US20150202226A1 - Pharmaceutical formulations useful for inhibiting acid secretion and methods for making and using them - Google Patents

Pharmaceutical formulations useful for inhibiting acid secretion and methods for making and using them Download PDF

Info

Publication number
US20150202226A1
US20150202226A1 US14/673,670 US201514673670A US2015202226A1 US 20150202226 A1 US20150202226 A1 US 20150202226A1 US 201514673670 A US201514673670 A US 201514673670A US 2015202226 A1 US2015202226 A1 US 2015202226A1
Authority
US
United States
Prior art keywords
proton pump
pump inhibitor
pharmaceutical formulation
antacid
meq
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
US14/673,670
Inventor
Warren Hall
Kay Olmstead
Laura Weston
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.)
Santarus Inc
Original Assignee
Santarus 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
Priority claimed from US10/893,203 external-priority patent/US20050037070A1/en
Application filed by Santarus Inc filed Critical Santarus Inc
Priority to US14/673,670 priority Critical patent/US20150202226A1/en
Publication of US20150202226A1 publication Critical patent/US20150202226A1/en
Assigned to BARCLAYS BANK PLC reassignment BARCLAYS BANK PLC SECURITY INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ATON PHARMA, INC., BAUSCH & LOMB INCORPORATED, BAUSH & LOMB PHARMA HOLDINGS CORP., DENDREON PHARMACEUTICALS, INC., DOW PHARMACEUTICAL SCIENCES, INC., MEDICIS PHARMACEUTICAL CORPORATION, OBAGI MEDICAL PRODUCTS, INC., OMP, INC., ORAPHARMA, INC., PRECISION DERMATOLOGY, INC., SALIX PHARMACEUTICALS, INC., Salix Pharmaceuticals, Ltd, SANTARUS, INC., SOLTA MEDICAL, INC., VALEANT CANADA LP, BY ITS GENERAL PARTNER VALEANT CANADA GP LIMITED, VALEANT HOLDINGS IRELAND (AS SUCCESSOR TO VALEANT INTERNATIONAL BERMUDA), VALEANT PHARMACEUTICALS INTERNATIONAL, INC., VALEANT PHARMACEUTICALS IRELAND, VALEANT PHARMACEUTICALS NORTH AMERICA LLC
Assigned to THE BANK OF NEW YORK MELLON reassignment THE BANK OF NEW YORK MELLON SECURITY INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SANTARUS, INC.
Assigned to BARCLAYS BANK PLC, AS COLLATERAL AGENT reassignment BARCLAYS BANK PLC, AS COLLATERAL AGENT SECURITY INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ATON PHARMA, INC., BAUSCH & LOMB INCORPORATED, BAUSCH & LOMB PHARMA HOLDINGS CORP., COMMONWEALTH LABORATORIES, LLC, DOW PHARMACEUTICAL SCIENCES, INC., ECR PHARMACEUTICALS CO., INC., LABORATOIRE CHAUVIN S.A.S., MEDICIS PHARMACEUTICAL CORPORATION, ONPHARMA INC., ORAPHARMA, INC., PRECISION DERMATOLOGY, INC., SALIX PHARMACEUTICALS, INC., SALIX PHARMACEUTICALS, LTD., SANTARUS, INC., SOLTA MEDICAL, INC., SYNERGETICS USA, INC., TECHNOLAS PERFECT VISION GMBH, VALEANT CANADA LP, VALEANT PHARMA POLAND SP. Z O.O., VALEANT PHARMACEUTICALS INTERNATIONAL, VALEANT PHARMACEUTICALS INTERNATIONAL, INC., VALEANT PHARMACEUTICALS IRELAND LIMITED, VALEANT PHARMACEUTICALS LUXEMBOURG S.A R.L., VALEANT PHARMACEUTICALS NORTH AMERICA LLC, WIRRA IP PTY LIMITED
Assigned to THE BANK OF NEW YORK MELLON, AS COLLATERAL AGENT reassignment THE BANK OF NEW YORK MELLON, AS COLLATERAL AGENT SECURITY INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ATON PHARMA, INC., BAUSCH & LOMB INCORPORATED, BAUSCH & LOMB PHARMA HOLDINGS CORP., COMMONWEALTH LABORATORIES, LLC, DOW PHARMACEUTICAL SCIENCES, INC., ECR PHARMACEUTICALS CO., INC., LABORATOIRE CHAUVIN S.A.S., MEDICIS PHARMACEUTICAL CORPORATION, ONPHARMA INC., ORAPHARMA, INC., PRECISION DERMATOLOGY, INC., SALIX PHARMACEUTICALS, INC., SALIX PHARMACEUTICALS, LTD., SANTARUS, INC., SOLTA MEDICAL, INC., SYNERGETICS USA, INC., TECHNOLAS PERFECT VISION GMBH, VALEANT CANADA LP, VALEANT PHARMA POLAND SP. Z O.O., VALEANT PHARMACEUTICALS INTERNATIONAL, VALEANT PHARMACEUTICALS INTERNATIONAL, INC., VALEANT PHARMACEUTICALS IRELAND LIMITED, VALEANT PHARMACEUTICALS LUXEMBOURG S.A R.L., VALEANT PHARMACEUTICALS NORTH AMERICA LLC, WIRRA IP PTY LIMITED
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/06Aluminium, calcium or magnesium; Compounds thereof, e.g. clay
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/06Aluminium, calcium or magnesium; Compounds thereof, e.g. clay
    • A61K33/08Oxides; Hydroxides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/02Inorganic compounds
    • 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
    • A61K9/0056Mouth soluble or dispersible forms; Suckable, eatable, chewable coherent forms; Forms rapidly disintegrating in the mouth; Lozenges; Lollipops; Bite capsules; Baked products; Baits or other oral forms for animals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2009Inorganic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2072Pills, tablets, discs, rods characterised by shape, structure or size; Tablets with holes, special break lines or identification marks; Partially coated tablets; Disintegrating flat shaped forms
    • A61K9/2077Tablets comprising drug-containing microparticles in a substantial amount of supporting matrix; Multiparticulate tablets
    • A61K9/2081Tablets comprising drug-containing microparticles in a substantial amount of supporting matrix; Multiparticulate tablets with microcapsules or coated microparticles according to A61K9/50
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/28Dragees; Coated pills or tablets, e.g. with film or compression coating
    • A61K9/2806Coating materials
    • A61K9/2833Organic macromolecular compounds
    • A61K9/284Organic macromolecular compounds obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyvinyl pyrrolidone
    • A61K9/2846Poly(meth)acrylates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/4808Preparations in capsules, e.g. of gelatin, of chocolate characterised by the form of the capsule or the structure of the filling; Capsules containing small tablets; Capsules with outer layer for immediate drug release
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/5005Wall or coating material
    • A61K9/5021Organic macromolecular compounds
    • A61K9/5036Polysaccharides, e.g. gums, alginate; Cyclodextrin
    • A61K9/5042Cellulose; Cellulose derivatives, e.g. phthalate or acetate succinate esters of hydroxypropyl methylcellulose
    • A61K9/5047Cellulose ethers containing no ester groups, e.g. hydroxypropyl methylcellulose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/5089Processes

Definitions

  • the present invention is related to pharmaceutical formulations comprising an antacid and a proton pump inhibitor microencapsulated with (1) a material that enhances the shelf-life of the composition, and/or (2) a taste-masking material.
  • pharmaceutical compositions comprising proton pump inhibitor and antacid wherein the proton pump inhibitor is dry coated.
  • methods for manufacture of the pharmaceutical formulations; uses of the pharmaceutical formulations in treating disease; and combinations of the pharmaceutical formulations with other therapeutic agents are described.
  • compositions with enteric-coatings have been designed to dissolve at a pH to ensure that the drug is released in the proximal region of the small intestine (duodenum), rather than the acidic environment of the stomach.
  • enteric-coated compositions due to the pH-dependent attributes of these enteric-coated compositions and the uncertainty of gastric retention time, in-vivo performance as well as both inter- and intra-subject variability are all major set backs of using enteric-coated systems for the controlled release of a drug.
  • compositions which allow for the immediate release of the pharmaceutically active ingredient into the stomach, involve the administration of one or more buffering agents with an acid labile pharmaceutical agent, such as a proton pump inhibitor.
  • the buffering agent is thought to prevent substantial degradation of the acid labile pharmaceutical agent in the acidic environment of the stomach by raising the pH. See, e.g., U.S. Pat. Nos. 5,840,737; 6,489,346; 6,645,988; and 6,699,885; and U.S. patent application Ser. No. 10/898,135.
  • a class of acid-labile pharmaceutical compounds that are administered as enteric-coated dosage forms are proton pump inhibiting agents.
  • proton pump inhibitors include, omeprazole (Prilosec®), lansoprazole (Prevacid®), esomeprazole (Nexium®), rabeprazole. Aciphex®), pantoprazole (Protonix®), pariprazole, tentaprazole, and leminoprazole.
  • the drugs of this class suppress gastrointestinal acid secretion by the specific inhibition of the H + /K + -ATPase enzyme system (proton pump) at the secretory surface of the gastrointestinal parietal cell.
  • Omeprazole is one example of a proton pump inhibitor which is a substituted bicyclic aryl-imidazole, 5-methoxy-2-[(4-methoxy-3,5-dimethyl-2-pyridinyl)methyl]sulfinyl]-1H-benzimidazole, that inhibits gastrointestinal acid secretion.
  • U.S. Pat. No. 4,786,505 to Lovgren et al. teaches that a pharmaceutical oral solid dosage form of omeprazole must be protected from contact with acidic gastrointestinal juice by an enteric-coating to maintain its pharmaceutical activity and describes an enteric-coated omeprazole preparation containing one or more subcoats between the core material and the enteric-coating.
  • Proton pump inhibitors are typically prescribed for short-term treatment of active duodenal ulcers, gastrointestinal ulcers, gastro esophageal reflux disease (GERD), severe erosive esophagitis, poorly responsive symptomatic GERD, and pathological hypersecretory conditions such as Zollinger Ellison syndrome. These above-listed conditions commonly arise in healthy or critically ill patients of all ages, and may be accompanied by significant upper gastrointestinal bleeding.
  • GGERD gastro esophageal reflux disease
  • severe erosive esophagitis severe erosive esophagitis
  • poorly responsive symptomatic GERD poorly responsive symptomatic GERD
  • pathological hypersecretory conditions such as Zollinger Ellison syndrome.
  • omeprazole, lansoprazole and other proton pump inhibiting agents reduce gastrointestinal acid production by inhibiting H + /K + -ATPase of the parietal cell during the final common pathway for gastrointestinal acid secretion.
  • Fellenius et al. Substituted Benzimidazoles Inhibit Gastrointestinal Acid Secretion by Blocking H + /K + -ATPase, Nature, 290: 159-161 (1981); Wallmark et al., The Relationship Between Gastrointestinal Acid Secretion and Gastrointestinal H + /K + -ATPase Activity, J. Biol.
  • Proton pump inhibitors have the ability to act as weak bases which reach parietal cells from the blood and diffuse into the secretory canaliculi. There the drugs become protonated and thereby trapped. The protonated compound can then rearrange to form a sulfenamide which can covalently interact with sulthydryl groups at critical sites in the extra cellular (luminal) domain of the membrane-spanning H + /K + -ATPase. See, e.g., Hardman et al., Goodman & Gilman's The Pharmacological Basis of Therapeutics , 907 (9th ed. 1996). As such, proton pump inhibitors are prodrugs that must be activated to be effective.
  • the specificity of the effects of proton pump inhibiting agents is also dependent upon: (a) the selective distribution of H + /K + -ATPase; (b) the requirement for acidic conditions to catalyze generation of the reactive inhibitor; and (c) the trapping of the protonated drug and the cationic sulfenamide within the acidic canaliculi and adjacent to the target enzyme. See, e.g., Hardman et al.
  • FIG. 1 is a graph comparing the pharmacokinetic release profiles of omeprazole of Prilosec®, non-microencapsulated omeprazole with antacid (as described in Example 13B), omeprazole microencapsulated with Klucel and antacid tablet (as described in Example 13C), and omeprazole microencapsulated with Methocel and antacid tablet (as described in Example 13 D) in human.
  • FIGS. 2A and 2B are SEM micrographs of micronized omeprazole and omeprazole microencapsulated with Klucel® Hydroxypropyl Cellulose.
  • FIG. 3 is a graph comparing the average pharmacokinetic release profiles of SAN-15A, SAN-15B, SAN-15C, SAN-20D and SAN-20E as compared to Priolosec® brand enteric coated omeprazole 40 mg. Formulations were prepared as described in Example 13.
  • FIG. 4 is a graph comparing the mean peak plasma concentration (Cmax) versus the time at which Cmax is observed (Tmax) for SAN-15 (20 mg and 40 mg), SAN-20D, SAN-20E and Priolosec® brand enteric coated omeprazole 20 mg and 40 mg, Day 1.
  • the data is from the human clinical trial described in Examples 14B and 15B.
  • FIG. 5 is a graph comparing the average pharmacokinetic release profiles for SAN-15 20 mg and Priolosec® brand enteric coated omeprazole 20 mg, Day 1. The data is from the human clinical trial described in Examples 14B.
  • FIG. 6 is a graph comparing the average pharmacokinetic release profiles for SAN-15 40 mg and Priolosec® brand enteric coated omeprazole 40 mg, Day 1. The data is from the human clinical trial described in Examples 15B.
  • FIG. 7 is a graph comparing the average pharmacokinetic release profiles for SAN-15 20 mg and Priolosec® brand enteric coated omeprazole 20 mg, Day 7. The data is from the human clinical trial described in Examples 14B.
  • FIG. 8 is a graph comparing the average pharmacokinetic release profiles for SAN-15 40 mg and Priolosec® brand enteric coated omeprazole 40 mg, Day 7. The data is from the human clinical trial described in Examples 15B.
  • FIG. 9 is a graph comparing the mean peak plasma concentration (Cmax) versus the time at which Cmax is observed (Tmax) for SAN-15 (20 mg and 40 mg) and Priolosec® brand enteric coated omeprazole 20 mg and 40 mg, Day 7. The data is from the human clinical trial described in Examples 14B and 15B.
  • FIG. 10 is a graph comparing the mean peak plasma concentration (Cmax) versus the time at which Cmax is observed (Tmax) for SAN-15 (20 mg and 40 mg) and Priolosec® brand enteric coated omeprazole 20 mg and 40 mg, Day 1 and Day 7. The data is from the human clinical trial described in Examples 14B and 15B.
  • compositions having enhanced shelf-lives comprising, at least one acid labile proton pump inhibitor which is microencapsulated with a material that enhances the shelf-life of the pharmaceutical formulation and at least one antacid; wherein an initial serum concentration of the proton pump inhibitor is greater than about 0.1 ⁇ g/ml at any time within about 30 minutes after administration of the pharmaceutical formulation.
  • taste-masked pharmaceutical formulations comprising at least one acid labile proton pump inhibitor which is microencapsulated with a taste-masking material and at least one antacid; wherein an initial serum concentration of the proton pump inhibitor is greater than about 0.1 ⁇ g/ml at any time within about 30 minutes after administration of the pharmaceutical formulation.
  • the proton pump inhibitor is microencapsulated with one or more compounds selected from cellulose hydroxypropyl ethers; low-substituted hydroxypropyl ethers; cellulose hydroxypropyl methyl ethers; methylcellulose polymers; ethylcelluloses and mixtures thereof; polyvinyl alcohol; hydroxyethylcelluloses; carboxymethylcelluloses and salts of carboxymethylcelluloses; polyvinyl alcohol and polyethylene glycol co-polymers; monoglycerides; triglycerides; polyethylene glycols, modified food starch, acrylic polymers; mixtures of acrylic polymers with cellulose ethers; cellulose acetate phthalate; sepifilms, cyclodextrins; and mixtures thereof.
  • the proton pump inhibitor is microencapsulated with one or more additives to enhance the processing or performance of microencapsulation.
  • additives maybe pH modifier, plastersizer, antioxidant, or sweetener or flavor.
  • compositions having enhanced shelf-lives comprising, at least one acid labile proton pump inhibitor and at least one antacid; wherein an initial serum concentration of the proton pump inhibitor is greater than about 0.1 ⁇ g/ml at any time within about 30 minutes after administration of the pharmaceutical formulation, wherein some or all of the proton pump inhibitor is dry coated.
  • the at least one antacid comprises at least one soluble antacid.
  • the soluble antacid is sodium bicarbonate.
  • the at least one buffer is selected from sodium bicarbonate, calcium carbonate, sodium carbonate, magnesium oxide, magnesium hydroxide, magnesium carbonate, aluminum hydroxide, and mixtures thereof.
  • kits for extending the shelf-life of pharmaceutical formulations comprising microencapsulating at least one acid labile proton pump inhibitor with a material that enhances the shelf-life; and combining the microencapsulated acid labile proton pump inhibitor with at least one antacid.
  • methods of masking the taste of a pharmaceutical formulation comprising microencapsulating at least one acid labile proton pump inhibitor with a taste-masking material; and combining the microencapsulated acid labile proton pump inhibitor with an antacid.
  • the proton pump inhibitor is combined with some or all of the antacid to form a slug or sheet of material. This intermediate product is then broken into granular material with is combined with other components present in the pharmaceutical formulation.
  • the dry coated proton pump inhibitor is combined with additional antacid.
  • the pharmaceutical formulations may further comprise one or more excipients selected from parietal cell activators, organic solvents, erosion facilitators, diffusion facilitators, antioxidants, flavoring agents and carrier materials selected from binders, suspending agents, disintegration agents, filling agents, surfactants, solubilizers, stabilizers, lubricants, wetting agents, diluents, anti-adherents, and antifoaming agents.
  • excipients selected from parietal cell activators, organic solvents, erosion facilitators, diffusion facilitators, antioxidants, flavoring agents and carrier materials selected from binders, suspending agents, disintegration agents, filling agents, surfactants, solubilizers, stabilizers, lubricants, wetting agents, diluents, anti-adherents, and antifoaming agents.
  • the present invention is directed to pharmaceutical formulations exhibiting enhanced shelf-life stability and/or improved taste masking properties useful for the treatment of a disease, condition or disorder. Methods of treatment using the pharmaceutical formulations of the present invention are also described.
  • compositions comprising (1) an acid labile proton pump inhibitor which is microencapsulated with a material that enhances the shelf-life of the pharmaceutical composition together with (2) one or more antacid, provide superior performance by enhancing shelf-life stability of the pharmaceutical formulation during manufacturing and storage.
  • compositions comprising (1) an acid labile proton pump inhibitor which has been dry coated and (2) one or more antacids, provide superior performance by enhancing shelf-life stability of the pharmaceutical formulation during manufacturing and storage.
  • the proton pump inhibitor is dry coated with some or all of the antacid.
  • the dry coated proton pump inhibitor is combined with a second antacid which can be the same antacid as used to dry coat the proton pump inhibitor or a different antacid.
  • the dry coated material is combined with one or more pharmaceutical excipients.
  • the proton pump inhibitor is dry coated with a material comprising antacid, sweetener(s), lubricant, and binder.
  • Certain taste-masking materials have also been discovered which, when used in the pharmaceutical formulations provide (1) more palatable forms of the drug by blocking the contact of the unpleasant taste of the pharmaceutical agent from the contact of the taste receptor, thereby increasing patient compliance; and/or (2) require lower amounts of traditional flavoring agents.
  • acid-labile pharmaceutical agent refers to any pharmacologically active drug subject to acid catalyzed degradation.
  • Aftertaste is a measurement of all sensation remaining after swallowing. Aftertaste can be measured, e.g., from 30 seconds after swallowing, 1 minutes after swallowing, 2 minutes after swallowing, 3 minutes after swallowing, 4 minutes after swallowing, 5 minutes after swallowing, and the like.
  • Amplitude is the initial overall perception of the flavors balance and fullness.
  • the amplitude scale is 0-none, 1-low, 2-moderate, and 3-high.
  • Anti-adherents prevent components of the formulation from aggregating or sticking and improve flow characteristics of a material.
  • Such compounds include, e.g., colloidal silicon dioxide such as Cab-o-sil®; tribasic calcium phosphate, talc, corn starch, DL-leucine, sodium lauryl sulfate, magnesium stearate, calcium stearate, sodium stearate, kaolin, and micronized amorphous silicon dioxide (Syloid®) and the like.
  • Antifoaming agents reduce foaming during processing which can result in coagulation of aqueous dispersions, bubbles in the finished film, or generally impair processing.
  • Exemplary anti-foaming agents include silicon emulsions or sorbitan sesquoleate.
  • Antioxidants include, e.g., butylated hydroxytoluene (BHT), sodium ascorbate, and tocopherol.
  • BHT butylated hydroxytoluene
  • sodium ascorbate sodium ascorbate
  • tocopherol sodium ascorbate
  • Binders impart cohesive qualities and include, e.g., alginic acid and salts thereof; cellulose derivatives such as carboxymethylcellulose, methylcellulose (e.g., Methocel®), hydroxypropylmethylcellulose, hydroxyethylcellulose, hydroxypropylcellulose (e.g., Klucel®), ethylcellulose (e.g., Ethocel®), and microcrystalline cellulose (e.g., Avicel®); microcrystalline dextrose; amylose; magnesium aluminum silicate; polysaccharide acids; bentonites; gelatin; polyvinylpyrrolidone/vinyl acetate copolymer; crospovidone; povidone; starch; pregelatinized starch; tragacanth, dextrin, a sugar, such as sucrose (e.g., Dipac®), glucose, dextrose, molasses, mannitol, sorbitol, xylitol (e.g., cellulose
  • Bioavailability refers to the extent to which an active moiety, e.g., drug, prodrug, or metabolite, is absorbed into the general circulation and becomes available at the site of drug action in the body. Thus, a proton pump inhibitor administered through IV is 100% bioavailable. “Oral bioavailability” refers to the extent to with the proton pump inhibitor is absorbed into the general circulation and becomes available at the site of the drug action in the body when the pharmaceutical formulation is taken orally.
  • Bioequivalence or “bioequivalent” means that the area under the serum concentration time curve (AUC) and the peak serum concentration (C max ) are each within 80% and 125% with a 90% confidence interval.
  • Carrier materials include any commonly used excipients in pharmaceutics and should be selected on the basis of compatibility with the proton pump inhibitor and the release profile properties of the desired dosage form.
  • Exemplary carrier materials include, e.g., binders, suspending agents, disintegration agents, filling agents, surfactants, solubilizers, stabilizers, lubricants, wetting agents, diluents, and the like.
  • “Pharmaceutically compatible carrier materials” may comprise, e.g., acacia, gelatin, colloidal silicon dioxide, calcium glycerophosphate, calcium lactate, maltodextrin, glycerine, magnesium silicate, sodium caseinate, soy lecithin, sodium chloride, tricalcium phosphate, dipotassium phosphate, sodium stearoyl lactylate, carrageenan, monoglyceride, diglyceride, pregelatinized starch, and the like. See, e.g., Remington: The Science and Practice of Pharmacy , Nineteenth Ed (Easton, Pa.: Mack Publishing Company, 1995); Hoover, John E., Remington's Pharmaceutical Sciences , Mack Publishing Co., Easton, Pa.
  • “Character notes” include, e.g., aromatics, basis tastes, and feeling factors.
  • the intensity of the character note can be scaled from 0-none, 1-slight, 2-moderate, or 3-strong.
  • a “derivative” is a compound that is produced from another compound of similar structure by the replacement of substitution of an atom, molecule or group by another suitable atom, molecule or group.
  • one or more hydrogen atom of a compound may be substituted by one or more alkyl, acyl, amino, hydroxyl, halo, haloalkyl, aryl, heteroaryl, cycloaolkyl, heterocycloalkyl, or heteroalkyl group to produce a derivative of that compound.
  • diffusion facilitators and “dispersing agents” include materials that control the diffusion of an aqueous fluid through a coating.
  • Exemplary diffusion facilitators/dispersing agents include, e.g., hydrophilic polymers, electrolytes, Tween® 60 or 80, PEG and the like. Combinations of one or more erosion facilitator with one or more diffusion facilitator can also be used in the present invention.
  • “Diluents” increase bulk of the composition to facilitate compression.
  • Such compounds include e.g., lactose; starch; mannitol; sorbitol; dextrose; microcrystalline cellulose such as Avicel®; dibasic calcium phosphate; dicalcium phosphate dihydrate; tricalcium phosphate; calcium phosphate; anhydrous lactose; spray-dried lactose; pregelatinzed starch; compressible sugar, such as Di-Pac® (Amstar); mannitol; hydroxypropylmethylcellulose; sucrose-based diluents; confectioner's sugar; monobasic calcium sulfate monohydrate; calcium sulfate dihydrate; calcium lactate trihydrate; dextrates; hydrolyzed cereal solids; amylose; powdered cellulose; calcium carbonate; glycine; kaolin; mannitol; sodium chloride; inositol; bentonite; and the like.
  • disintegrate includes both the dissolution and dispersion of the dosage form when contacted with gastrointestinal fluid.
  • Disintegration agents facilitate the breakup or disintegration of a substance.
  • disintegration agents include a starch, e.g., a natural starch such as corn starch or potato starch, a pregelatinized starch such as National 1551 or Amijel®, or sodium starch glycolate such as Promogel® or Explotab®; a cellulose such as a wood product, methylcrystalline cellulose, e.g., Avicel®, Avicel® PH101, Avicel® PH102, Avicel® PH105, Elcema® P100, Emcocel®, Vivacel®, Ming Tia®, and Solka-Floc®, methylcellulose, croscarmellose, or a cross-linked cellulose, such as cross-linked sodium carboxymethylcellulose (Ac-Di-Sol®), cross-linked carboxymethylcellulose, or cross-linked croscarmellose; a cross-linked starch such as sodium starch glycolate; a cross-linked polymer such as sodium starch
  • Drug absorption or “absorption” refers to the process of movement from the site of administration of a drug toward the systemic circulation, e.g., into the bloodstream of a subject.
  • “Dry coating” is a method of coating the proton pump inhibitor with one or more other components without using water or other solvents.
  • “Dry granulation” is a method of converting powder particles into granules using the application of pressure without the use of a liquid.
  • enteric coating is a substance that remains substantially intact in the stomach but dissolves and releases the drug once the small intestine is reached.
  • the enteric coating comprises a polymeric material that prevents release in the low pH environment of the stomach but that ionizes at a slightly higher pH, typically a pH of 4 or 5, and thus dissolves sufficiently in the small intestines to gradually release the active agent therein.
  • the “enteric form of the proton pump inhibitor” is intended to mean that some or most of the proton pump inhibitor has been enterically coated to ensure that at least some of the drug is released in the proximal region of the small intestine (duodenum), rather than the acidic environment of the stomach.
  • Erosion facilitators include materials that control the erosion of a particular material in gastrointestinal fluid. Erosion facilitators are generally known to those of ordinary skill in the art. Exemplary erosion facilitators include, e.g., hydrophilic polymers, electrolytes, proteins, peptides, and amino acids.
  • Filling agents include compounds such as lactose, calcium carbonate, calcium phosphate, dibasic calcium phosphate, calcium sulfate, microcrystalline cellulose, cellulose powder, dextrose, dextrates, dextran, starches, pregelatinized starch, sucrose, xylitol, lactitol, mannitol, sorbitol, sodium chloride, polyethylene glycol, and the like.
  • “Flavoring agents” or “sweeteners” useful in the pharmaceutical compositions of the present invention include, e.g., acacia syrup, acesulfame K, alitame, anise, apple, aspartame, banana, Bavarian cream, berry, black currant, butterscotch, calcium citrate, camphor, caramel, cherry, cherry cream, chocolate, cinnamon, bubble gum, citrus, citrus punch, citrus cream, cotton candy, cocoa, cola, cool cherry, cool citrus, cyclamate, cylamate, dextrose, eucalyptus, eugenol, fructose, fruit punch, ginger, glycyrrhetinate, glycyrrhiza (licorice) syrup, grape, grapefruit, honey, isomalt, lemon, lime, lemon cream, monoammonium glyrrhizinate (MagnaSweet®), maltol, mannitol, maple, marshmallow, menthol, mint
  • stomach secretion is the fluid of stomach secretions of a subject or the saliva of a subject after oral administration of a composition of the present invention, or the equivalent thereof.
  • An “equivalent of stomach secretion” includes, e.g., an in vitro fluid having similar content and/or pH as stomach secretions such as a 1% sodium dodecyl sulfate solution or 0.1N HCl solution in water.
  • “Half-life” refers to the time required for the plasma drug concentration or the amount in the body to decrease by 50% from its maximum concentration.
  • “Lubricants” are compounds that prevent, reduce or inhibit adhesion or friction of materials.
  • Exemplary lubricants include, e.g., stearic acid; calcium hydroxide; talc; sodium stearyl fumerate; a hydrocarbon such as mineral oil, or hydrogenated vegetable oil such as hydrogenated soybean oil (Sterotex®); higher fatty acids and their alkali-metal and alkaline earth metal salts, such as aluminum, calcium, magnesium, zinc, stearic acid, sodium stearates, glycerol, talc, waxes, Stearowet®, boric acid, sodium benzoate, sodium acetate, sodium chloride, leucine, a polyethylene glycol or a methoxypolyethylene glycol such as CarbowaxTM, sodium oleate, glyceryl behenate, polyethylene glycol, magnesium or sodium lauryl sulfate, colloidal silica such as SyloidTM, Carb-O-Sil®,
  • a “measurable serum concentration” or “measurable plasma concentration” describes the blood serum or blood plasma concentration, typically measured in mg, ⁇ g, or ng of therapeutic agent per ml, dl, or l of blood serum, of a therapeutic agent that is absorbed into the bloodstream after administration.
  • measurable serum concentration or “measurable plasma concentration” describes the blood serum or blood plasma concentration, typically measured in mg, ⁇ g, or ng of therapeutic agent per ml, dl, or l of blood serum, of a therapeutic agent that is absorbed into the bloodstream after administration.
  • One of ordinary skill in the art would be able to measure the serum concentration or plasma concentration of a proton pump inhibitor or other therapeutic agent. See, e.g., Gonzalez H. et al., J. Chromatogr. B. Analyt. Technol. Biomed. Life Sci ., vol. 780, pp 459-65, (Nov. 25, 2002).
  • Parietal cell activators stimulate the parietal cells and enhance the pharmaceutical activity of the proton pump inhibitor.
  • Parietal cell activators include, e.g., chocolate; alkaline substances such as sodium bicarbonate; calcium such as calcium carbonate, calcium gluconate, calcium hydroxide, calcium acetate and calcium glycerophosphate; peppermint oil; spearmint oil; coffee; tea and colas (even if decaffeinated); caffeine; theophylline; theobromine; amino acids (particularly aromatic amino acids such as phenylalanine and tryptophan); and combinations thereof.
  • “Pharmacodynamics” refers to the factors that determine the biologic response observed relative to the concentration of drug at a site of action.
  • “Pharmacokinetics” refers to the factors that determine the attainment and maintenance of the appropriate concentration of drug at a site of action.
  • Plasma concentration refers to the concentration of a substance in blood plasma or blood serum of a subject. It is understood that the plasma concentration of a therapeutic agent may vary many-fold between subjects, due to variability with respect to metabolism of therapeutic agents. In accordance with one aspect of the present invention, the plasma concentration of a proton pump inhibitors and/or other therapeutic agent may vary from subject to subject. Likewise, values such as maximum plasma concentration (C max ) or time to reach maximum serum concentration (T max ), or area under the serum concentration time curve (AUC) may vary from subject to subject. Due to this variability, the amount necessary to constitute “a therapeutically effective amount” of proton pump inhibitor or other therapeutic agent, may vary from subject to subject. It is understood that when mean plasma concentrations are disclosed for a population of subjects, these mean values may include substantial variation.
  • Plasticizers are compounds used to soften the microencapsulation material or film coatings to make them less brittle. Suitable plasticizers include, e.g., polyethylene glycols such as PEG 300, PEG 400, PEG 600, PEG 1450, PEG 3350, and PEG 800, stearic acid, propylene glycol, oleic acid, and triacetin.
  • Prevent or “prevention” when used in the context of a gastric acid related disorder means no gastrointestinal disorder or disease development if none had occurred, or no further gastrointestinal disorder or disease development if there had already been development of the gastrointestinal disorder or disease. Also considered is the ability of one to prevent some or all of the symptoms associated with the gastrointestinal disorder or disease.
  • a “prodrug” refers to a drug or compound in which the pharmacological action results from conversion by metabolic processes within the body.
  • Prodrugs are generally drug precursors that, following administration to a subject and subsequent absorption, are converted to an active, or a more active species via some process, such as conversion by a metabolic pathway.
  • Some prodrugs have a chemical group present on the prodrug that renders it less active and/or confers solubility or some other property to the drug. Once the chemical group has been cleaved and/or modified from the prodrug the active drug is generated.
  • Prodrugs may be designed as reversible drug derivatives, for use as modifiers to enhance drug transport to site-specific tissues.
  • prodrugs to date has been to increase the effective water solubility of the therapeutic compound for targeting to regions where water is the principal solvent. See, e.g., Fedorak et al., Am. J. Physiol., 269:G210-218 (1995); McLoed et al., Gastroenterol, 106:405-413 (1994); Hochhaus et al., Biomed. Chrom., 6:283-286 (1992); J. Larsen and H. Bundgaard, Int. J. Pharmaceutics, 37, 87 (1987); J. Larsen et al., Int. J. Pharmaceutics, 47, 103 (1988); Sinkula et al., J. Pharm.
  • Proton pump inhibitor product refers to a product sold on the market.
  • Proton pump inhibitor products include, for example, Priolosec®, Nexium®, Prevacid®, Protonix®, and Aciphex®.
  • “Serum concentration” refers to the concentration of a substance such as a therapeutic agent, in blood plasma or blood serum of a subject. It is understood that the serum concentration of a therapeutic agent may vary many-fold between subjects, due to variability with respect to metabolism of therapeutic agents. In accordance with one aspect of the present invention, the serum concentration of a proton pump inhibitors and/or prokinetic agent may vary from subject to subject. Likewise, values such as maximum serum concentration (C max ) or time to reach maximum serum concentration (T max ), or total area under the serum concentration time curve (AUC) may vary from subject to subject. Due to this variability, the amount necessary to constitute “a therapeutically effective amount” of proton pump inhibitor, prokinetic agent, or other therapeutic agent, may vary from subject to subject. It is understood that when mean serum concentrations are disclosed for a population of subjects, these mean values may include substantial variation.
  • Solidizers include compounds such as citric acid, succinic acid, fumaric acid, malic acid, tartaric acid, maleic acid, glutaric acid, sodium bicarbonate, sodium carbonate and the like.
  • Stabilizers include compounds such as any antioxidation agents, buffers, acids, and the like.
  • “Suspending agents” or “thickening agents” include compounds such as polyvinylpyrrolidone, e.g., polyvinylpyrrolidone K12, polyvinylpyrrolidone K17, polyvinylpyrrolidone K25, or polyvinylpyrrolidone K30; polyethylene glycol, e.g., the polyethylene glycol can have a molecular weight of about 300 to about 6000, or about 3350 to about 4000, or about 7000 to about 5400; sodium carboxymethylcellulose; methylcellulose; hydroxypropylmethylcellulose; polysorbate-80; hydroxyethylcellulose; sodium alginate; gums, such as, e.g., gum tragacanth and gum acacia; guar gum; xanthans, including xanthan gum; sugars; cellulosics, such as, e.g., sodium carboxymethylcellulose, methylcellulose, sodium carboxymethylcellulose, hydroxypropylmethylcellulose
  • “Surfactants” include compounds such as sodium lauryl sulfate, sorbitan monooleate, polyoxyethylene sorbitan monooleate, polysorbates, polaxomers, bile salts, glyceryl monostearate, copolymers of ethylene oxide and propylene oxide, e.g., Pluronic® (BASF); and the like.
  • a “therapeutically effective amount” or “effective amount” is that amount of a pharmaceutical agent to achieve a pharmacological effect.
  • the term “therapeutically effective amount” includes, for example, a prophylactically effective amount.
  • An “effective amount” of a proton pump inhibitor is an amount effective to achieve a desired pharmacologic effect or therapeutic improvement without undue adverse side effects.
  • an effective amount of a proton pump inhibitor refers to an amount of proton pump inhibitor that reduces acid secretion, or raises gastrointestinal fluid pH, or reduces gastrointestinal bleeding, or reduces the need for blood transfusion, or improves survival rate, or provides for a more rapid recovery from a gastric acid related disorder.
  • an effect amount” or “a therapeutically effective amount” can vary from subject to subject, due to variation in metabolism of therapeutic agents such as proton pump inhibitors and/or prokinetic agents, age, weight, general condition of the subject, the condition being treated, the severity of the condition being treated, and the judgment of the prescribing physician.
  • Total intensity of aroma is the overall immediate impression of the strength of the aroma and includes both aromatics and nose feel sensations.
  • Total intensity of flavor is the overall immediate impression of the strength of the flavor including aromatics, basic tastes and mouth feel sensations.
  • Treat” or “treatment” as used in the context of a gastric acid related disorder refers to any treatment of a disorder or disease associated with a gastrointestinal disorder, such as preventing the disorder or disease from occurring in a subject which may be predisposed to the disorder or disease, but has not yet been diagnosed as having the disorder or disease; inhibiting the disorder or disease, e.g., arresting the development of the disorder or disease, relieving the disorder or disease, causing regression of the disorder or disease, relieving a condition caused by the disease or disorder, or stopping the symptoms of the disease or disorder.
  • the term “treat” is used synonymously with the term “prevent.”
  • Weight agents include compounds such as oleic acid, glyceryl monostearate, sorbitan monooleate, sorbitan monolaurate, triethanolamine oleate, polyoxyethylene sorbitan monooleate, polyoxyethylene sorbitan monolaurate, sodium oleate, sodium lauryl sulfate, and the like.
  • proton pump inhibitor PPI
  • proton pump inhibiting agent can be used interchangeably to describe any acid labile pharmaceutical agent possessing pharmacological activity as an inhibitor of H+/K+-ATPase.
  • a proton pump inhibitor may, if desired, be in the form of free base, free acid, salt, ester, hydrate, anhydrate, amide, enantiomer, isomer, tautomer, prodrug, polymorph, derivative, or the like, provided that the free base, salt, ester, hydrate, amide, enantiomer, isomer, tautomer, prodrug, or any other pharmacologically suitable derivative is therapeutically active.
  • the proton pump inhibitor can be a substituted bicyclic aryl-imidazole, wherein the aryl group can be, e.g., a pyridine, a phenyl, or a pyrimidine group and is attached to the 4- and 5-positions of the imidazole ring.
  • the aryl group can be, e.g., a pyridine, a phenyl, or a pyrimidine group and is attached to the 4- and 5-positions of the imidazole ring.
  • Proton pump inhibitors comprising a substituted bicyclic aryl-imidazoles include, but are not limited to, omeprazole, hydroxyomeprazole, esomeprazole, lansoprazole, pantoprazole, rabeprazole, dontoprazole, dontoprazole, dontoprazole, dontoprazole, dontoprazole, dontoprazole, dontoprazole, dontoprazole, dontoprazole, dontoprazole, dontoprazole, dontoprazole, dontoprazole, dontoprazole, dontoprazole, dontoprazole, dontoprazole, dontoprazole, dontoprazole, dontoprazole, dontoprazole, dontoprazole, dontoprazole, dontoprazole, dontoprazole, dontoprazole, dontoprazole, dontoprazole, dontoprazole, dontoprazole, dontop
  • proton pump inhibitors include but are not limited to: soraprazan (Altana); ilaprazole (U.S. Pat. No. 5,703,097) (Il-Yang); AZD-0865 (AstraZeneca); YH-1885 (PCT Publication WO 96/05177) (SB-641257) (2-pyrimidinamine, 4-(3,4-dihydro-1-methyl-2(1H)-isoquinolinyl)-N-(4-fluorophenyl)-5,6-dimethyl-monohydrochloride)(YuHan); BY-112 (Altana); SPI-447 (Imidazo(1,2-a)thieno(3,2-c)pyridin-3-amine,5-methyl-2-(2-methyl-3-thienyl) (Shinnippon); 3-hydroxymethyl-2-methyl-9-phenyl-7H-8,9-dihydro-pyrano(2,3-c)-imidazo(1,
  • Still other proton pump inhibitors contemplated by the present invention include those described in the following U.S. Pat. Nos. 4,628,098; 4,689,333; 4,786,505; 4,853,230; 4,965,269; 5,021,433; 5,026,560; 5,045,321; 5,093,132; 5,430,042; 5,433,959; 5,576,025; 5,639,478; 5,703,110; 5,705,517; 5,708,017; 5,731,006; 5,824,339; 5,855,914; 5,879,708; 5,948,773; 6,017,560; 6,123,962; 6,187,340; 6,296,875; 6,319,904; 6,328,994; 4,255,431; 4,508,905; 4,636,499; 4,738,974; 5,690,960; 5,714,504; 5,753,265; 5,817,338; 6,093,734; 6,013,281; 6,
  • “Pharmaceutically acceptable salts,” or “salts,” include, e.g., the salt of a proton pump inhibitor prepared from formic, acetic, propionic, succinic, glycolic, gluconic, lactic, malic, tartaric, citric, ascorbic, glucuronic, maleic, fumaric, pyruvic, aspartic, glutamic, benzoic, anthranilic, mesylic, stearic, salicylic, p-hydroxybenzoic, phenylacetic, mandelic, embonic, methanesulfonic, ethanesulfonic, benzenesulfonic, pantothenic, toluenesulfonic, 2-hydroxyethanesulfonic, sulfanilic, cyclohexylaminosulfonic, algenic, ⁇ -hydroxybutyric, galactaric and galacturonic acids.
  • a proton pump inhibitor prepared from formic, acetic
  • acid addition salts are prepared from the free base using conventional methodology involving reaction of the free base with a suitable acid.
  • suitable acids for preparing acid addition salts include both organic acids, e.g., acetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, malic acid, malonic acid, succinic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid, and the like, as well as inorganic acids, e.g., hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like.
  • an acid addition salt is reconverted to the free base by treatment with a suitable base.
  • the acid addition salts of the proton pump inhibitors are halide salts, which are prepared using hydrochloric or hydrobromic acids.
  • the basic salts are alkali metal salts, e.g., sodium salt.
  • Salt forms of proton pump inhibiting agents include, but are not limited to: a sodium salt form such as esomeprazole sodium, omeprazole sodium, rabeprazole sodium, pantoprazole sodium; or a magnesium salt form such as esomeprazole magnesium or omeprazole magnesium, described in U.S. Pat. No. 5,900,424; a calcium salt form; or a potassium salt form such as the potassium salt of esomeprazole, described in U.S. patent application Ser. No. 02/0198239 and U.S. Pat. No. 6,511,996. Other salts of esomeprazole are described in U.S. Pat. No. 4,738,974 and U.S. Pat. No. 6,369,085. Salt forms of pantoprazole and lansoprazole are discussed in U.S. Pat. Nos. 4,758,579 and 4,628,098, respectively.
  • esters in one embodiment, preparation of esters involves functionalizing hydroxyl and/or carboxyl groups that may be present within the molecular structure of the drug.
  • the esters are acyl-substituted derivatives of free alcohol groups, e.g., moieties derived from carboxylic acids of the formula RCOOR 1 where R 1 is a lower alkyl group.
  • Esters can be reconverted to the free acids, if desired, by using conventional procedures such as hydrogenolysis or hydrolysis.
  • amides may be prepared using techniques known to those skilled in the art or described in the pertinent literature. For example, amides may be prepared from esters, using suitable amine reactants, or they may be prepared from an anhydride or an acid chloride by reaction with an amine group such as ammonia or a lower alkyl amine.
  • “Tautomers” of substituted bicyclic aryl-imidazoles include, e.g., tautomers of omeprazole such as those described in U.S. Pat. Nos. 6,262,085; 6,262,086; 6,268,385; 6,312,723; 6,316,020; 6,326,384; 6,369,087; and 6,444,689; and U.S. Patent Publication No. 02/0156103.
  • An exemplary “isomer” of a substituted bicyclic aryl-imidazole is the isomer of omeprazole including but not limited to isomers described in: Oishi et al., Acta Cryst. (1989), C45, 1921-1923; U.S. Pat. No. 6,150,380; U.S. Patent Publication No. 02/0156284; and PCT Publication No. WO 02/085889.
  • Exemplary “polymorphs” include, but are not limited to, those described in PCT Publication No. WO 92/08716, and U.S. Pat. Nos. 4,045,563; 4,182,766; 4,508,905; 4,628,098; 4,636,499; 4,689,333; 4,758,579; 4,783,974; 4,786,505; 4,808,596; 4,853,230; 5,026,560; 5,013,743; 5,035,899; 5,045,321; 5,045,552; 5,093,132; 5,093,342; 5,433,959; 5,464,632; 5,536,735; 5,576,025; 5,599,794; 5,629,305; 5,639,478; 5,690,960; 5,703,110; 5,705,517; 5,714,504; 5,731,006; 5,879,708; 5,900,424; 5,948,773; 5,997,903; 6,017
  • Particle size of the proton pump inhibitor can affect the solid dosage form in numerous ways. Because decreased particle size increases in surface area (S), the particle size reduction provides an increase in the rate of dissolution (dM/dt) as expressed in the Noyes-Whitney equation below:
  • the average particle size of at least about 90% the micronized proton pump inhibitor is less than about 200 ⁇ m, 150 ⁇ m, 100 ⁇ m, 80 ⁇ m, 60 ⁇ m, 40 ⁇ m, or less than about 35 ⁇ m, or less than about 30 ⁇ m, or less than about 25 ⁇ m, or less than about 20 ⁇ m, or less than about 15 ⁇ m, or less than about 10 ⁇ m.
  • At least 80% of the micronized proton pump inhibitor has an average particle size of less than about 200 ⁇ m, 150 ⁇ m, 100 ⁇ m, 80 ⁇ m, 60 ⁇ m, 40 ⁇ m, or less than about 35 ⁇ m, or less than about 30 ⁇ m, or less than about 25 ⁇ m, or less than about 20 ⁇ m, or less than about 15 ⁇ m, or less than about 10 ⁇ m.
  • At least 70% of the micronized proton pump inhibitor has an average particle size of less than about 200 ⁇ m, 150 ⁇ m, 100 ⁇ m, 80 ⁇ m, 60 ⁇ m, 40 ⁇ m, or less than about 35 ⁇ m, or less than about 30 ⁇ m, or less than about 25 ⁇ m, or less than about 20 ⁇ m, or less than about 15 ⁇ m, or less than about 10 ⁇ m.
  • the micronized proton pump inhibitor is of a size that allows greater than 50% of the proton pump inhibitor to be released within about 1 hour or 50 minutes, 40 minutes, 30 minutes, 20 minutes or 10 minutes of dissolution testing. In other embodiments, the micronized proton pump inhibitor allows greater than 25% of the proton pump inhibitor to be released within about 45 minutes, 30 minutes, or 15 minutes of dissolution testing.
  • compositions are provided wherein the micronized proton pump inhibitor is of a size which allows greater than 75% of the proton pump inhibitor to be released within about 1.5 hours, or within about 1.25 hours, or within about 1 hour, or within about 50 minutes, or within about 40 minutes, or within about 30 minutes, or within about 20 minutes, or within about 10 minutes, or within about 5 minutes of dissolution testing.
  • the micronized proton pump inhibitor is of a size which allows greater than 90% of the proton pump inhibitor to be released within about 1.5 hours, or within about 1.25 hours, or within about 1 hour, or within about 50 minutes, or within about 40 minutes, or within about 30 minutes, or within about 20 minutes, or within about 10 minutes, or within about 5 minutes of dissolution testing. See U.S.
  • the particle size of the proton pump inhibitor, antacid and excipients is an important factor which can effect bioavailability, blend uniformity, segregation, and flow properties.
  • smaller particle sizes of a drug increases the bioabsorption rate of the drug with substantially poor water solubility by increasing the surface area.
  • the particle size of the drug and excipients can also affect the suspension properties of the pharmaceutical formulation. For example, smaller particles are less likely to settle and therefore form better suspensions.
  • the average particle size of the dry powder (which can be administered directly, as a powder for suspension, or used in a solid dosage form) is less than about 500 microns in diameter, or less than about 450 microns in diameter, or less than about 400 microns in diameter, or less than about 350 microns in diameter, or less than about 300 microns in diameter, or less than about 250 microns in diameter, or less than about 200 microns in diameter, or less than about 150 microns in diameter, or less than about 100 microns in diameter, or less than about 75 microns in diameter, or less than about 50 microns in diameter, or less than about 25 microns in diameter, or less than about 15 microns in diameter.
  • the average particle size of the aggregates is between about 25 microns in diameter to about 300 microns in diameter. In still other embodiments, the average particle size of the aggregates is between about 25 microns in diameter to about 150 microns in diameter. And, in still further embodiments, the average particle size of the aggregates is between about 25 microns in diameter to about 100 microns in diameter.
  • the term “average particle size” is intended to describe the average diameter of the particles and/or agglomerates used in the pharmaceutical formulation.
  • the average particle size of the insoluble excipients is between about 5 ⁇ m to about 500 ⁇ m, or less than about 400 ⁇ m, or less than about 300 ⁇ m, or less than about 200 ⁇ m, or less than about 150 ⁇ m, or less than about 100 ⁇ m, or less than about 90 ⁇ m, or less than about 80 ⁇ m, or less than about 70 ⁇ m, or less than about 60 ⁇ m, or less than about 50 ⁇ m, or less than about 40 ⁇ m, or less than about 30 ⁇ m, or less than about 25 ⁇ m, or less than about 20 ⁇ m, or less than about 15 ⁇ m, or less than about 10 ⁇ m, or less than about 5 ⁇ m.
  • At least about 80% of the particles have a particle size of less than about 300 ⁇ m, or less than about 250 ⁇ m, or less than about 200 ⁇ m, or less than about 150 ⁇ m, or less than about 100 ⁇ m, or less than about 500 ⁇ m.
  • at least about 85% of the dry powder particles have a particle size of less than about 300 ⁇ m, or less than about 250 ⁇ m, or less than about 200 ⁇ m, or less than about 150 ⁇ m, or less than about 100 ⁇ m, or less than about 50 ⁇ m.
  • At least about 90% of the dry powder particles have a particle size of less than about 300 ⁇ m, or less than about 250 ⁇ m, or less than about 200 ⁇ m, or less than about 150 ⁇ m, or less than about 100 ⁇ m, or less than about 50 ⁇ m.
  • at least about 95% of the dry powder particles have a particle size of less than about 300 ⁇ m, or less than about 250 ⁇ m, or less than about 200 ⁇ m, or less than about 150 ⁇ m, or less than about 100 ⁇ m, or less than about 50 ⁇ m.
  • the average particle size of the insoluble material is between about 5 ⁇ m to about 250 ⁇ m in diameter. In other embodiments, the average particle size of the insoluble excipients is between about 5 ⁇ m to about 100 ⁇ m, or between about 5 ⁇ m to about 80 ⁇ m, or between about 5 ⁇ m to about 50 ⁇ m in diameter.
  • the particle size of other excipients is chosen to be about the same as the particle size of the antacid.
  • the particle size of the insoluble excipients is chosen to be about the same as the particle size of the proton pump inhibitor.
  • the excipient should be pharmaceutically acceptable. Also, in some examples, rapid dissolution and neutralization of gastric acid to maintain the gastric pH at about 6.5 for at least one hour.
  • the excipients which will be in contact with the proton pump inhibitor, if any, should also be chemically compatible with the proton pump inhibitor. “Chemically compatible” is intended to mean that the material does not lead to more than 10% degradation of the proton pump inhibitor when stored at room temperature for at least about 1 year.
  • Parietal cell activators are administered in an amount sufficient to produce the desired stimulatory effect without causing untoward side effects to patients.
  • the parietal cell activator is administered in an amount of about 5 mg to about 2.5 grams per 20 mg dose of the proton pump inhibitor.
  • the pharmaceutical composition of the invention comprises one or more antacids.
  • a class of antacids useful in the present invention include, e.g., antacids possessing pharmacological activity as a weak base or a strong base.
  • the antacid when formulated or delivered with an proton pump inhibiting agent, functions to substantially prevent or inhibit the acid degradation of the proton pump inhibitor by gastrointestinal fluid for a period of time, e.g., for a period of time sufficient to preserve the bioavailability of the proton pump inhibitor administered.
  • the antacid can be delivered before, during, and/or after delivery of the Proton Pump Inhibitor.
  • the antacid includes a salt of a Group IA metal, including, e.g., a bicarbonate salt of a Group IA metal (alkali metal), a carbonate salt of a Group IA metal, an alkali earth metal antacid (Group IIA metal), an aluminum antacid, a calcium antacid, or a magnesium antacid.
  • alkali metal a Group IA metal including, but not limited to, lithium, sodium, potassium, rubidium, cesium, and francium
  • alkaline earth metal Group IIA metal including, but not limited to, beryllium, magnesium, calcium, strontium, barium, radium
  • carbonates phosphates, bicarbonates, citrates, borates, acetates, phthalates, tartrate, succinates and the like, such as sodium or potassium phosphate, citrate, borate, acetate, bicarbonate and carbonate.
  • an antacid includes, e.g., an amino acid, an alkali salt of an amino acid, aluminum hydroxide, aluminum hydroxide/magnesium carbonate/calcium carbonate co-precipitate, aluminum magnesium hydroxide, aluminum hydroxide/magnesium hydroxide co-precipitate, aluminum hydroxide/sodium bicarbonate co-precipitate, aluminum glycinate, calcium acetate, calcium bicarbonate, calcium borate, calcium carbonate, calcium citrate, calcium gluconate, calcium glycerophosphate, calcium hydroxide, calcium lactate, calcium phthalate, calcium phosphate, calcium succinate, calcium tartrate, dibasic sodium phosphate, dipotassium hydrogen phosphate, dipotassium phosphate, disodium hydrogen phosphate, disodium succinate, dry aluminum hydroxide gel, L-arginine, magnesium acetate, magnesium aluminate, magnesium borate, magnesium bicarbonate, magnesium carbonate, magnesium citrate, magnesium gluconate, aluminum g
  • the antacids useful in the present invention also include antacids or combinations of antacids that interact with HCl (or other acids in the environment of interest) faster than the proton pump inhibitor interacts with the same acids. When placed in a liquid phase, such as water, these antacids produce and maintain a pH greater than the pKa of the proton pump inhibitor.
  • the antacid is selected from sodium bicarbonate, sodium carbonate, calcium carbonate, magnesium oxide, magnesium hydroxide, magnesium carbonate, aluminum hydroxide, and mixtures thereof.
  • the antacid is present in the pharmaceutical formulations of the present invention in an amount greater than about 5 mEq of antacid.
  • the antacid is present in the pharmaceutical formulations of the present invention in an amount greater than about 7 mEq of antacid. In other embodiments, the antacid is present in the pharmaceutical formulations of the present invention in an amount greater than about 10 mEq of antacid. In other embodiments, the antacid is present in the pharmaceutical formulations of the present invention in an amount greater than about 15 mEq of antacid. In other embodiments, the antacid is present in the pharmaceutical formulations of the present invention in an amount greater than about 20 mEq of antacid.
  • the antacid comprises sodium bicarbonate in about 0.1 mEq/mg proton pump inhibitor to about 5 mEq/mg proton pump inhibitor.
  • the antacid comprises a mixture of sodium bicarbonate and magnesium hydroxide, wherein the sodium bicarbonate and magnesium hydroxide are each present in about 0.1 mEq/mg proton pump inhibitor to about 5 mEq/mg proton pump inhibitor.
  • the antacid comprises a mixture of sodium bicarbonate, calcium carbonate, and magnesium hydroxide, wherein the sodium bicarbonate, calcium carbonate, and magnesium hydroxide are each present in about 0.1 mEq/mg proton pump inhibitor to about 5 mEq/mg of the proton pump inhibitor.
  • the antacid comprises a mixture of sodium bicarbonate and magnesium oxide, wherein the sodium bicarbonate and magnesium hydroxide are each present in about 0.1 mEq/mg proton pump inhibitor to about 5 mEq/mg proton pump inhibitor.
  • the antacid is present in an amount of about 0.1 mEq/mg to about 5 mEq/mg of the proton pump inhibitor, or about 0.5 mEq/mg to about 3 mEq/mg of the proton pump inhibitor, or about 0.6 mEq/mg to about 2.5 mEq/mg of the proton pump inhibitor, or about 0.7 mEq/mg to about 2.0 mEq/mg of the proton pump inhibitor, or about 0.8 mEq/mg to about 1.8 mEq/mg of the proton pump inhibitor, or about 1.0 mEq/mg to about 1.5 mEq/mg of the proton pump inhibitor, or at least 0.5 mEq/mg of the proton pump inhibitor.
  • the antacid is present in the pharmaceutical formulations of the present invention in an amount from about 5 to about 50 mEq of antacid. In other embodiments, the antacid is present in the pharmaceutical formulations of the present invention in an amount from about 5 to about 40 mEq of antacid. In other embodiments, the antacid is present in the pharmaceutical formulations of the present invention in an amount from about 10 to about 30 mEq of antacid. In other embodiments, the antacid is present in the pharmaceutical formulations of the present invention in an amount from about 10 to about 20 mEq of antacid. In other embodiments, the antacid is present in the pharmaceutical formulations of the present invention in an amount from about 5 to about 15 mEq of antacid.
  • the antacid is present in the pharmaceutical formulations of the present invention in an amount of about 0.1 mEq to about 15 mEq/mg of proton pump inhibitor, or about 0.1 mEq/mg of proton pump inhibitor, or about 0.5 mEq/mg of proton pump inhibitor, or about 1 mEq/mg of proton pump inhibitor, or about 2 mEq/mg of proton pump inhibitor, or about 2.5 mEq/mg of proton pump inhibitor, or about 3 mEq/mg of proton pump inhibitor, or about 3.5 mEq/mg of proton pump inhibitor, or about 4 mEq/mg of proton pump inhibitor, or about 4.5 mEq/mg of proton pump inhibitor, or about 5 mEq/mg of proton pump inhibitor, or about 6 mEq/mg of proton pump inhibitor, or about 7 mEq/mg of proton pump inhibitor, or about 8 mEq/mg of proton pump inhibitor
  • the antacid is present in the pharmaceutical formulations of the present invention in an amount of about 1 mEq to about 160 mEq per dose, or about 1 mEq, or about 5 mEq, or about 10 mEq, or about 15 mEq, or about 20 mEq, or about 25 mEq, or about 30 mEq, or about 35 mEq, or about 40 mEq, or about 45 mEq, or about 50 mEq, or about 60 mEq, or about 70 mEq, or about 80 mEq, or about 90 mEq, or about 100 mEq, or about 110 mEq, or about 120 mEq, or about 130 mEq, or about 140 mEq, or about 150 mEq, or about 160 mEq per dose.
  • the antacid is present in an amount of more than about 5 times, or more than about 10 times, or more than about 20 times, or more than about 30 times, or more than about 40 times, or more than about 50 times, or more than about 60 times, or more than about 70 times, or more than about 80 times, or more than about 90 times, or more than about 100 times the amount of the proton pump inhibiting agent on a weight to weight basis in the composition.
  • the amount of antacid present in the pharmaceutical formulation is between 200 and 3500 mg. In other embodiments, the amount of antacid present in the pharmaceutical formulation is about 200 mgs, or about 300 mgs, or about 400 mgs, or about 500 mgs, or about 600 mgs, or about 700 mgs, or about 800 mgs, or about 900 mgs, or about 1000 mgs, or about 1100 mgs, or about 1200 mgs, or about 1300 mgs, or about 1400 mgs, or about 1500 mgs, or about 1600 mgs, or about 1700 mgs, or about 1800 mgs, or about 1900 mgs, or about 2000 mgs, or about 2100 mgs, or about 2200 mgs, or about 2300 mgs, or about 2400 mgs, or about 2500 mgs, or about 2600 mgs, or about 2700 mgs, or about 2800 mgs, or about 2900 mgs, or about 3000 mgs, or about 3200 mgs, or about 3500 mgs.
  • the at least one buffering agent is a combination of two or more buffering agents
  • the combination comprises at least two non-amino acid buffering agents, wherein the combination of at least two non-amino acid buffering agents comprises substantially no aluminum hydroxide-sodium bicarbonate co-precipitate.
  • the pharmaceutical composition comprises an amino acid buffering agent
  • the total amount of buffering agent present in the pharmaceutical composition is less than about 5 mEq, or less than about 4 mEq, or less than about 3 mEq.
  • amino acid buffering agent as used herein includes amino acids, amino acid salts, and amino acid alkali salts.
  • non-amino acid buffering agent includes buffering agents as defined hereinabove but does not include amino acid buffering agents.
  • the pharmaceutical composition comprises substantially no or no poly[phosphoryl/sulfon]-ated carbohydrate and is in the form of a solid dosage unit.
  • a composition comprises a poly[phosphoryl/sulfon]-ated carbohydrate (e.g. sucralfate or sucrose octasulfate)
  • the weight ratio of poly[phosphoryl/sulfon]-ated carbohydrate to buffering agent is less than 1:5 (0.2), less than 1:10 (0.1) or less than 1:20 (0.05).
  • the poly[phosphoryl/sulfon]-ated carbohydrate is present in the composition, if at all, in an amount less than 50 mg, less than 25 mg, less than 10 mg or less than 5 mg.
  • the antacid is sodium bicarbonate and is present in about 0.1 mEq/mg proton pump inhibitor to about 5 mEq/mg proton pump inhibitor.
  • the antacid is a mixture of sodium bicarbonate and magnesium hydroxide, wherein the sodium bicarbonate and magnesium hydroxide are each present in about 0.1 mEq/mg proton pump inhibitor to about 5 mEq/mg proton pump inhibitor.
  • the antacid is a mixture of sodium bicarbonate and magnesium oxide, wherein the sodium bicarbonate and magnesium oxide are each present in about 0.1 mEq/mg proton pump inhibitor to about 5 mEq/mg proton pump inhibitor.
  • the term “soluble antacid” as used herein refers to an antacid that has a solubility of at least 500 mg/mL, or 300 mg/mL, or 200 mg/mL, or 100 mg/mL, or 50 mg/mL in the gastrointestinal fluid.
  • the soluble antacid is sodium bicarbonate.
  • Particle size of the buffer especially that an insoluble buffer can affect the onset of in-vivo neutralization of the stomach acid. Since decreased particle size increases in surface area, the particle size reduction provides an increase in the rate of acid neutralization, leading to superior protection of PPI from gastric acid degradation. On the other hand, extremely fine particle size of buffer will result in the powder mixture that is difficult to manufacture in commercial scale due to their poor flow and difficulties in processing (i.e., compression and encapsulation).
  • the antacid is a specific particle size.
  • the average particle size is no greater than 20 ⁇ m, or no greater than 30 ⁇ m, or no greater than 40 ⁇ m, or no greater than 50 ⁇ m, or no greater than 60 ⁇ m, or no greater than 70 ⁇ m, or no greater than 80 ⁇ m, or no greater than 90 ⁇ m or no greater than 100 ⁇ m in diameter.
  • At least about 70% of the antacid is no greater than 20 ⁇ m, or no greater than 30 ⁇ m, or no greater than 40 ⁇ m, or no greater than 50 ⁇ m, or no greater than 60 ⁇ m, or no greater than 70 ⁇ m, or no greater than 80 ⁇ m, or no greater than 90 ⁇ m or no greater than 100 ⁇ m in diameter.
  • At least about 85% of the antacid is no greater than 20 ⁇ m, or no greater than 30 ⁇ m, or no greater than 40 ⁇ m, or no greater than 50 ⁇ m, or no greater than 60 ⁇ m, or no greater than 70 ⁇ m, or no greater than 80 ⁇ m, or no greater than 90 ⁇ m or no greater than 100 ⁇ m in diameter.
  • the antacid is micronized.
  • particle size of at least 90% of antacid (D 90 ) is less than about 300 ⁇ m, or less than about 250 ⁇ m, or less than about 200 ⁇ m, or less than about 150 ⁇ m, or less than about 100 ⁇ m.
  • at least 75% of the antacid (D 75 ) has particle size of less than about 300 ⁇ m, or less than about 250 ⁇ m, or less than about 200 ⁇ m, or less than about 150 ⁇ m, or less than about 100 ⁇ m.
  • At least 50% of the antacid (D 50 ) has particle size of less than about 300 ⁇ m, or less than about 250 ⁇ m, or less than about 200 ⁇ m, or less than about 150 ⁇ m, or less than about 100 ⁇ m.
  • Spray dried antacid can also facilitate the speed of neutralization by fast reacting with acid upon contact.
  • Sprayed dried antacid typically has spherical particle shape which aids with achieving homogeneous blend during manufacturing process.
  • the antacid is spray dried with at least 15% of coating material such as maltodextrin or starch.
  • the antacid is spray dried with at least 10% of coating material such as maltodextrin or starch.
  • the antacid is spray dried with at least 5% of coating material such as maltodextrin or starch.
  • the antacid is spray dried with between about 1% to about 10% of a coating material.
  • the antacid is spray dried with about 5% of a coating material such as maltodextrin or starch.
  • Materials useful for enhancing the shelf-life of the pharmaceutical formulations of the present invention include materials compatible with the proton pump inhibitor of the pharmaceutical formulations which sufficiently isolate the proton pump inhibitor from other non-compatible excipients.
  • Materials compatible with the proton pump inhibitors of the present invention are those that enhance the shelf-life of the proton pump inhibitor, i.e., by slowing or stopping degradation of the proton pump inhibitor.
  • a pharmaceutical formulation of the present invention may have an enhanced shelf-life stability if, e.g., the microencapsulated proton pump inhibitor has less than about 0.5% degradation after one month of storage at room temperature, or less than about 1% degradation after one month at room temperature, or less than about 1.5% degradation after one month of storage at room temperature, or less than about 2% degradation after one month storage at room temperature, or less than about 2.5% degradation after one month of storage at room temperature, or less than about 3% degradation after one month of storage at room temperature.
  • a pharmaceutical formulation of the present invention may have an enhanced shelf-life stability if the pharmaceutical formulation contains less than about 5% total impurities after about 3 years of storage, or after about 2.5 years of storage, or about 2 years of storage, or about 1.5 years of storage, or about 1 year of storage, or after 11 months of storage, or after 10 months of storage, or after 9 months of storage, or after 8 months of storage, or after 7 months of storage, or after 6 months of storage, or after 5 months of storage, or after 4 months of storage, or after 3 months of storage, or after 2 months of storage, or after 1 month of storage.
  • pharmaceutical formulations of the present invention may have enhanced shelf-life stability if the pharmaceutical formulation contains less degradation of the proton pump inhibitor than proton pump inhibitor in the same formulation which is not microencapsulated or dry coated, or “bare”. For example, if bare proton pump inhibitor in the pharmaceutical formulation degrades at room temperature by more than about 2% after one month of storage and the microencapsulated or dry coated material degrades at room temperature by less than about 2% after one month of storage, then the proton pump inhibitor has been microencapsulated or dry coated with a compatible material that enhances the shelf-life of the pharmaceutical formulation.
  • the material useful for enhancing the shelf-life of the pharmaceutical formulations increases the shelf-life stability of the pharmaceutical formulation for at least about 5 days at room temperature, or at least about 10 days at room temperature, or at least about 15 days at room temperature, or at least about 20 days at room temperature, or at least about 25 days at room temperature, or at least about 30 days at room temperature or at least about 2 months at room temperature, or at least about 3 months at room temperature, or at least about 4 months at room temperature, or at least about 5 months at room temperature, or at least about 6 months at room temperature, or at least about 7 months at room temperature, or at least about 8 months at room temperature, or at least about 9 months at room temperature, or at least about 10 months at room temperature, or at least about 11 months at room temperature, or at least about one year at room temperature, or at least about 1.5 years at room temperature, or at least about 2 years at room temperature, or at least about 2.5 years at room temperature, or about 3 years at room temperature.
  • Exemplary microencapsulation materials useful for enhancing the shelf-life of pharmaceutical formulations comprising a proton pump inhibitor include, e.g., cellulose hydroxypropyl ethers (HPC) such as EF Klucel®, Nisso HPC and PrimaFlo HP22; low-substituted hydroxypropyl ethers (L-HPC); cellulose hydroxypropyl methyl ethers (HPMC) such as Seppifilm-LC, Pharmacoat®, Metolose SR, Opadry YS, PrimaFlo, MP3295A, Benecel MP824, and Benecel MP843; methylcellulose polymers such as Methocel® and Metolose®; Ethylcelluloses (EC) and mixtures thereof such as E461, Ethocel®, Aqualon®-EC, Surelease; Polyvinyl alcohol (PVA) such as Opadry AMB; hydroxyethylcelluloses such as Natrosol®; carboxymethylcelluloses and salts of carboxymethylcelluloses (
  • the microencapsulation material is selected from hydroxypropylcellulose and cellulose ethers. In still other embodiments, the microencapsulation material is selected from Klucel EF, Klucel EXF, Methocel E5, Methocel E15, and Methocel A15. In other embodiments, the material that enhances the shelf-life has a viscosity of 100-800 cps at 10% solution; or a viscosity of 200-600 cps at 10% solution; or a viscosity of 300-400 cps at 10% solution.
  • a buffering agent such as sodium bicarbonate is incorporated into the microencapsulation material.
  • an antioxidant such as BHT or BHA is incorporated into the microencapsulation material.
  • plasticizers such as polyethylene glycols, e.g., PEG 300, PEG 400, PEG 600, PEG 1450, PEG 3350, and PEG 800, stearic acid, propylene glycol, oleic acid, and triacetin are incorporated into the microencapsulation material.
  • the microencapsulating material useful for enhancing the shelf-life of the pharmaceutical formulations is from the USP or the National Formulary (NF).
  • one or more other compatible materials are present in the microencapsulation material.
  • exemplary materials include, e.g., parietal cell activators, organic solvents, erosion facilitators, diffusion facilitators, anti-adherents, anti-foaming agents, antioxidants, sweetening agents, flavoring agents, and carrier materials such as binders, suspending agents, disintegration agents, filing agents, surfactants, solubilizers, stabilizers, lubricants, wetting agents, and diluents.
  • the final formulation of the pharmaceutical formulation will be in the form of a tablet and at least about 50%, or at least about 55%, or at least about 60%, or at least about 65%, or at least about 70%, or at least about 75%, or at least about 80%, or at least about 85% or at least about 90%, or at least about 92%, or at least about 95%, or at least about 98%, or at least about 99% of the microspheres survive the tableting process, wherein microspheres that have survived the tableting process are those which provide the desired properties described herein.
  • the final formulation of the pharmaceutical formulation is in the form of a powder for oral suspension and the microencapsulation material surrounding the proton pump inhibitor will sufficiently dissolve in water, with or without stirring, in less than 1 hour, or less than 50 minutes, or less than 40 minutes, or less than 30 minutes, or less than 25 minutes, or less than 20 minutes, or less than 15 minutes, or less than 10 minutes or less than 5 minutes, or less than 1 minute.
  • Sufficiently dissolves means that at least about 50% of the encapsulation material has dissolved.
  • the microencapsulating material useful for enhancing the shelf-life of the pharmaceutical formulation sufficiently disintegrates to release the proton pump inhibitor into the gastrointestinal fluid of the stomach within less than about 1.5 hours, or within about 10 minutes, or within about 20 minutes, or within about 30 minutes, or within about or within about 40 minutes, or within about 50 minutes, or within about 1 hour, or within about 1.25 hours, or within about 1.5 hours after exposure to the gastrointestinal fluid.
  • Sufficiently disintegrates means that at least about 50% of the microencapsulation material has dissolved.
  • the average particle sizes of the microencapsulated drugs range from submicron to less than about 1,000 microns in diameter, or less than about 900 microns in diameter, or less than about 800 microns in diameter, or less than about 700 microns in diameter, or less than about 600 microns in diameter, or less than about 500 microns in diameter, or less than about 450 microns in diameter, or less than about 400 microns in diameter, or less than about 350 microns in diameter, or less than about 300 microns in diameter, or less than about 250 microns in diameter, or less than about 200 microns in diameter, or less than about 150 microns in diameter, or less than about 100 microns in diameter, or less than about 75 microns in diameter, or less than about 50 microns in diameter, or less than about 25 microns in diameter, or less than about 15 microns in diameter.
  • the average particle size of the aggregates is between about 25 microns in diameter to about 300 microns in diameter. In still other embodiments, the average particle size of the aggregates is between about 100 microns in diameter to about 200 microns in diameter. And in still further embodiments, the average particle size of the aggregates is between about 25 microns in diameter to about 100 microns in diameter.
  • the term “average particle size” is intended to describe the average diameter of the particles and/or agglomerates used in the pharmaceutical formulation.
  • Proton pump inhibitors are inherently bitter tasting and in one embodiment of the present invention, these bitter proton pump inhibitors are microencapsulated with a taste-masking material.
  • Materials useful Tor masking the taste of pharmaceutical formulations include those materials capable of microencapsulating the proton pump inhibitor, thereby protecting the senses from its bitter taste.
  • Taste-masking materials of the present invention provide superior pharmaceutical formulations by e.g., creating a more palatable pharmaceutical formulation as compared to pharmaceutical formulations and/or by creating a dosage form requiring less of the traditional flavoring or tastemasking agents.
  • flavor leadership criteria used to develop a palatable product include (1) immediate impact of identifying flavor, (2) rapid development of balanced, full flavor, (3) compatible mouth feel factors, (4) no “off” flavors, and (5) short aftertaste. See, e.g., Worthington, A Matter of Taste, Pharmaceutical Executive (April 2001).
  • the pharmaceutical formulations of the present invention improve upon one or more of these criteria.
  • Taste of a pharmaceutical formulation is important for both increasing patient compliance as well as for competing with other marketed products used for similar diseases, conditions and disorders.
  • Taste, especially bitterness is particularly important in pharmaceutical formulations for children since, because they cannot weigh the positive benefit of getting better against the immediate negative impact of the bitter taste in their mouth, they are more likely to refuse a drug that tastes bad. Thus, for pharmaceutical formulations for children, it becomes even more important to mask the bitter taste.
  • Microencapsulation of the proton pump inhibitor can (1) lower the amount of flavoring agents necessary to create a palatable product and/or (2) mask the bitter taste of the proton pump inhibitor by separating the drug from the taste receptors.
  • Taste-masking materials include, e.g., cellulose hydroxypropyl ethers (HPC) such as Klucel®, Nisswo HPC and PrimaFlo HP22; low-substituted hydroxypropyl ethers (L-HPC); cellulose hydroxypropyl methyl ethers (HPMC) such as Seppifilm-LC, Pharmacoat®, Metolose SR, Opadry YS, PrimaFlo, MP3295A, Benecel MP824, and Benecel MP843; methylcellulose polymers such as Methocel® and Metolose®; Ethylcelluloses (EC) and mixtures thereof such as E461, Ethocel®, Aqualon®-EC, Surelease; Polyvinyl alcohol (PVA) such as Opadry AMB; hydroxyethylcelluloses such as Natrosol®; carboxymethylcelluloses and salts of carboxymethylcelluloses (CMC) such as Aualon®-CMC; polyvinyl alcohol and polyethylene glycol
  • taste-masking materials contemplated are those described in U.S. Pat. Nos. 4,851,226, 5,075,114, and 5,876,759.
  • taste-masking materials see, e.g., Remington: The Science and Practice of Pharmacy , Nineteenth Ed. (Easton, Pa.: Mack Publishing Company, 1995); Hoover, John E., Remington's Pharmaceutical Sciences (Mack Publishing Co., Easton, Pa. 1975); Liberman, H. A. and Lachman, L., Eds., Pharmaceutical Dosage Forms (Marcel Decker, New York, N.Y., 1980); and Pharmaceutical Dosage Forms and Drug Delivery Systems , Seventh Ed. (Lippincott Williams & Wilkins 1999).
  • a pH modifier such as sodium carbonate or sodium bicarbonate is incorporated into the microencapsulation material.
  • an antioxidant such as BHT or BHA is incorporated into the microencapsulation material.
  • sucrose or sucralose is incorporated into the taste masking material.
  • plasticizers such as polyethylene glycol and/or stearic acid are incorporated into the microencapsulation material.
  • one or more other compatible materials are present in the microencapsulation material.
  • exemplary materials include, e.g., parietal cell activators, organic solvents, erosion facilitators, diffusion facilitators, anti-adherents, anti-foaming agents, antioxidants, flavoring agents, and carrier materials such as binders, suspending agents, disintegration agents, filing agents, surfactants, solubilizers, stabilizers, lubricants, wetting agents, diluents.
  • the pharmaceutical formulations of the present invention may also comprise one or more flavoring agents.
  • “Flavoring agents” or “sweeteners” useful in the pharmaceutical formulations of the present invention include, e.g., acacia syrup, acesulfame K, alitame, anise, apple, aspartame, banana, Bavarian cream, berry, black currant, butterscotch, calcium citrate, camphor, caramel, cherry, cherry cream, chocolate, cinnamon, bubble gum, citrus, citrus punch, citrus cream, cotton candy, cocoa, cola, cool cherry, cool citrus, cyclamate, cylamate, dextrose, eucalyptus, eugenol, fructose, fruit punch, ginger, glycyrrhetinate, glycyrrhiza (licorice) syrup, grape, grapefruit, honey, isomalt, lemon, lime, lemon cream, monoammonium glyrrhizinate (MagnaSweet®), maltol, mannitol, maple, marshmallow, menthol, mint
  • one or more flavoring agents are mixed with the taste-masking material prior to microencapsulating the proton pump inhibitor and, as such, are part of the taste-masking material.
  • the flavoring agent is mixed with the non-compatible excipients during the formulation process and is therefore not in contact with the proton pump inhibitor, and not part of the microencapsulation material.
  • an antacid such as sodium bicarbonate
  • the weight fraction of the taste masking material is, e.g., about 98% or less, about 95% or less, about 90% or less, about 85% or less, about 80% or less, about 75% or less, about 70% or less, about 65% or less, about 60% or less, about 55% or less, about 50% or less, about 45% or less, about 40% or less, about 35% or less, about 30% or less, about 25% or less, about 20% or less, about 15% or less, about 10% or less, about 5% or less, about 2%, or about 1% or less of the total weight of the pharmaceutical composition.
  • the amount of flavoring agent necessary to create a palatable product, as compared to a pharmaceutical formulation comprising non-microencapsulated proton pump inhibitor is decreased by 5% or less, or by 5% to 10%, or by 10% to 20%, or by 20% to 30%, or by 30% to 40%, or by 40% to 50%, or by 50% to 60%, or by 60% to 70%, or by 70% to 80%, or by 80% to 90%, or by 90% to 95%, or by greater than 95%.
  • no flavoring agent is necessary to create a more palatable pharmaceutical formulation as compared to a similar pharmaceutical formulation comprising non-microencapsulated proton pump inhibitor.
  • the total amount of flavoring agent present in the pharmaceutical formulation is less than 20 grams, or less than 15 grams, or less than 10 grams, or less than 8 grams, or less than 5 grams, or less than 4 grams, or less than 3.5 grams, or less than 3 grams, or less than 2.5 grams or less than 2 grams, or less than 1.5 grams, or less than 1 gram, or less than 500 mg, or less than 250 mg, or less than 150 mg, or less than 100 mg, or less than 50 mg.
  • the formulation comprises a microencapsulated omeprazole having approximately 37% omeprazole, the remainder being the immediate release coating designed to protect the micronized omeprazole from degradation by flavor components and other excipients.
  • the shelf life of a chewable product is thus improved. Additionally the flavor of the product can be enhanced, as the immediate release coating provides protection against a wide variety of acidic excipients.
  • the proton pump inhibitor may be microencapsulated by methods known by one of ordinary skill in the art. Such known methods include, e.g., spray drying processes, spinning disk processes, hot melt processes, spray chilling methods, fluidized bed, electrostatic deposition, centrifugal extrusion, rotational suspension separation, polymerization at liquid-gas or solid-gas interface, pressure extrusion, or spraying solvent extraction bath.
  • spray drying processes e.g., spray drying processes, spinning disk processes, hot melt processes, spray chilling methods, fluidized bed, electrostatic deposition, centrifugal extrusion, rotational suspension separation, polymerization at liquid-gas or solid-gas interface, pressure extrusion, or spraying solvent extraction bath.
  • several chemical techniques e.g., complex coacervation, solvent evaporation, polymer-polymer incompatibility, interfacial polymerization in liquid media, in situ polymerization, in-liquid drying, and desolvation in liquid media could also be used.
  • other methods such as dry granulation (i
  • the spinning disk method allows for: 1) an increased production rate due to higher feed rates and use of higher solids loading in feed solution, 2) the production of more spherical particles, 3) the production of a more even coating, and 4) limited clogging of the spray nozzle during the process.
  • the material used in the spray-dry encapsulation process is emulsified or dispersed into the core material in a concentrated form, e.g., 10-60% solids. In some embodiments of the present invention, the solid loading is between about 10-20%, or between about 10-40%, or between about 40-60%.
  • the microencapsulation material is, in one embodiment, is emulsified until about 1 to 3 ⁇ m droplets are obtained. In other embodiments, the microencapsulation material is emulsified until about 1 to 200 ⁇ m droplets are obtained, or until about 1 to 100 ⁇ m droplets are obtained.
  • the median droplet size of the microencapsulation material is between about 1 to about 300 ⁇ m, or between about 1 to about 200 ⁇ m, or between about 50 to about 150 ⁇ m.
  • Coacervation involves microencapsulation of materials such as active pharmaceutical ingredients and involves a three part process of particle or droplet formation, coacerate wall formation, and capsule isolation. This method can produce very small particle size microcapsules (10-70 microns).
  • Extrusion/spheronization is another method that involves wet massing of active pharmaceutical ingredients, followed by the extrusion of the wet mass through a perforated plate to produce short cylindrical rods. These rods are subsequently placed into a rapidly rotating spheronizer to shape the cylindrical rods into uniform spheres. The spheres are subsequently dried using a fluid bed drier and then coated with a functional coating using a fluid bed equipped with a Wurster insert and spray nozzle. This method produces smooth, uniform spheres that are ideal for receiving a functional coating. Drug loadings as high as 80% are possible (depending on drug characteristics).
  • the microspheres have irregular geometries. In other embodiments, the microspheres are aggregates of smaller particles.
  • the drug loading of the proton pump inhibitor in the microspheres is greater than 1%, greater than 2.5%, greater than 5%, greater than 10%, greater than 15%, greater than 20%, greater than 25%, greater than 30%, greater than 35%, greater than 40%, greater than 45%, greater than 50%, greater than 55%, greater than 60%, greater than 65%, greater than 70%, greater than 75%, greater than 80% weight percent of the proton pump inhibitor to the microencapsulated drug.
  • the drug loading of the proton pump inhibitor in the microspheres is between about 5-60 wt %, or about 5-50 wt-%, or about 10-40 wt-%.
  • the stability of the proton pump inhibitors used in the present invention may be increased by alternative methods such as dry coating and nano-particle coating.
  • Dry coating involves the formation of granules of coated proton pump inhibitor which are then mixed with other components. Dry granulation is achieved by forming dense compacts which are subsequently milled to a desired particle size and then blended with other components of the pharmaceutical composition. Dry granulation and nano-particle coating can provide enhanced stability and tastemasking characteristics to active pharmaceutical by diluting and isolating such components in a granulated matrix of compatible ingredients that can enhance the shelf life of proton pump inhibitor products as well as tastemask the bitterness if sweetener or flavors are used in coating material.
  • Typical technique for dry granulation is to use slugging or roller compaction.
  • the dry powders are compressed using a conventional tablet machine, or more usually, a large heavy duty rotary press.
  • the resulting compacts or “slug” are then milled to a desired particle size.
  • Roller compaction is an alternative gentler method, the powder mix being squeezed between two rollers to form a compressed sheet.
  • the sheet normally is weak and brittle and breaks immediately into flakes. These flakes need gentler treatment to break them into granules, and this can be usually be achieved by screening alone. Parikh, D. M., Handbook of Pharmaceutical Granulation Technology , (Marcel Dekker ed. 1997).
  • Nano particle coating is another method that involves a nano particle deposited onto the drug core using Physical Vapour Deposition (PVD) methods.
  • PVD Physical Vapour Deposition
  • This method can coat a 10-20 micro drug core with various thickness of metal, or metal salt (e.g., SB, MgO, MgOH, CaCO 3 , etc.).
  • the average particle sizes of the dry coated proton pump inhibitor ranges from submicron to less than about 1,000 microns in diameter, or less than about 900 microns in diameter, or less than about 800 microns in diameter, or less than about 700 microns in diameter, or less than about 600 microns in diameter, or less than about 500 microns in diameter, or less than about 450 microns in diameter, or less than about 400 microns in diameter, or less than about 350 microns in diameter, or less than about 300 microns in diameter, or less than about 250 microns in diameter, or less than about 200 microns in diameter, or less than about 150 microns in diameter, or less than about 100 microns in diameter, or less than about 75 microns in diameter, or less than about 50 microns in diameter, or less than about 25 microns in diameter, or less than about 15 microns in diameter.
  • the average particle size of the aggregates is between about 25 microns in diameter to about 300 microns in diameter. In still other embodiments, the average particle size of the aggregates is between about 100 microns in diameter to about 200 microns in diameter. And in still further embodiments, the average particle size of the aggregates is between about 25 microns in diameter to about 100 microns in diameter.
  • the term “average particle size” is intended to describe the average diameter of the particles and/or agglomerates used in the pharmaceutical formulation.
  • the dry coated proton pump inhibitor granules are less than about 2000 microns, or less than about 1500 microns, or less than about 1000 microns. In some embodiments, the average particle size of the dry coated proton pump inhibitor granules is between about 100 to about 2000 microns, or between about 100 to about 1000 microns, or between about 200 to about 800 microns, or between about 300 to about 600 microns.
  • the dry coated proton pump inhibitor granules comprise antacid, binder, lubricant and/or sweeteners.
  • the antacid is sodium bicarbonate.
  • the binder is hydroxypropyl cellulose.
  • the sweetener is sucralose and/or xylitab.
  • the lubricant is magnesium stearate.
  • the dry coated proton pump inhibitor is combined with additional antacid.
  • the additional antacid is the same antacid as used in the material used to dry coat the proton pump inhibitor.
  • the antacid is a different antacid.
  • the antacid is a combination of two or more antacids.
  • one or more pharmaceutically acceptable excipients are mixed with the dry coated proton pump inhibitor to form the pharmaceutical composition.
  • the additional pharmaceutical excipients include one or more flavors.
  • one or more other compatible materials are present in the dry coating material.
  • exemplary materials include, e.g., parietal cell activators, organic solvents, erosion facilitators, diffusion facilitators, anti-adherents, anti-foaming agents, antioxidants, flavoring agents, and carrier materials such as binders, suspending agents, disintegration agents, filing agents, surfactants, solubilizers, stabilizers, lubricants, wetting agents, diluents.
  • the additional compatible materials are binders, lubricants and sweeteners.
  • one or more sweeteners are incorporated into the material used to dry coat the proton pump inhibitor.
  • one or more flavoring agents are incorporated into the material used to dry coat the proton pump inhibitor.
  • the material used to dry coat the proton pump inhibitor comprises a sweetener and/or a flavoring agent. In some embodiments, this dry coated proton pump inhibitor is then mixed with additional sweeteners and/or flavoring agents.
  • “Flavoring agents” or “sweeteners” useful in the pharmaceutical formulations of the present invention include, e.g., acacia syrup, acesulfame K, alitame, anise, apple, aspartame, banana, Bavarian cream, berry, black currant, butterscotch, calcium citrate, camphor, caramel, cherry, cherry cream, chocolate, cinnamon, bubble gum, citrus, citrus punch, citrus cream, cotton candy, cocoa, cola, cool cherry, cool citrus, cyclamate, cylamate, dextrose, eucalyptus, eugenol, fructose, fruit punch, ginger, glycyrrhetinate, glycyrrhiza (licorice) syrup, grape, grapefruit, honey, isomalt, lemon, lime, lemon cream, monoammonium glyrrhizinate (MagnaSweet®), maltol, mannitol, maple, marshmallow, menthol, mint
  • the weight percent of the proton pump inhibitor in the dry coated granules is about 2-70%. In some embodiments, the weight percent of the proton pump inhibitor in the dry coated granules is about 5-50%, or about 5-30%. In yet other embodiments, the weight percent of the proton pump inhibitor in the granules is about 5%, or about 7%, or about 10%, or about 15%, or about 20%, or about 25%, or about 30%, or about 35%, or about 40%.
  • compositions and methods described herein as containing microencapsulated proton pump inhibitors can, in addition to or in the alternative, contain dry coated proton pump inhibitors.
  • the acid neutralizing capacity and pH profile of various antacid combinations can be evaluated by using an in-vitro stomach model.
  • Several of these simulated dynamic models are known in the art. See, e.g., Smyth et al., Correlation of In-Vivo Methodology for Evaluation of Antacids, J. Pharm. Sci. Vol. 65, 1045 (1976); Hobert, Fordham et al., In-Vivo Evaluation of Liquid Antacids, New England Journal of Med.
  • the antacid increases the gastric pH to at least about 3.5 for no more than about 90 minutes as measured by a simulated stomach model such as Fuch's kinetic in-vitro pH model. In other embodiments, the antacid increases the pH to at least about 3.5 for no more than about 60 minutes. In still other embodiments, the antacid increases the pH to at least about 3.5 for no more than 45 minutes. Depending on the buffer system used (i.e., type of antacid and amount) some embodiments of the present invention, the antacid increases the gastric pH to at least about 3.5 for no more than about 30 minutes as measured by a simulated stomach model such as Fuchs' kinetic in-vitro pH model.
  • the antacid increases the gastric pH to at least about 3.5 for less than about 25 minutes as measured by a simulated stomach model such as Fuch's kinetic in-vitro pH model. In yet other embodiments, the antacid increases the gastric pH to at least about 3.5 for less than about 20 minutes, or less than about 15 minutes, or less than about 10 minutes as measured by a stimulated stomach model such as Fuch's kinetic in-vitro pH model.
  • the antacid protects at least some of the proton pump inhibitor and a therapeutically effective amount of the proton pump inhibitor is delivered to the subject.
  • the proton pump inhibiting agent is administered and dosed in accordance with good medical practice, taking into account the clinical condition of the individual patient, the site and method of administration, scheduling of administration, and other factors known to medical practitioners.
  • human therapy it is important to provide a dosage form that delivers the required therapeutic amount of the drug in vivo, and renders the drug bioavailable in a rapid manner.
  • the dosage forms described by Phillips et al. in U.S. Pat. Nos. 5,840,737, 6,489,346, 6,699,885, and 6,645,988 are incorporated herein by reference.
  • the percent of intact drug that is absorbed into the bloodstream is not narrowly critical, as long as a therapeutic-disorder-effective amount, e.g., a gastrointestinal-disorder-effective amount of a proton pump inhibiting agent, is absorbed following administration of the pharmaceutical composition to a subject. It is understood that the amount of proton pump inhibiting agent and/or antacid that is administered to a subject is dependent on, e.g., the sex, general health, diet, and/or body weight of the subject.
  • a relatively low amount of the proton pump inhibitor e.g., about 1 mg to about 30 mg, will often provide blood serum concentrations consistent with therapeutic effectiveness.
  • achievement of a therapeutically effective blood serum concentration will require larger dosage units, e.g., about 10 mg, about 15 mg, about 20 mg, about 30 mg, about 40 mg, about 80 mg, or about 120 mg dose for an adult human, or about 150 mg, or about 200 mg, or about 400 mg, or about 800 mg, or about 1000 mg dose, or about 1500 mg dose, or about 2000 mg dose, or about 2500 mg dose, or about 3000 mg dose, or about 3200 mg dose, or about 3500 mg dose for an adult horse.
  • larger dosage units e.g., about 10 mg, about 15 mg, about 20 mg, about 30 mg, about 40 mg, about 80 mg, or about 120 mg dose for an adult human, or about 150 mg, or about 200 mg, or about 400 mg, or about 800 mg, or about 1000 mg dose, or about 1500 mg dose, or about 2000 mg dose, or about 2500 mg dose, or about 3000 mg dose, or about 3200 mg dose, or about 3500 mg dose for an adult horse.
  • the amount of proton pump inhibitor administered to a subject is, e.g., about 1-2 mg/Kg of body weight, or about 0.5 mg/Kg of body weight, or about 1 mg/Kg of body weight, or about 1.5 mg/Kg of body weight, or about 2 mg/Kg of body weight.
  • Treatment dosages generally may be titrated to optimize safety and efficacy. Typically, dosage-effect relationships from in vitro and/or in vivo tests initially can provide useful guidance on the proper doses for subject administration. Studies in animal models generally may be used for guidance regarding effective dosages for treatment of gastrointestinal disorders or diseases in accordance with the present invention. In terms of treatment protocols, it should be appreciated that the dosage to be administered will depend on several factors, including the particular agent that is administered, the route chosen for administration, the condition of the particular subject.
  • unit dosage forms for humans contain about 1 mg to about 120 mg, or about 1 mg, or about 5 mg, or about 10 mg, or about 15 mg, or about 20 mg, or about 30 mg, or about 40 mg, or about 50 mg, or about 60 mg, or about 70 mg, or about 80, mg, or about 90 mg, or about 100 mg, or about 110 mg, or about 120 mg of a proton pump inhibitor.
  • the pharmaceutical formulation is administered in an amount to achieve a measurable serum concentration of a non-acid degraded proton pump inhibiting agent greater than about 100 ng/ml within about 30 minutes after administration of the pharmaceutical formulation.
  • the pharmaceutical formulation is administered to the subject in an amount to achieve a measurable serum concentration of a non-acid degraded or non-acid reacted proton pump inhibiting agent greater than about 100 ng/ml within about 15 minutes after administration of the pharmaceutical formulation.
  • the pharmaceutical formulation is administered to the subject in an amount to achieve a measurable serum concentration of a non-acid degraded or non-acid reacted proton pump inhibiting agent greater than about 100 ng/ml within about 10 minutes after administration of the pharmaceutical formulation.
  • the composition is administered to the subject in an amount to achieve a measurable serum concentration of the proton pump inhibiting agent greater than about 150 ng/ml within about 15 minutes and to maintain a serum concentration of the proton pump inhibiting agent of greater than about 150 ng/ml from about 15 minutes to about 1 hour after administration of the composition.
  • the composition is administered to the subject in an amount to achieve a measurable serum concentration of the proton pump inhibiting agent greater than about 250 ng/ml within about minutes and to maintain a serum concentration of the proton pump inhibiting agent of greater than about 150 ng/ml from about 15 minutes to about 1 hour after administration of the composition.
  • the composition is administered to the subject in an amount to achieve a measurable serum concentration of the proton pump inhibiting agent greater than about 350 ng/ml within about 15 minutes and to maintain a serum concentration of the proton pump inhibiting agent of greater than about 150 ng/ml from about 15 minutes to about 1 hour after administration of the composition.
  • the composition is administered to the subject in an amount to achieve a measurable serum concentration of the proton pump inhibiting agent greater than about 450 ng/ml within about 15 minutes and to maintain a serum concentration of the proton pump inhibiting agent of greater than about 150 ng/m1 from about 15 minutes to about 1 hour after administration of the composition.
  • the composition is administered to the subject in an amount to achieve a measurable serum concentration of the proton pump inhibiting agent greater than about 150 ng/ml within about 30 minutes and to maintain a serum concentration of the proton pump inhibiting agent of greater than about 150 ng/ml from about 30 minutes to about 1 hour after administration of the composition.
  • the composition is administered to the subject in an amount to achieve a measurable serum concentration of the proton pump inhibiting agent greater than about 250 ng/ml within about 30 minutes and to maintain a serum concentration of the proton pump inhibiting agent of greater than about 150 ng/ml from about 30 minutes to about 1 hour after administration of the composition.
  • the composition is administered to the subject in an amount to achieve a measurable serum concentration of the proton pump inhibiting agent greater than about 350 ng/ml within about 30 minutes and to maintain a serum concentration of the proton pump inhibiting agent of greater than about 150 ng/ml from about 30 minutes to about 1 hour after administration of the composition.
  • the composition is administered to the subject in an amount to achieve a measurable serum concentration of the proton pump inhibiting agent greater than about 450 ng/ml within about 30 minutes and to maintain a serum concentration of the proton pump inhibiting agent of greater than about 150 ng/ml from about 30 minutes to about 1 hour after administration of the composition.
  • the composition is administered to the subject in an amount to achieve a measurable serum concentration of a non-acid degraded or non-acid reacted proton pump inhibiting agent greater than about 500 ng/ml within about 1 hour after administration of the composition.
  • the composition is administered to the subject in an amount to achieve a measurable serum concentration of a non-acid degraded or non-acid reacted proton pump inhibiting agent greater than about 300 ng/ml within about 45 minutes after administration of the composition.
  • the composition is administered to the subject in an amount sufficient to achieve a maximum serum concentration (Cmax) at a time (Tmax) that is within about 90, 70, 60, 50, 40, 30 or 20 minutes after administration of the composition according to the present invention.
  • the composition is administered to the subject in an amount sufficient to achieve a maximum serum concentration (Cmax) at a time (Tmax) that is between about 10 and about 90 minutes, between about 10 to about 60 minutes, between about 15 to about 60 minutes or between about 20 to about 60 minutes after administration of the composition according to the present invention.
  • Cmax maximum serum concentration
  • Tmax time
  • the values of Cmax and Tmax are averages over a test population. In other specific embodiments, the values of Cmax and Tmax are the values for an individual.
  • the composition is administered in an amount sufficient to achieve a maximum serum concentration (Cmax) of from about 400 to about 3000 ng/mL, from about 400 to about 2500 ng/mL, from about 400 to about 2000 ng/mL, from about 400 to about 1500 ng/mL, from about 1000 to about 1500 ng/mL, from about 400 to about 1000 ng/mL or from about 400 to about 700 ng/mL.
  • Cmax serum concentration
  • the values of Cmax and Tmax are averages over a test population. In other specific embodiments, the values of Cmax and Tmax are the values for an individual.
  • the composition is administered in an amount sufficient to achieve a maximum serum concentration (Cmax) of greater than 400 ng/mL, greater than 600 ng/mL, greater than 1000 ng/mL.
  • Cmax maximum serum concentration
  • the values of Cmax and Tmax are averages over a test population. In other specific embodiments, the values of Cmax and Tmax are the values for an individual.
  • Contemplated compositions of the present invention provide a therapeutic effect as proton pump inhibiting agent medications over an interval of about 5 minutes to about 24 hours after administration, enabling, for example, once-a-day, twice-a-day, three times a day, etc. administration if desired.
  • a therapeutic effect as proton pump inhibiting agent medications over an interval of about 5 minutes to about 24 hours after administration, enabling, for example, once-a-day, twice-a-day, three times a day, etc. administration if desired.
  • one will desire to administer an amount of the compound that is effective to achieve a serum level commensurate with the concentrations found to be effective in vivo for a period of time effective to elicit a therapeutic effect. Determination of these parameters is well within the skill of the art. These considerations are well known in the art and are described in standard textbooks.
  • the composition is administered to a subject in a gastrointestinal-disorder-effective amount, that is, the composition is administered in an amount that achieves a therapeutically-effective dose of a proton pump inhibiting agent in the blood serum of a subject for a period of time to elicit a desired therapeutic effect.
  • a therapeutically-effective dose of a proton pump inhibiting agent in the blood serum of a subject for a period of time to elicit a desired therapeutic effect.
  • the composition is administered to achieve a therapeutically-effective dose of a proton pump inhibiting agent in the blood serum of a subject within about 45 minutes after administration of the composition.
  • a therapeutically-effective dose of the proton pump inhibiting agent is achieved in the blood serum of a subject within about 30 minutes from the time of administration of the composition to the subject. In yet another embodiment, a therapeutically-effective dose of the proton pump inhibiting agent is achieved in the blood serum of a subject within about 20 minutes from the time of administration to the subject. In still another embodiment of the present invention, a therapeutically-effective dose of the proton pump inhibiting agent is achieved in the blood serum of a subject at about 15 minutes from the time of administration of the composition to the subject.
  • greater than about 98%; or greater than about 95%; or greater than about 90%; or greater than about 75%; or greater than about 50% of the drug absorbed into the bloodstream is in a non-acid degraded or a non-acid reacted form.
  • the pharmaceutical formulations provide a release profile of the proton pump inhibitor, using USP dissolution methods, whereby greater than about 50% of the proton pump inhibitor is released from the composition within about 2 hours; or greater than 50% of the proton pump inhibitor is released from the composition within about 1.5 hours; or greater than 50% of the proton pump inhibitor is released from the composition within about 1 hour after exposure to gastrointestinal fluid.
  • greater than about 60% of the proton pump inhibitor is released from the composition within about 2 hours; or greater than 60% of the proton pump inhibitor is released from the composition within about 1.5 hours; or greater than 60% of the proton pump inhibitor is released from the composition within about 1 hour after exposure to gastrointestinal fluid.
  • greater than about 70% of the proton pump inhibitor is released from the composition within about 2 hours; or greater than 70% of the proton pump inhibitor is released from the composition within about 1.5 hours; or greater than 70% of the proton pump inhibitor is released from the composition within about 1 hour after exposure to gastrointestinal fluid.
  • the pharmaceutical formulations of the present invention contain desired amounts of microencapsulated proton pump inhibitor and/or dry coated proton pump inhibitor and antacid can be in the form of, e.g., a tablet; including a suspension tablet, a chewable tablet, an effervescent tablet or caplet; a pill; a powder such as a sterile packaged powder, a dispensable powder, and an effervescent powder; a capsule including both soft or hard gelatin capsules such as HPMC capsules; a lozenge; a sachet; a troche; pellets; granules; or aerosol.
  • These pharmaceutical formulations of the present invention can be manufactured by conventional pharmacological techniques.
  • buffers The amount and types of buffers, proton pump inhibitors, and other excipients useful in each of these dosage forms are described throughout the specification and examples. It should be recognized that where a combination of buffer, proton pump inhibitor and/or excipient, including specific amounts of these components, is described with one dosage form that the same combination could be used for any other suitable dosage form. Moreover, it should be understood that one of skill in the art would, with the teachings found within this application, be able to make any of the dosage forms listed above by combining the components (i.e., amounts and types of PPIs, buffers, and other excipients) described in the different sections of the specification.
  • each of the dosage forms may comprise one or more additional materials such as a pharmaceutically compatible carrier, binder, filling agent, suspending agent, flavoring agent, sweetening agent, disintegrating agent, surfactant, preservative, lubricant, colorant, diluent, solubilizer, moistening agent, stabilizer, wetting agent, anti-adherent, parietal cell activator, anti-foaming agent, antioxidant, chelating agent, antifungal agent, antibacterial agent, or one or more combination thereof.
  • additional materials such as a pharmaceutically compatible carrier, binder, filling agent, suspending agent, flavoring agent, sweetening agent, disintegrating agent, surfactant, preservative, lubricant, colorant, diluent, solubilizer, moistening agent, stabilizer, wetting agent, anti-adherent, parietal cell activator, anti-foaming agent, antioxidant, chelating agent, antifungal agent, antibacterial agent, or one or more combination thereof.
  • Parietal cell activators are administered in an amount sufficient to produce the desired stimulatory effect without causing untoward side effects to patients.
  • the parietal cell activator is administered in an amount of about 5 mg to about 2.5 grams per 20 mg dose of the proton pump inhibitor.
  • compositions of the present invention can be manufactured by conventional pharmacological techniques.
  • Conventional pharmacological techniques include, e.g., one or a combination of methods: (1) dry mixing, (2) direct compression, (3) milling, (4) dry or non-aqueous granulation, (5) wet granulation, or (6) fusion. See, e.g., Lachman et al., The Theory and Practice of Industrial Pharmacy (1986).
  • Other methods include, e.g., prilling, spray drying, pan coating, melt granulation, granulation, wurster coating, tangential coating, top spraying, extruding, coacervation and the like.
  • the proton pump inhibitor is microencapsulated or dry coated prior to being formulated into one of the above forms.
  • some or all of the antacid is also microencapsulated prior to being further formulated into one of the above forms.
  • a film coating is provided around the pharmaceutical formulation.
  • compositions wherein some or all of the proton pump inhibitor and some or all of the antacid are microencapsulated or dry coated. In some embodiments, only some of the proton pump inhibitor is microencapsulated or dry coated. In other embodiments, all of the proton pump inhibitor is microencapsulated or dry coated. In still other embodiments, only some of the antacid is microencapsulated or dry coated.
  • one or more layers of the pharmaceutical formulation are plasticized.
  • a plasticizer is generally a high boiling point solid or liquid. Suitable plasticizers can be added from about 0.01% to about 50% by weight (w/w) of the coating composition.
  • Plasticizers include, e.g., diethyl phthalate, citrate esters, polyethylene glycol, glycerol, acetylated glycerides, triacetin, polypropylene glycol, polyethylene glycol, triethyl citrate, dibutyl sebacate, stearic acid, stearol, stearate, and castor oil.
  • the pharmaceutical compositions of the present invention contain desired amounts of proton pump inhibiting inhibitor and antacid and are in a solid dosage form. In other embodiments, the pharmaceutical compositions of the present invention contain desired amounts of proton pump inhibitor and antacid and are administered in the form of a capsule (including both soft or hard capsules, e.g., capsules made from animal-derived gelatin or plant-derived HPMC).
  • Solid compositions e.g., tablets, chewable tablets, effervescent tablets, and capsules, are prepared by mixing the microencapsulated proton pump inhibitor or dry coated proton pump inhibitor with one or more antacid and pharmaceutical excipients to form a bulk blend composition.
  • dry coated proton pump inhibitor mixing with additional antacid is optional.
  • these bulk blend compositions as homogeneous, it is meant that the microencapsulated or dry coated proton pump inhibitor and antacid are dispersed evenly throughout the composition so that the composition may be readily subdivided into equally effective unit dosage forms, such as tablets, pills, and capsules.
  • the individual unit dosages may also comprise film coatings, which disintegrate upon oral ingestion or upon contact with diluent.
  • Compressed tablets are solid dosage forms prepared by compacting the bulk blend compositions described above.
  • compressed tablets of the present invention will comprise one or more flavoring agents.
  • the compressed tablets will comprise a film surrounding the final compressed tablet.
  • the compressed tablets comprise one or more excipients and/or flavoring agents.
  • a capsule may be prepared, e.g., by placing the bulk blend composition, described above, inside of a capsule.
  • the pharmaceutical compositions of the present invention contain desired amounts of proton pump inhibiting inhibitor and antacid and are in a solid dosage form.
  • the pharmaceutical compositions of the present invention contain desired amounts of proton pump inhibitor and antacid and are administered in the form of a capsule (including both soft and hard capsules, e.g., capsules made from animal-derived gelatin or plant-derived HPMC).
  • the pharmaceutical compositions of the present invention can be manufactured by conventional pharmacological techniques.
  • a chewable tablet may be prepared by compacting bulk blend compositions, described above.
  • the chewable tablet comprises a material useful for enhancing the shelf-life of the pharmaceutical formulation.
  • the proton pump inhibitor is dry coated.
  • microencapsulated material has taste-masking properties.
  • the chewable tablet comprises one or more flavoring agents and one ore more taste-masking materials.
  • the chewable tablet comprises both a material useful for enhancing the shelf-life of the pharmaceutical formulation and one or more flavoring agents.
  • the microencapsulated or dry coated proton pump inhibitor, antacid, and optionally one or more excipients are dry blended and compressed into a mass, such as a tablet, having a hardness sufficient to provide a pharmaceutical composition that substantially disintegrates within less than about 30 minutes, less than about 35 minutes, less than about 40 minutes, less than about 45 minutes, less than about 50 minutes, less than about 55 minutes, or less than about 60 minutes, after oral administration, thereby releasing the antacid and the proton pump inhibitor into the gastrointestinal fluid.
  • a mass such as a tablet
  • chewable tablets are prepared using micronized proton pump inhibitor that has been combined with various combinations of potential excipients.
  • micronized omeprazole is combined with various colorants, flavorings and other taste masking excipients.
  • acidity in particular colorants, flavorings or other taste masking excipients can give rise to instability of the proton pump inhibitor.
  • some embodiments provide for encapsulation of micronized proton pump inhibitor in an immediate release coating.
  • the immediate release coating is designed to protect the micronized proton pump inhibitor from degradation by acidic excipients, such as flavor components.
  • a powder for suspension may be prepared by combining microencapsulated proton pump inhibitor and one or more antacid.
  • the powder may comprise one or more pharmaceutical excipients.
  • the proton pump inhibitor is micronized.
  • Other embodiments of the present invention also comprise a suspending agent and/or a wetting agent.
  • Effervescent powders are also prepared in accordance with the present invention.
  • Effervescent salts have been used to disperse medicines in water for oral administration.
  • Effervescent salts are granules or coarse powders containing a medicinal agent in a dry mixture, usually composed of sodium bicarbonate, citric acid and/or tartaric acid.
  • a medicinal agent in a dry mixture, usually composed of sodium bicarbonate, citric acid and/or tartaric acid.
  • Examples of effervescent salts include the following ingredients: sodium bicarbonate or a mixture of sodium bicarbonate and sodium carbonate, citric acid and/or tartaric acid. Any acid-base combination that results in the liberation of carbon dioxide can be used in place of the combination of sodium bicarbonate and citric and tartaric acids, as long as the ingredients were suitable for pharmaceutical use and result in a pH of about 6 or higher.
  • the method of preparation of the effervescent granules of the present invention employs three basic processes: wet granulation, dry granulation and fusion.
  • the fusion method is used for the preparation of most commercial effervescent powders. It should be noted that, although these methods are intended for the preparation of granules, the formulations of effervescent salts of the present invention could also be prepared as tablets, according to known technology for tablet preparation.
  • wet granulation is one of the oldest methods of granule preparation.
  • the individual steps in the wet granulation process of tablet preparation include milling and sieving of the ingredients, dry powder mixing, wet massing, granulation, and final grinding.
  • the microencapsulated omeprazole is added to the other excipients of the pharmaceutical formulation after they have been wet granulated.
  • Dry granulation by slugging involves compressing a powder mixture into a rough tablet or “slug” on a heavy-duty rotary tablet press. The slugs are then broken up into granular particles by a grinding or milling operation, usually by passage through an oscillation granulator. The individual steps include mixing of the powders, compressing (slugging) and grinding (slug reduction or granulation). In a larger scale operation, roller compaction can be used instead of “slugging”. Roller compaction procedure is well-known to ones skilled in the art. No wet binder or moisture is involved in any of the dry granulation steps.
  • the microencapsulated omeprazole is dry granulated with other excipients in the pharmaceutical formulation. In other embodiments, the microencapsulated omeprazole is added to other excipients of the pharmaceutical formulation after they have been dry granulated.
  • compositions suitable for buccal (sublingual) administration include, e.g., lozenges in a flavored base, such as sucrose, acacia, tragacanth, and pastilles comprising microencapsulated proton pump inhibitor in an inert base such as gelatin, glycerin, sucrose, and acacia are also provided herein.
  • release delivery systems include, e.g., polymer-based systems, such as polylactic and polyglycolic acid, plyanhydrides and polycaprolactone; nonpolymer-based systems that are lipids, including sterols, such as cholesterol, cholesterol esters and fatty acids, or neutral fats, such as mono-, di- and triglycerides; hydrogel release systems; silastic systems; peptide-based systems; wax coatings; compressed tablets using conventional binders and excipients partially fused implants and the like. See, e.g., Liberman et al., Pharmaceutical Dosage Forms, 2 Ed., Vol. 1, pp. 209-214 (1990).
  • the pharmaceutical composition comprises (a) microencapsulated proton pump inhibitor; and (b) at least one antacid; wherein the pharmaceutical composition is made by the process of (a) microencapsulating some or all of the proton pump inhibitor; and (b) dry blending the microencapsulated material with some or all of the at least one antacid.
  • the pharmaceutical composition comprises (a) microencapsulated proton pump inhibitor, and (b) at least one antacid, wherein the microencapsulated proton pump inhibitor is made by the process of spray drying the proton pump inhibitor with a microencapsulating material.
  • the pharmaceutical composition comprises (a) microencapsulated proton pump inhibitor, and (b) at least one antacid, wherein the pharmaceutical composition is made by the process of (a) microencapsulating some or all of the proton pump inhibitor, and (b) blending the microencapsulated material with some or all of the at least one antacid.
  • Initial treatment of a subject suffering from a disease, condition or disorder where treatment with an inhibitor of H + /K + -ATPase is indicated can begin with the dosages indicated above. Treatment is generally continued as necessary over a period of hours, days, or weeks to several months or years until the disease, condition or disorder has been controlled or eliminated. Subjects undergoing treatment with the compositions disclosed herein can be routinely monitored by any of the methods well known in the art to determine the effectiveness of therapy. Continuous analysis of such data permits modification of the treatment regimen during therapy so that optimal effective amounts of compounds of the present invention are administered at any point in time, and so that the duration of treatment can be determined as well.
  • the treatment regimen/dosing schedule can be rationally modified over the course of therapy so that the lowest amount of an inhibitor of H + /K + -ATPase exhibiting satisfactory effectiveness is administered, and so that administration is continued only so long as is necessary to successfully treat the disease, condition or disorder.
  • the pharmaceutical formulations are useful for treating a condition, disease or disorder where treatment with a proton pump inhibitor is indicated.
  • the treatment method comprises oral administration of one or more compositions of the present invention to a subject in need thereof in an amount effective at treating the condition, disease, disorder.
  • the disease, condition or disorder is a gastrointestinal disorder.
  • the dosage regimen to prevent, give relief from, or ameliorate the disease, condition or disorder can be modified in accordance with a variety of factors. These factors include the type, age, weight, sex, diet, and medical condition of the subject and the severity of the disorder or disease. Thus, the dosage regimen actually employed can vary widely and therefore can deviate from the dosage regimens set forth herein.
  • the pharmaceutical formulation is administered post meal.
  • the pharmaceutical formulation administered post meal is in the form of a chewable tablet.
  • the present invention also includes methods of treating, preventing, reversing, halting or slowing the progression of a gastrointestinal disorder once it becomes clinically evident, or treating the symptoms associated with, or related to the gastrointestinal disorder, by administering to the subject a composition of the present invention.
  • the subject may already have a gastrointestinal disorder at the time of administration, or be at risk of developing a gastrointestinal disorder.
  • the symptoms or conditions of a gastrointestinal disorder in a subject can be determined by one skilled in the art and are described in standard textbooks.
  • the method comprises the oral administration a gastrointestinal-disorder-effective amount of one or more compositions of the present invention to a subject in need thereof.
  • Gastrointestinal disorders include, e.g., duodenal ulcer disease, gastrointestinal ulcer disease, gastroesophageal reflux disease, erosive esophagitis, poorly responsive symptomatic gastroesophageal reflux disease, pathological gastrointestinal hypersecretory disease, Zollinger Ellison Syndrome, and acid dyspepsia.
  • the gastrointestinal disorder is heartburn.
  • the present invention is also useful for other subjects including veterinary animals, reptiles, birds, exotic animals and farm animals, including mammals, rodents, and the like.
  • Mammals include primates, e.g., a monkey, or a lemur, horses, dogs, pigs, or cats.
  • Rodents includes rats, mice, squirrels, or guinea pigs.
  • compositions are designed to produce release of the proton pump inhibitor to the site of delivery (typically the stomach), while substantially preventing or inhibiting acid degradation of the proton pump inhibitor.
  • compositions are designed to produce release of the proton pump inhibitor for immediate release in vitro dissolution profile, which is well accepted to by those in the arts to mean drug release of greater than 70% (Q70) in 45 minutes in a USP dissolution test.
  • In vitro dissolution rate contemplated by the present invention include drug release of greater than 70% (Q70) in 15 minutes in a media with a wide pH range of 1-8.
  • compositions can also be used in combination (“combination therapy”) with another pharmaceutical agent that is indicated for treating or preventing a gastrointestinal disorder, such as, e.g., an anti-bacterial agent, an alginate, a prokinetic agent, a H2 antagonist, an antacid, or sucralfate, which are commonly administered to minimize the pain and/or complications related to this disorder.
  • a gastrointestinal disorder such as, e.g., an anti-bacterial agent, an alginate, a prokinetic agent, a H2 antagonist, an antacid, or sucralfate, which are commonly administered to minimize the pain and/or complications related to this disorder.
  • Combination therapies contemplated by the present invention include administration of a pharmaceutical formulation of the present invention in conjunction with another pharmaceutically active agent that is indicated for treating or preventing a gastrointestinal disorder in a subject, as part of a specific treatment regimen intended to provide a beneficial effect from the co-action of these therapeutic agents for the treatment of a gastrointestinal disorder.
  • the beneficial effect of the combination includes, but is not limited to, pharmacokinetic or pharmacodynamic co-action resulting from the combination of therapeutic agents.
  • Administration of these therapeutic agents in combination typically is carried out over a defined time period (usually substantially simultaneously, minutes, hours, days, weeks, months or years depending upon the combination selected).
  • Combination therapies of the present invention are also intended to embrace administration of these therapeutic agents in a sequential manner, that is, where each therapeutic agent is administered at a different time, as Well as administration of these therapeutic agents, or at least two of the therapeutic agents, in a substantially simultaneous manner.
  • Substantially simultaneous administration can be accomplished, e.g., by administering to the subject a single tablet or capsule having a fixed ratio of each therapeutic agent or in multiple, single capsules, or tablets for each of the therapeutic agents.
  • Sequential or substantially simultaneous administration of each therapeutic agent can be effected by any appropriate route.
  • composition of the present invention can be administered orally or nasogastrointestinal, while the other therapeutic agent of the combination can be administered by any appropriate route for that particular agent, including, but not limited to, an oral route, a percutaneous route, an intravenous route, an intramuscular route, or by direct absorption through mucous membrane tissues.
  • the composition of the present invention is administered orally or nasogastrointestinal and the therapeutic agent of the combination may be administered orally, or percutaneously.
  • the sequence in which the therapeutic agents are administered is not narrowly critical.
  • Combination therapy also can embrace the administration of the therapeutic agents as described above in further combination with other biologically active ingredients, such as, but not limited to, a pain reliever, such as a steroidal or nonsteroidal anti-inflammatory drug, or an agent for improving stomach motility, e.g., and with non-drug therapies, such as, but not limited to, surgery.
  • a pain reliever such as a steroidal or nonsteroidal anti-inflammatory drug
  • an agent for improving stomach motility e.g.
  • non-drug therapies such as, but not limited to, surgery.
  • the therapeutic compounds which make up the combination therapy may be a combined dosage form or in separate dosage forms intended for substantially simultaneous administration.
  • the therapeutic compounds that make up the combination therapy may also be administered sequentially, with either therapeutic compound being administered by a regimen calling for two step administration.
  • a regimen may call for sequential administration of the therapeutic compounds with spaced-apart administration of the separate, active agents.
  • the time period between the multiple administration steps may range from, e.g., a few minutes to several hours to days, depending upon the properties of each therapeutic compound such as potency, solubility, bioavailability, plasma half-life and kinetic profile of the therapeutic compound, as well as depending upon the effect of food ingestion and the age and condition of the subject. Circadian variation of the target molecule concentration may also determine the optimal dose interval.
  • the therapeutic compounds of the combined therapies contemplated by the present invention may involve a regimen calling for administration of one therapeutic compound by oral route and another therapeutic compound by an oral route, a percutaneous route, an intravenous route, an intramuscular route, or by direct absorption through mucous membrane tissues, for example.
  • a regimen calling for administration of one therapeutic compound by oral route and another therapeutic compound by an oral route, a percutaneous route, an intravenous route, an intramuscular route, or by direct absorption through mucous membrane tissues for example.
  • the therapeutic compounds of the combined therapy are administered orally, by inhalation spray, rectally, topically, buccally, sublingually, or parenterally (e.g., subcutaneous, intramuscular, intravenous and intradermal injections, or infusion techniques), separately or together, each such therapeutic compound will be contained in a suitable pharmaceutical formulation of pharmaceutically-acceptable excipients, diluents or other formulations components.
  • the pharmaceutical formulations of the present invention are administered with low strength enteric coated Aspirin.
  • the second active pharmaceutical e.g., Aspirin or an NSAID, used in combination with the pharmaceutical formulations of the present invention, is enteric coated.
  • antacid present in the pharmaceutical formulations of the present invention increase the pH level of the gastrointestinal fluid, thereby allowing part or all of the enteric coating on the second active pharmaceutical to dissolve in the stomach.
  • the basic operation for the spinning disk process used was as follows: An encapsulation solution or suspension was prepared by dissolving the encapsulation (or shell) material in the appropriate solvent. Omeprazole (or the proton pump inhibitor) was dispersed in the coating solution and fed onto the center of the spinning disk or spray dryer which is pre-heated. One of three atomization methods can be used to make droplets of the feed solution/suspension. The microspheres were formed by removal of the solvent using heated airflow inside the drying chamber and collected as a powder using a cyclone separator.
  • Atomization of the feed solution/suspension can be achieved by major methods in a spray dryer, by rotary atomization using a spinning disc, by high pressure-nozzle, or by two fluid nozzles.
  • a thin film was produced across the surface of the disk and atomization occurs as the coating material left the periphery of the disk in a rotary atomization process.
  • the microspheres were formed by removal of the solvent using heated airflow inside the atomization chamber and collected as a free-flowing powder using a cyclone separator.
  • High pressure nozzle utilizes a high pressure pump to feed the solution/suspension through a various size nozzle so droplets form when injected into the drying chamber.
  • the pressure energy is converted to kinetic energy and the feed issues form the orifice as a high speed film that readily disintegrates into droplets.
  • the atomization is created due to high frictional shearing forces between the liquid surface and the air having a high velocity even at sonic velocities and sometimes rotated to obtain maximum atomization.
  • the basic operation for the microencapsulation by spray drying was as follows: An encapsulation solution or suspension was prepared by dissolving the shell material in the appropriate solvent. Proton pump inhibitor was dispersed in the coating solution and fed onto the spray dryer which is pre-heated. One of the three atomization method can be used to make droplet of the feed solution/suspension. The microspheres were formed by removal of the solvent using heated airflow inside the drying chamber and collected as a powder using a cyclone separator.
  • a spray dryer with attached fluid-bed dryer for sizing of dried particles and/or agglomeration if desired can be also used. Recycling of the super-fine particles from the cyclones back to the spray dryer inlet would allow the agglomeration to form desired particle size distribution.
  • the dissolution profiles of the microencapsulated omeprazole were determined by a method similar to the HPLC method outlined in Example 10, described below.
  • the size of the microspheres was determined by using a microscopic optical method similar to the one outlined in Example 11.
  • a suspension containing Klucel EF, sodium bicarbonate and omeprazole was prepared (total solid content of 16.23%) and spray dried using a rotary atomizer.
  • the pH of the suspension was 8.1.
  • Spray rate was 35 Kg/hour and the resulting outlet temperature was 70-85° C.
  • Atomizer speed was 22,000 rpm.
  • the viscosity of the suspension was 680 cps and the pumping system had no difficulty in delivering the suspension to the atomizer.
  • White, fine particles were collected.
  • the median particle size of sample was approximately 80-110 ⁇ m.
  • USP No. 2 in vitro dissolution test showed drug release of >90% in 15 minutes. The amounts of each component are shown below:
  • a suspension containing Klucel EF, sodium bicarbonate and omeprazole was prepared (total solid content of 16.23%) and spray dried using a rotary atomizer.
  • the pH of the suspension was 8.1.
  • the inlet temperature was 130° C. and the outlet temperature was 84-85° C.
  • Atomizer speed was 27,000 rpm.
  • the viscosity of the suspension was 680 cps and the pumping system had no difficulty in delivering the suspension to the atomizer.
  • White, fine particles were collected.
  • the median particle size of sample was approximately 35-40 ⁇ m USP No. 2 in vitro dissolution test showed drug release of >35% in 30 minutes.
  • the amounts of each component are shown below:
  • omeprazole The microencapsulation of omeprazole consisted of two main steps: 1) suspension preparation and 2) spray drying operation. Methocel E5 was weighed out and added into water extremely slowly until a clear, homogeneous. PEG400 was added to the Methocel E5 polymer solution. Omeprazole and sodium bicarbonate were added to the polymer solution with agitation until a milky white, homogeneous suspension was formed. The finished omeprazole suspension was agitated at a minimum speed to prevent settling. The omeprazole suspension was then sprayed using the parameter shown below.
  • a suspension containing hydroxypropyl cellulose, sodium bicarbonate and omeprazole was prepared (total solid content of 15.0%) and spray dried using a high pressure nozzle.
  • the pH of the suspension was 8.5.
  • the inlet temperature was 140-150° C. and the outlet temperature was 79-85° C.
  • Core and the Nozzle size (SK value) used in the spray drying was 72 and 17, respectively.
  • the viscosity of the suspension was in the range of 600-700 cps and the pumping system had no difficulty in delivering the suspension to the atomizer.
  • White, fine particles were collected.
  • the median particle size of sample was approximately 60 ⁇ m.
  • the amounts of each component are shown below:
  • the chart below summarizes the wt %, the feed rates used, and the inlet/outlet temperatures for eleven different omeprazole microspheres.
  • the amount of encapsulated omeprazole used in each tablet batch varies based on the actual payload of each set of microcapsules to achieve the theoretical dose of 40 mg.
  • the omeprazole was microencapsulated in a similar manner as that described in Example 1. All ingredients are mixed well to achieve a homogeneous blend.
  • Tablets containing omeprazole microspheres were prepared using a high-speed rotary tablet press (TBCB Pharmaceutical Equipment Group, Model ZPY15). Round, convex tablets with diameters of about 10 mm and an average weight of approximately 600 mg per tablet were prepared.
  • Chewable tablets were manufactured using the following materials: Encapsulated omeprazole (varied based on payload, to deliver 40 mg potency), sodium bicarbonate (1260 mg), calcium carbonate (790 mg), croscarmellose sodium (64 mg), Klucel (160 mg), Xylitab 100 (380 mg), microcrystalline cellulose (128 mg), sucralose (162 mg), peppermint flavor (34 mg), peach flavor (100 mg), masking powder (60 mg), FD&C Lake No. 40 Red (3 mg), and magnesium stearate (32 mg).
  • Table 2B summarizes various drug wt %, shell materials, and in-vitro dissolution data for eighteen different proton pump inhibitor microspheres.
  • the atomization method for the spray drying is also listed and is performed in a manner analogous to that listed in Example 1 above.
  • the amount of microencapsulated lansoprazole used in each tablet batch is based on the actual payload of microcapsules to achieve the theoretical dose of 30 mg lansoprazole per tablet.
  • Lansoprazole is microencapsulated in a similar manner as that described in Example 1 and 2. All ingredients are mixed well to achieve a homogenous bulk blend which is then compressed to round tablets.
  • Omeprazole magnesium tablets are manufactured using the following materials: Microencapsulated Omeprazole-Mg salt (45% drug loading-Entry 2B-14 in Table 2B, 89 mg to deliver 40 mg omeprazole), sodium bicarbonate (420 mg), direct-compression grade magnesium hydroxide (contains 5% starch, 337 mg to deliver 320 mg magnesium hydroxide), croscarmellose sodium (50 mg), Klucel-EXF (50 mg), and magnesium stearate (7 mg) [total 16 mEq in swallowable tablet or caplet].
  • omeprazole magnesium salt is microencapsulated in a similar manner as that described in Example 1 and 2. All ingredients are mixed well to achieve a homogenous bulk blend which is then compressed to capsule shaped tablets.
  • pantoprazole used in each tablet batch is based on the actual payload of microcapsules to achieve the theoretical dose of 40 mg pentoprazole per tablet.
  • Pantoprazole is microencapsulated in a similar manner as that described in Example 1 and 2. All ingredients are mixed well to achieve a homogenous bulk blend which is then compressed to round tablets.
  • Rabeprazole sodium caplets (capsule-shaped tablets) are manufactured using the following materials: Microencapsulated Rabeprazole sodium salt (40% drug loading-Entry 2B-10 Table 2B, 106 mg to deliver 40 mg rabeprazole), sodium bicarbonate (336 mg), Direct-compression grade magnesium hydroxide (contains 5% starch, 307 mg to deliver 292 mg magnesium hydroxide), magnesium oxide (101 mg), croscarmellose sodium (50 mg), Klucel-EXF (40 mg), and magnesium stearate (7 mg) [total 14mEq in swallowable tablet or caplet].
  • Microencapsulated Rabeprazole sodium salt 40% drug loading-Entry 2B-10 Table 2B, 106 mg to deliver 40 mg rabeprazole
  • sodium bicarbonate (336 mg)
  • Direct-compression grade magnesium hydroxide contains 5% starch, 307 mg to deliver 292 mg magnesium hydroxide
  • magnesium oxide 101 mg
  • croscarmellose sodium 50 mg
  • rabeprazole sodium salt is microencapsulated in a similar manner as that described in Example 1 and 2. All ingredients are mixed well to achieve a homogenous bulk blend which is then compressed to tablets.
  • omeprazole used in each capsule batch varies based on the actual payload of each set of microcapsules to achieve the theoretical dose of 30 mg.
  • Lansoprazole was microencapsulated in a similar manner as that described in Example 1 and 2. All ingredients are mixed well to achieve a homogenous bulk blend which is then filled into a capsule such as a size (00) hard gelatin capsule.
  • omeprazole magnesium salt used in each capsule batch varies based on the actual payload of each set of microcapsules to achieve the theoretical dose of 30 mg lansoprazole per capsule.
  • Omeprazole magnesium salt was microencapsulated in a similar manner as that described in Example 1 and 2. All ingredients are mixed well to achieve a homogenous bulk blend which is then filled into a capsule such as a size zero (0) hard gelatin capsule.
  • omeprazole magnesium salt was microencapsulated in a similar manner as that described in Example 1 and 2. All ingredients are mixed well to achieve a homogenous bulk blend which is then filled into a capsule such as a size zero (0) hard gelatin capsule.
  • pentoprazole sodium salt used in each capsule batch varies based on the actual payload of each set of microcapsules to achieve the theoretical dose of 40 mg neutral pentoprazole per capsule.
  • Pentoprazole sodium salt was microencapsulated in a similar manner as that described in Example 1 and 2. All ingredients are mixed well to achieve a homogenous bulk blend which is then filled into a capsule such as a size one hard gelatin capsule.
  • Various chewable tablets are manufactured using the microencapsulate materials described in Examples 1 and 2.
  • the amount of microencapsulated omeprazole used in each chewable tablet batch is based on the actual payload of each set of microcapsules to achieve the theoretical dose. All ingredients are mixed well to achieve a homogenous bulk blend which is then compressed to chewable tablets.
  • PPI Buffering Agent Sleep Aid 20 mg per chewable tablet 20.6 mEq or 600 mg Mg(OH) 2 0.125 triazolam mg per 170 mg Xylitab (microencapsulated) 5.0 mEq or 420 mg NaHCO 3 tablet 30 mg Ac-Di-Sol 25.6 mEq or 1020 mg total buffer 100 mg Klucel 25 mg cherry flavor 15 mg magnesium stearate 3 mg Red #40 Lake
  • PPI Buffering Agent Sleep Aid Excipient 30 mg lansoprazole 24.0 mEq or 700 mg Mg(OH) 2 25 mg diphenhydramine per 170 mg Xylitab per chewable tablet 5 mEq or 420 mg NaHCO 3 tablet 30 mg Ac-Di-Sol (microencapsulated) 29.0 mEq or 1120 mg total buffer 100 mg Klucel 25 mg cherry flavor 15 mg magnesium stearate 3 mg Red #40 Lake
  • PPI Buffering Agent Prokinetic Agent 20 mg 20.6 mEq or 600 mg Mg(OH) 2 5 mg Mosapride per tablet 170 mg Xylitab microencapsulated 5.0 mEq or 420 mg NaHCO 3 30 mg Ac-Di-Sol omeprazole per 25.6 mEq or 1020 mg total buffer 100 mg Klucel chewable tablet 40 mg Sucralose 25 mg cherry flavor 15 mg magnesium stearate 3 mg Red #40 Lake
  • PPI Buffering Agent Prokinetic Agent 40 mg 24.0 mEq or 700 mg Mg(OH) 2 10 mg Domperidone per 170 mg Dipac sugar microencapsulated 7.1 mEq or 600 mg NaHCO 3 tablet 30 mg Ac-Di-Sol omeprazole per 31.1 mEq or 1300 mg total buffer 120 mg Klucel chewable tablet 27 mg grape flavor 15 mg magnesium stearate 1 mg Red #40 Lake 1 mg Blue #2 Lake
  • PPI Buffering Agent Prokinetic Agent 30 mg 24.0 mEq or 700 mg Mg(OH) 2 20 mg Clebopride per 170 mg Xylitab microencapsulated 5.0 mEq or 420 mg NaHCO 3 tablet 30 mg Ac-Di-Sol lansoprazole per 29.0 mEq or 1120 mg total buffer 100 mg Klucel chewable tablet 25 mg cherry flavor 15 mg magnesium stearate 3 mg Red #40 Lake
  • PPI Buffering Agent Prokinetic Agent Excipient 60 mg 15 mEq or 750 mg Ca(OH) 2 10 mg Norcisapride per 170 mg Xylitab microencapsulated 15 mEq or 1260 mg NaHCO 3 tablet 30 mg Ac-Di-Sol omeprazole per 30 mEq or 2010 mg total buffer 100 mg Klucel chewable tablet 25 mg cherry flavor 15 mg magnesium stearate 3 mg Red #40 Lake
  • PPI Buffering Agent Prokinetic Agent Excipient 40 mg 15 mEq or 750 mg Ca(OH) 2 10 mg Cisapride per tablet 170 mg Xylitab microencapsulated 10 mEq or 840 mg NaHCO 3 30 mg Ac-Di-Sol omprazole per chewable 25 mEq or 1590 mg total buffer 100 mg Klucel tablet 15 mg mint flavor 15 mg magnesium stearate
  • PPI Buffering Agent Prokinetic Agent 40 mg 24.0 mEq or 700 mg Mg(OH) 2 10 mg Cisapride per tablet 60 mg sucralose microencapsulated 7.1 mEq or 600 mg NaHCO 3 60 mg Ac-Di-Sol omeprazole per bite- 31.1 mEq or 1300 mg total buffer 60 mg pregelatinized starch disintegration chewable 30 mg Klucel tablet 27 mg grape flavor 15 mg magnesium stearate 1 mg Red #40 Lake 1 mg Blue #2 Lake
  • PPI Buffering Agent Prokinetic Agent Excipient 30 mg 17.1 mEq or 500 mg Mg(OH) 2 2.5 mg Mosapride per 60 mg sucralose microencapsulated 5 mEq or 420 mg NaHCO 3 tablet 60 mg Ac-Di-Sol lansoprazole per bite- 22.1 mEq or 920 mg total buffer 70 mg pregelatinized starch disintegration chewable 30 mg Klucel tablet 25 mg cherry flavor 15 mg magnesium stearate 3 mg Red #40 Lake
  • Prokinetic PPI Buffering Agent Agent Excipient 60 mg 15 mEq or 750 mg 10 mg 60 mg sucralose micro- Ca(OH) 2 Domperidone 60 mg Ac-Di-Sol encapsulated 15 mEq or 1260 mg per tablet 60 mg pregela- omeprazole NaHCO 3 tinized starch per bite- 30 mEq or 2010 mg 30 mg Klucel disinte- total buffer 25 mg cherry flavor gration 15 mg magnesium chewable stearate tablet 3 mg Red #40 Lake
  • Prokinetic PPI Buffering Agent Agent Excipient 40 mg 15 mEq or 750 mg 10 mg 60 mg sucralose micro- Ca(OH) 2 Norcisapride 60 mg Ac-Di-Sol encapsulated 10 mEq or 840 mg 60 mg pregela- omprazole NaHCO 3 tinized starch per bite- 25 mEq or 1590 mg 30 mg Klucel disinte- total buffer 15 mg mint flavor gration 15 mg magnesium chewable stearate tablet
  • PPI Antacid Excipient 10 mg 3.4 mEq or 100 mg 15 mg Ac-Di-Sol micro- Mg(OH) 2 40 mg Klucel encapsulated 3.0 mEq or 250 mg 6 mg magnesium omeprazole NaHCO 3 stearate per c chewable 6.4 mEq or 350 mg tablet total antacid
  • PPI Antacid Excipient 60 mg 4.4 mEq or 220 mg 80 mg Xylitab micro- Ca(OH) 2 30 mg Ac-Di-Sol encapsulated 3.6 mEq or 300 mg 100 mg Klucel omeprazole NaHCO 3 35 mg Sucralose per chewable 8.0 mEq or 520 mg 10 mg cherry flavor tablet total antacid 9 mg magnesium stearate 2 mg Red #40 Lake
  • caplets are manufactured using the microencapsulate materials described in Examples 1 and 2.
  • the amount of microencapsulated omeprazole used in each caplet batch is based on the actual payload of each set of microcapsules to achieve the theoretical dose. All ingredients are mixed well to achieve a homogenous bulk blend which is then compressed to caplets.
  • Prokinetic PPI Buffering Agent Agent 40 mg 17.1 mEq or 500 mg 20 mg 20 mg Ac-Di-Sol micro- Mg(OH) 2 Clebopride 80 mg Klucel encapsulated 3.0 mEq or 250 mg per tablet 10 mg magnesium omeprazole NaHCO 3 stearate per caplet 20.1 mEq or 750 mg total buffer
  • Prokinetic PPI Buffering Agent Agent Excipient 15 mg 17.1 mEq or 500 mg 10 mg 20 mg Ac-Di-Sol micro- Mg(OH) 2 Clebopride 80 mg Klucel encapsulated 3.0 mEq or 250 mg per tablet 10 mg magnesium lansoprazole NaHCO 3 stearate per caplet 20.1 mEq or 750 mg total buffer
  • Prokinetic PPI Buffering Agent Agent 10 mg 13.7 mEq or 400 mg 10 mg 20 mg Ac-Di-Sol micro- Mg(OH) 2 Cisapride 80 mg Klucel encapsulated 3.0 mEq or 250 mg pre tablet 10 mg magnesium omeprazole NaHCO 3 stearate per caplet 16.7 mEq or 650 mg total buffer
  • Prokinetic PPI Buffering Agent Agent 40 mg 20.6 mEq or 600 mg 10 mg 20 mg Ac-Di-Sol micro- Mg(OH) 2 Metoclo- 80 mg Klucel encapsulated 3.0 mEq or 250 mg pramide 10 mg magnesium omeprazole NaHCO 3 per tablet stearate per caplet 23.6 mEq or 850 mg total buffer
  • PPI Buffering Agent NSAID 40 mg 17.1 mEq or 500 mg 81 mg 20 mg Ac-Di-Sol micro- Mg(OH) 2 aspirin 80 mg Klucel encapsulated 3.0 mEq or 250 mg per tablet 10 mg magnesium omeprazole NaHCO 3 stearate per caplet 20.1 mEq or 750 mg total buffer
  • PPI Buffering Agent NSAID Excipient 15 mg 17.1 mEq or 500 mg 75 mg 20 mg Ac-Di-Sol micro- Mg(OH) 2 indo- 80 mg Klucel encapsulated 3.0 mEq or 250 mg methacin 10 mg magnesium lansoprazole NaHCO 3 per tablet stearate per caplet 20.1 mEq or 750 mg total buffer
  • PPI Buffering Agent NSAID 10 mg 13.7 mEq or 400 mg 200 mgs 20 mg Ac-Di-Sol micro- Mg(OH) 2 Ibupro- 80 mg Klucel encapsulated 3.0 mEq or 250 mg pene 10 mg magnesium omeprazole NaHCO 3 per tablet stearate per caplet 16.7 mEq or 650 mg total buffer
  • PPI Buffering Agent NSAID 40 mg 20.6 mEq or 600 mg 100 mgs 20 mg Ac-Di-Sol micro- Mg(OH) 2 aspirin 80 mg Klucel encapsulated 3.0 mEq or 250 mg per tablet 10 mg magnesium omeprazole NaHCO 3 stearate per caplet 23.6 mEq or 850 mg total buffer
  • PPI Buffering Agent NSAID 20 mg 20.6 mEq or 600 mg 25 mg 170 mg Xylitab micro- Mg(OH) 2 rofecoxib 30 mg Ac-Di-Sol encapsulated 5.0 mEq or 420 mg per tablet 100 mg Klucel omeprazole NaHCO 3 40 mg Sucralose per caplet 25.6 mEq or 1020 mg 25 mg cherry flavor total buffer 15 mg magnesium stearate 3 mg Red #40 Lake
  • PPI Buffering Agent NSAID 40 mg 24.0 mEq or 700 mg 100 mg 170 mg Dipac sugar micro- Mg(OH) 2 diclofenac 30 mg Ac-Di-Sol encapsulated 7.1 mEq or 600 mg per tablet 120 mg Klucel omeprazole NaHCO 3 27 mg grape flavor per caplet 31.1 mEq or 1300 mg 15 mg magnesium total buffer stearate 1 mg Red #40 Lake 1 mg Blue #2 Lake
  • PPI Buffering Agent NSAID 30 mg 24.0 mEq or 700 mg 400 mg 170 mg Xylitab micro- Mg(OH) 2 aspirin 30 mg Ac-Di-Sol encapsulated 5.0 mEq or 420 mg and 400 mg 100 mg Klucel lansoprazole NaHCO 3 enteric 25 mg cherry flavor per caplet 29.0 mEq or 1120 mg coated 15 mg magnesium total buffer aspirin stearate per tablet 3 mg Red #40 Lake
  • PPI Buffering Agent NSAID Excipient 60 mg 15 mEq or 750 mg 600 mg 170 mg Xylitab micro- Ca(OH) 2 oxaprozin 30 mg Ac-Di-Sol encapsulated 15 mEq or 1260 mg per tablet 100 mg Klucel omeprazole NaHCO 3 25 mg cherry flavor per caplet 30 mEq or 2010 mg 15 mg magnesium total buffer stearate 3 mg Red #40 Lake
  • PPI Buffering Agent NSAID 40 mg 15 mEq or 750 mg 100 mg 170 mg Xylitab micro- Ca(OH) 2 aspirin 30 mg Ac-Di-Sol encapsulated 10 mEq or 840 mg per tablet 100 mg Klucel omprazole NaHCO 3 15 mg mint flavor per caplet 25 mEq or 1590 mg 15 mg magnesium total buffer stearate
  • PPI Buffering Agent NSAID Excipient 40 mg 24.0 mEq or 700 mg 100 mg 60 mg sucralose micro- Mg(OH) 2 diclofenac 60 mg Ac-Di-Sol encapsulated 7.1 mEq or 600 mg per tablet 60 mg pregela- omeprazole NaHCO 3 tinized starch per caplet 31.1 mEq or 1300 mg 30 mg Klucel total buffer 27 mg grape flavor 15 mg magnesium stearate 1 mg Red #40 Lake 1 mg Blue #2 Lake
  • PPI Buffering Agent NSAID 30 mg 17.1 mEq or 500 mg 200 mg 60 mg sucralose micro- Mg(OH) 2 micro- 60 mg Ac-Di-Sol encapsulated 5 mEq or 420 mg encapsulated 70 mg pregela- lansoprazole NaHCO 3 asprin tinized starch per caplet 22.1 mEq or 920 mg per tablet 30 mg Klucel total buffer 25 mg cherry flavor 15 mg magnesium stearate 3 mg Red #40 Lake
  • PPI Buffering Agent NSAID Excipient 60 mg 15 mEq or 750 mg 100 mg 60 mg sucralose micro- Ca(OH) 2 ketoprofen 60 mg Ac-Di-Sol encapsulated 15 mEq or 1260 mg per tablet 60 mg pregela- omeprazole NaHCO 3 tinized starch per caplet 30 mEq or 2010 mg 30 mg Klucel total buffer 25 mg cherry flavor 15 mg magnesium stearate 3 mg Red #40 Lake
  • PPI Buffering Agent NSAID Excipient 40 mg 15 mEq or 750 mg 100 mg 60 mg sucralose micro- Ca(OH) 2 aspirin 60 mg Ac-Di-Sol encapsulated 10 mEq or 840 mg per tablet 60 mg pregela- omprazole NaHCO 3 tinized starch per tablet 25 mEq or 1590 mg 30 mg Klucel total buffer 15 mg mint flavor 15 mg magnesium stearate
  • microencapsulate materials described in Examples 1 and 2.
  • the amount of microencapsulated omeprazole used in each capsule batch is based on the actual payload of each set of microcapsules to achieve the theoretical dose. All ingredients are mixed well to achieve a homogenous bulk blend which is then filled into a hard gelatin capsule such as a size 00 hard gelatin capsule from Capsugel.
  • Prokinetic PPI Buffering Agent Agent Excipient 15 mg 17.1 mEq or 500 mg 5 mg 30 mg Ac-Di-Sol micro- Mg(OH) 2 Mosapride 15 mg Klucel encapsulated 3.0 mEq or 250 mg per capsule 7 mg magnesium lansoprazole NaHCO 3 stearate per capsule 20.1 mEq or 750 mg total buffer
  • Prokinetic PPI Buffering Agent Agent 40 mg 15.4 mEq or 450 mg 10 mg 30 mg Ac-Di-Sol micro- Mg(OH) 2 Norcisapride 7 mg magnesium encapsulated 2.4 mEq or 200 mg per capsule stearate omeprazole NaHCO 3 per capsule 17.8 mEq or 650 mg total buffer
  • PPI Buffering Agent NSAID Excipient 15 mg 17.1 mEq or 500 mg 75 mg 30 mg Ac-Di-Sol micro- Mg(OH) 2 ketoprofen 15 mg Klucel encapsulated 3.0 mEq or 250 mg per capsule 7 mg magnesium lansoprazole NaHCO 3 stearate per capsule 20.1 mEq or 750 mg total buffer
  • PPI Buffering Agent NSAID 40 mg 15.4 mEq or 450 mg 100 mg enteric 30 mg Ac-Di-Sol micro- Mg(OH) 2 coated aspirin 7 mg magnesium encapsulated 2.4 mEq or 200 mg per capsule stearate omeprazole NaHCO 3 per capsule 17.8 mEq or 650 mg total buffer
  • the following procedure was used to determine the potency of omeprazole in the tablets.
  • the tablet was accurately weighed and placed into 100 ml volumetric flask.
  • 1.0 ml of Nanopure water was added to wet and soften the tablet.
  • the solution was allowed to stand for 30 minutes. After sitting, the sample was vortexed and sonicated for 30 minutes or until completely dissolved.
  • 1.0 ml of chloroform was then added and the sample was vortexed and sonicated for an additional 15 minutes.
  • the solution was then brought to volume with methanol and vortexed again to mix solution. 10 ml was then decanted into a 10 cc syringe fitted with a 0.45-micron filter.
  • a 5-point calibration curve was prepared in methanol ranging from 15 to 300 ⁇ g/ml.
  • Microspheres that exhibited dissolution results with greater than 80% omeprazole release after 2 hours were placed on stability.
  • the microspheres were stored in opened vials at 25° C. All samples showed degradation after 4 weeks at elevated temperatures.
  • the open vials stored at 25° C. were analyzed after 6-8 weeks for potency and for impurities using the Omeprazole EP method.
  • the stability results are summarized in the table 7A below.
  • HPLC samples for the omeprazole assay of various microspheres were prepared as follows: 5 mg of the microsphere were accurately weighed into a screw cap culture tube. To that, 200 ⁇ L of chloroform was added. The microspheres were allowed to dissolve, sonicated and vortex for approximately one minute. Then, 10 ml of methanol was added and the sample was again vortexed for one minute. Once completed, an aliquot of the sample was removed for HPLC analysis.
  • a 5-point calibration curve was prepared in methanol ranging from 20 to 500 ⁇ g/mL to calculate payload.
  • the chromatographic conditions were: Mobile phase: 75.5% Na 2 PO 4 pH 8.0, 24.5% Acetonitrile; Flow Rate: 1.0 mL/min; Run Time: 15 min; Injection Volume: 20 ⁇ L; Detector: U.V., 280 nm; Column: Waters SymmetryShield RP8.
  • HPLC samples for the omeprazole assay of various microspheres were prepared in the following manner: 5 mgs of the omeprazole microspheres were weighed into a screw cap culture tube. To that, 200 ⁇ L of chloroform were added. The microspheres were allowed to dissolve, sonicate and vortex for approximately one minute each. 10 mL of methanol was then added and the sample was again vortexed for 1 minute. Once complete, an aliquot was removed for HPLC analysis.
  • omeprazole concentration 100 ⁇ g/mL concentration of omeprazole in methanol for a marker was prepared. A 0.1 ⁇ g/mL concentration of omeprazole was then prepared to set one-half the minimal detection limit. Then, a 1 ⁇ g/mL concentration of omeprazole impurity D in methanol was prepared.
  • the chromatographic conditions were: Mobile Phase: 75% Na 2 PO 4 pH 7.6, 25% acetonitrile; Flow Rate: 1.0 mL/min; Run Time: 30 min; Injection Volume: 20 ⁇ L; Detector: U.V., 280 nm; Column: Waters SymmetryShield RP8.
  • the omeprazole potency method was used for the dissolution testing.
  • the HPLC samples for the omeprazole assay of various microspheres were prepared according to the following method. 5 mgs of the microspheres were accurately weighed into an 8 ounce amber bottle. To that, 100 ml of pH 7.4 monobasic phosphate buffer was added. The samples were placed in a 37° C. water bath and vigorously shaken until the end of the release study. Using an Eppendorf pipette, 100 ⁇ L was removed and the outside part of the tip was rinsed with 100 ⁇ L of buffer back into the sample bottle. The sample was then transferred into a limited insert for HPLC analysis using a 1 cc syringe fitted with a 45 micron filter. Samples were then taken at 30, 45, and 120 minutes.
  • a 6-point calibration curve was prepared in diluent (70% sodium phosphate pH 10.0/30% acetonitrile) ranging from 1 to 120 ⁇ g/mL to determine sample release rates.
  • the chromatographic conditions were: Mobile phase: 75.5% Na 2 PO 4 pH 8.0, 24.5% Acetonitrile; Flow Rate: 1.0 mL/min; Run Time: 15 min; Injection Volume: 20 ⁇ L; Detector: U.V., 280 nm; Column: Waters SymmetryShield RP8.
  • omeprazole microspheres were observed using an Olympus BX60 optical microscope equipped with an Olympus DP 10 digital camera to determine their particle size and morphology characteristics. The microspheres were observed at either 100 ⁇ or 200 ⁇ magnification.
  • microspheres prepared by spray drying were in the size range of 5 to 30 microns.
  • the microspheres prepared by spinning disk-solvent process were in the size range of 25 to 100 microns.
  • the microspheres prepared by spinning disk-hot melt process were in the size range of 30 to 125 microns.
  • the percent weight loss up to 140° C. was recorded to determine the amount of volatiles present. Most samples exhibit a weight loss of less than 1% up to 140° C. except the samples that contained sodium bicarbonate which have a greater weight loss, from 7-32%.
  • the following TGA run conditions were used: nitrogen atmosphere; Isothermal for 5 minutes at 25° C.; ramp 10° C./minute to 250° C.; platinum sample pan.
  • Antacid tablet without any omeprazole was prepared using the components listed in the table below. All ingredients were blended homogeneously in 1 cubic feet V-blender. The resulting blend was compressed into a tablet using a Stokes 16 station rotary tablet press equipped with plain, round 3 ⁇ 4′′, FFBE punches.
  • Micronized, non-microencapsulated omeprazole chewable tablets were prepared using the components listed in the table. All ingredients were blended homogeneously in 1 cubic feet V-blender. The resulting blend was compressed into a tablet using a Stokes 16 station rotary tablet press equipped with plain, round 3 ⁇ 4′′, FFBE punches.
  • Microencapsulated omeprazole powder (40 mg) prepared in Example 1C was co-administered with the antacid chewable tablet prepared in Example 13A (total ANC of 30.7 mEq).
  • the powder and the placebo chewable tablet were chewed together in the mouth to simulate administration of 40 mg of microencapsulated omeprazole incorporated in the said placebo tablet.
  • Microencapsulated omeprazole powder (40 mg) prepared in Example 1D was co-administered with an antacid chewable tablet prepared in Example 13A (total ANC of 30.7 mEq).
  • the powder and the placebo chewable tablet were chewed together in the mouth to simulate administration of 40 mg of microencapsulated omeprazole incorporated in the said placebo tablet.
  • Microencapsulated omeprazole powder prepared in Example 1C was blended homogeneously with other ingredients shown in the table. The powder blend was then compressed into a capsule shaped tablet using a Stokes rotary press.
  • SAN-20D Ingredients mg/cap % Microencapsulated Omeprazole (28% w/w) 142.86 14.1% Sodium Bicarbonate #2 280 27.7% Magnesium Hydroxide (95%) 473.68 46.9% Croscarmellose Sodium 50 4.9% Hydroxypropyl Cellulose 55 5.4% Magnesium Stearate 9 0.9% Total Fill Wt: 1010.5 100%
  • Microencapsulated omeprazole powder prepared in Example 1C was blended homogeneously with other ingredients shown in the table. The powder blend was then compressed into a capsule shaped tablet using a Stokes rotary press.
  • SAN-20E Ingredients mg/cap % Microencapsulated Omeprazole (28% w/w) 142.86 13.7% Sodium Bicarbonate #2 280 26.9% Magnesium Hydroxide (95%) 473.68 45.4% Croscarmellose Sodium 82 7.9% Hydroxypropyl Cellulose 55 5.3% Magnesium Stearate 9 0.9% Total Fill Wt: 1042.5 100%
  • This trial was conducted as an open-label, single-dose, crossover trial, with each subject receiving up to twelve different oral omeprazole formulations, one in each of the twelve treatment periods. Each dose was followed by a minimum 7-day washout. Omeprazole was administered at a dose of 40 mg.
  • the compositions administered are set forth in Examples 13A-13F and Table 13.G.1. All formulations were administered with about 120 ml (4 oz) of water after an overnight fast and 1 hour prior to a standardized, high-fat breakfast. Within a given treatment period, the same treatment was administered to all subjects.
  • the non-enteric coated formulation study drugs were compared to Prilosec® which contained enteric-coated granules.
  • This trial was designed to assess the pharmacokinetics of immediate-release omeprazole chewable tablets and caplets versus the Prilosec® 40 mg delayed-release formulation.
  • the duration of the trial for each subject was approximately 24 weeks, including up to 14 days for screening and a minimum 7 day wash-out period between omeprazole doses.
  • Data from 12 healthy male subjects were expected to provide adequate power to assess pharmacokinetics and safety using descriptive statistics.
  • the descriptive statistics were assessed using the pharmacokinetic parameters: Tmax, Cmax, AUC(0-t), and AUC(0-inf).
  • the treatments administered to subjects in this trial are listed in Table 13.G.1, above.
  • the treatment protocol entailed a 14 day assessment period, followed by a first period (Period 1) in which Prilosec® 40 mg delayed release capsule was administered to the subjects, after an overnight fast, and 1 hour prior to a standardized high-fat breakfast.
  • Plasma sampling was conducted for 6 hour post-dose.
  • Period 1 was followed by a 7-14 day washout period, during which the plasma levels of omeprazole were expected to decrease to a steady baseline.
  • the other periods listed in Tablet 13.G.1 were conducted in a similar manner to Period 1, except substituting a dosage form according to the invention for the delayed-release formulation used in Period 1.
  • Blood samples (3 mL) were obtained by venipuncture within 30 minutes before each dose and at 0, 5, 10, 15, 20, 30, 45, 60, 90, 120, 180, 240, 300, 360 minutes (6 hours) after delivery of each dose during each trial period.
  • Zero time was the time that the subject swallowed a capsule, caplet or chewable tablet of trial drug.
  • Plasma omeprazole concentrations were measured using a previously validated liquid chromatography mass spectrometry (LC-MSIMS) assay (MDS Pharma Services, Lincoln, Nebr.). The linear assay range was 5.0 to 750 ng/mL. The following pharmacokinetic parameters were measured for each subject:
  • an analysis of variance (ANOVA) model was used to test the bioequivalence of each of the tested drug formulations.
  • the model included the following factors: treatment, period, sequence, and subject nested within sequence.
  • CIs percent confidence intervals
  • equivalence was declared for each parameter if the bounds of the 90% CIs for the percent mean ratio, comparing a composition according to the invention (Periods 2-12) with Prilosec, were between 80% and 125%.
  • Omeprazole was microencapsulated as described in Example 1B.
  • omeprazole pre-blend containing microencapsulated omeprazole, antacid excipients and other formulation components was prepared.
  • a flavor pre-blend containing sensory components was then prepared.
  • the main blend was then prepared by combining the omeprazole and flavor pre-blends. Magnesium stearate was then added to the main blend and mixed to form a final blend. All blending operations were carried out in appropriately sized V-blenders. Blend uniformity was ensured by testing at various stages of blending.
  • the final blend was then compressed on a high speed rotary tablet press to form the final tablets.
  • the tablet press was a Stokes® instrumented tablet press using less than a full set of tablet tooling in order to conserve powder.
  • a compression force of 50 kN using 3 ⁇ 4′′ round FFBE tooling gave an acceptable tablet harness and friability in all prototype batches. The amount of each component is listed below in Table 14.A.2.
  • the primary objective was to show that SAN-15 20 mg Chewable Tablets are pharmacokinetically bioequivalent to Prilosec 20 mg with respect to area under the curve.
  • Subjects who had received SAN-15 chewable tablets 20 mg in Period 1 were given an eighth dose on Day 8 in Period 1, 1 hour after the start of the standardized high-fat breakfast. Blood samples were collected for 12 hours after the eighth dose. After a 10- to 14-day washout period, subjects returned for Period 2 and received the alternate treatment from that received in Period 1. Procedures in Period 2 were identical to those in Period 1 except that no eighth dose of SAN-15 chewable tablets 20 mg was given.
  • SAN-15 SAN-15 chewable tablets (omeprazole immediate-release chewable tablets) 20 mg to be administered orally and chewed for 30 seconds, followed by drinking 120 mL water each morning after an overnight fast, 1 hour before starting breakfast.
  • Prilosec Prilosec Capsules (omeprazole delayed-release capsules) 20 mg to be administered orally with 120 mL water each morning after an overnight fast, 1 hour before starting breakfast.
  • Blood samples (3 mL) were obtained by venipuncture on Days 1 and 7 of both periods and Day 8 of Period 1 (for SAN-15 chewable tablets) within 30 minutes before each dose, and at 5, 10, 15, 20, 30, 45, 60, 90, 120, 150, 180, 210, 240, 300, 360, 420, 480, 540, 600, 660, and 720 minutes (12 hours) after each dose.
  • Zero time was the time that the subject ingested a chewable tablet or swallowed a capsule of Prilosec.
  • Plasma omeprazole concentrations were measured using a validated liquid chromatography mass spectrometry (LC-MS/MS) assay (MDS Pharma Services, Lincoln, Nebr.). The linear assay range was 5.0 to 750 ng/mL. The following pharmacokinetic parameters were measured for each subject:
  • Omeprazole was microencapsulated as described in Example 1B.
  • Dry-coated omeprazole are prepared by mixing the ingredients listed in table 16A-1 and dry-granulate the powder using roller compactor. Resulting granules are screened through a 10-mesh screen.
  • the dry granules are then mixed with the components shown in table 16A-2 until a homogeneous blend is obtained. This blend is then compressed using a 18 mm diameter tooling in a Stokes rotary press.
  • Dry-coated omeprazole are prepared by mixing the ingredients listed in the table 16B-1 and dry-granulate the powder using roller compactor. Resulting granules are screened through a 10-mesh screen.
  • the dry granules are then mixed with the components shown in table 16B-2 until a homogeneous blend is obtained. This blend is then compressed using a 18 mm diameter tooling in a Stokes rotary press.
  • Dry-coated omeprazole are prepared by mixing the ingredients listed in the table 16C-1 and dry-granulate the powder using roller compactor. Resulting granules are screened through a 10-mesh screen.
  • Example 16C-1 The dry granules prepared in Example 16C-1 are mixed with the ingredients shown in Table 16C-2 until a homogeneous blend is obtained. This blend is then compressed using oval-shaped tooling in a Stokes rotary press.

Abstract

In one general aspect of the present invention, pharmaceutical formulations comprising both a proton pump inhibitor microencapsulated or dry coated with a material that enhances the shelf-life of the pharmaceutical composition and one or more antacid are described. In another general aspect of the present invention, pharmaceutical formulations comprising both a proton pump inhibitor microencapsulated or dry coated with a taste-masking material and one or more antacid are described.

Description

  • This application is a continuation-in-part of U.S. patent application Ser. No. 10/893,203, filed on Jul. 16, 2004, which claims benefit under 35 U.S.C. §119(e) of U.S. Provisional Application No. 60/488,321, filed Jul. 18, 2003; the contents of these applications are incorporated by reference herein in their entirety.
  • FIELD OF THE INVENTION
  • The present invention is related to pharmaceutical formulations comprising an antacid and a proton pump inhibitor microencapsulated with (1) a material that enhances the shelf-life of the composition, and/or (2) a taste-masking material. Also provided herein are pharmaceutical compositions comprising proton pump inhibitor and antacid wherein the proton pump inhibitor is dry coated. In addition, methods for manufacture of the pharmaceutical formulations; uses of the pharmaceutical formulations in treating disease; and combinations of the pharmaceutical formulations with other therapeutic agents are described.
  • BACKGROUND OF THE INVENTION
  • Upon ingestion, most acid-labile pharmaceutical compounds must be protected from contact with acidic stomach secretions to maintain their pharmaceutical activity. To accomplish this, compositions with enteric-coatings have been designed to dissolve at a pH to ensure that the drug is released in the proximal region of the small intestine (duodenum), rather than the acidic environment of the stomach. However, due to the pH-dependent attributes of these enteric-coated compositions and the uncertainty of gastric retention time, in-vivo performance as well as both inter- and intra-subject variability are all major set backs of using enteric-coated systems for the controlled release of a drug.
  • Phillips et al. has described non-enteric coated pharmaceutical compositions. These compositions, which allow for the immediate release of the pharmaceutically active ingredient into the stomach, involve the administration of one or more buffering agents with an acid labile pharmaceutical agent, such as a proton pump inhibitor. The buffering agent is thought to prevent substantial degradation of the acid labile pharmaceutical agent in the acidic environment of the stomach by raising the pH. See, e.g., U.S. Pat. Nos. 5,840,737; 6,489,346; 6,645,988; and 6,699,885; and U.S. patent application Ser. No. 10/898,135.
  • A class of acid-labile pharmaceutical compounds that are administered as enteric-coated dosage forms are proton pump inhibiting agents. Exemplary proton pump inhibitors include, omeprazole (Prilosec®), lansoprazole (Prevacid®), esomeprazole (Nexium®), rabeprazole. Aciphex®), pantoprazole (Protonix®), pariprazole, tentaprazole, and leminoprazole. The drugs of this class suppress gastrointestinal acid secretion by the specific inhibition of the H+/K+-ATPase enzyme system (proton pump) at the secretory surface of the gastrointestinal parietal cell. Most proton pump inhibitors are susceptible to acid degradation and, as such, are rapidly destroyed as pH falls to an acidic level. Therefore, if the enteric-coating of these formulated products is disrupted (e.g., trituration to compound a liquid, or chewing the capsule or tablet) or the buffering agent fails to sufficiently neutralize the gastrointestinal pH, the drug will be exposed to degradation by the gastrointestinal acid in the stomach.
  • Omeprazole is one example of a proton pump inhibitor which is a substituted bicyclic aryl-imidazole, 5-methoxy-2-[(4-methoxy-3,5-dimethyl-2-pyridinyl)methyl]sulfinyl]-1H-benzimidazole, that inhibits gastrointestinal acid secretion. U.S. Pat. No. 4,786,505 to Lovgren et al. teaches that a pharmaceutical oral solid dosage form of omeprazole must be protected from contact with acidic gastrointestinal juice by an enteric-coating to maintain its pharmaceutical activity and describes an enteric-coated omeprazole preparation containing one or more subcoats between the core material and the enteric-coating.
  • Proton pump inhibitors are typically prescribed for short-term treatment of active duodenal ulcers, gastrointestinal ulcers, gastro esophageal reflux disease (GERD), severe erosive esophagitis, poorly responsive symptomatic GERD, and pathological hypersecretory conditions such as Zollinger Ellison syndrome. These above-listed conditions commonly arise in healthy or critically ill patients of all ages, and may be accompanied by significant upper gastrointestinal bleeding.
  • It is believed that omeprazole, lansoprazole and other proton pump inhibiting agents reduce gastrointestinal acid production by inhibiting H+/K+-ATPase of the parietal cell during the final common pathway for gastrointestinal acid secretion. See, e.g., Fellenius et al., Substituted Benzimidazoles Inhibit Gastrointestinal Acid Secretion by Blocking H+/K+-ATPase, Nature, 290: 159-161 (1981); Wallmark et al., The Relationship Between Gastrointestinal Acid Secretion and Gastrointestinal H+/K+-ATPase Activity, J. Biol. Chem., 260: 13681-13684 (1985); and Fryklund et al., Function and Structure of Parietal Cells After H+/K+-ATPase Blockade, Am. J. Physiol., 254 (1988).
  • Proton pump inhibitors have the ability to act as weak bases which reach parietal cells from the blood and diffuse into the secretory canaliculi. There the drugs become protonated and thereby trapped. The protonated compound can then rearrange to form a sulfenamide which can covalently interact with sulthydryl groups at critical sites in the extra cellular (luminal) domain of the membrane-spanning H+/K+-ATPase. See, e.g., Hardman et al., Goodman & Gilman's The Pharmacological Basis of Therapeutics, 907 (9th ed. 1996). As such, proton pump inhibitors are prodrugs that must be activated to be effective. The specificity of the effects of proton pump inhibiting agents is also dependent upon: (a) the selective distribution of H+/K+-ATPase; (b) the requirement for acidic conditions to catalyze generation of the reactive inhibitor; and (c) the trapping of the protonated drug and the cationic sulfenamide within the acidic canaliculi and adjacent to the target enzyme. See, e.g., Hardman et al.
  • Still, there remains a need for a pharmaceutical formulation that releases a proton pump inhibitor into the gastrointestinal tract for absorption of an intact, non-acid degraded or non-acid reacted form of a proton pump inhibitor into the bloodstream of a subject in either a fed or fasted state which exhibits enhanced shelf-life stability and improved patient compliance. The discussion that follows discloses pharmaceutical formulations comprising microencapsulated or dry coated proton pump inhibitors and one or more antacids which help to fulfill these needs.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 is a graph comparing the pharmacokinetic release profiles of omeprazole of Prilosec®, non-microencapsulated omeprazole with antacid (as described in Example 13B), omeprazole microencapsulated with Klucel and antacid tablet (as described in Example 13C), and omeprazole microencapsulated with Methocel and antacid tablet (as described in Example 13 D) in human.
  • FIGS. 2A and 2B are SEM micrographs of micronized omeprazole and omeprazole microencapsulated with Klucel® Hydroxypropyl Cellulose.
  • FIG. 3 is a graph comparing the average pharmacokinetic release profiles of SAN-15A, SAN-15B, SAN-15C, SAN-20D and SAN-20E as compared to Priolosec® brand enteric coated omeprazole 40 mg. Formulations were prepared as described in Example 13.
  • FIG. 4 is a graph comparing the mean peak plasma concentration (Cmax) versus the time at which Cmax is observed (Tmax) for SAN-15 (20 mg and 40 mg), SAN-20D, SAN-20E and Priolosec® brand enteric coated omeprazole 20 mg and 40 mg, Day 1. The data is from the human clinical trial described in Examples 14B and 15B.
  • FIG. 5 is a graph comparing the average pharmacokinetic release profiles for SAN-15 20 mg and Priolosec® brand enteric coated omeprazole 20 mg, Day 1. The data is from the human clinical trial described in Examples 14B.
  • FIG. 6 is a graph comparing the average pharmacokinetic release profiles for SAN-15 40 mg and Priolosec® brand enteric coated omeprazole 40 mg, Day 1. The data is from the human clinical trial described in Examples 15B.
  • FIG. 7 is a graph comparing the average pharmacokinetic release profiles for SAN-15 20 mg and Priolosec® brand enteric coated omeprazole 20 mg, Day 7. The data is from the human clinical trial described in Examples 14B.
  • FIG. 8 is a graph comparing the average pharmacokinetic release profiles for SAN-15 40 mg and Priolosec® brand enteric coated omeprazole 40 mg, Day 7. The data is from the human clinical trial described in Examples 15B.
  • FIG. 9 is a graph comparing the mean peak plasma concentration (Cmax) versus the time at which Cmax is observed (Tmax) for SAN-15 (20 mg and 40 mg) and Priolosec® brand enteric coated omeprazole 20 mg and 40 mg, Day 7. The data is from the human clinical trial described in Examples 14B and 15B.
  • FIG. 10 is a graph comparing the mean peak plasma concentration (Cmax) versus the time at which Cmax is observed (Tmax) for SAN-15 (20 mg and 40 mg) and Priolosec® brand enteric coated omeprazole 20 mg and 40 mg, Day 1 and Day 7. The data is from the human clinical trial described in Examples 14B and 15B.
  • SUMMARY OF THE INVENTION
  • Provided herein are pharmaceutical formulations having enhanced shelf-lives comprising, at least one acid labile proton pump inhibitor which is microencapsulated with a material that enhances the shelf-life of the pharmaceutical formulation and at least one antacid; wherein an initial serum concentration of the proton pump inhibitor is greater than about 0.1 μg/ml at any time within about 30 minutes after administration of the pharmaceutical formulation. Also provided herein are taste-masked pharmaceutical formulations comprising at least one acid labile proton pump inhibitor which is microencapsulated with a taste-masking material and at least one antacid; wherein an initial serum concentration of the proton pump inhibitor is greater than about 0.1 μg/ml at any time within about 30 minutes after administration of the pharmaceutical formulation.
  • In various embodiments provided herein, the proton pump inhibitor is microencapsulated with one or more compounds selected from cellulose hydroxypropyl ethers; low-substituted hydroxypropyl ethers; cellulose hydroxypropyl methyl ethers; methylcellulose polymers; ethylcelluloses and mixtures thereof; polyvinyl alcohol; hydroxyethylcelluloses; carboxymethylcelluloses and salts of carboxymethylcelluloses; polyvinyl alcohol and polyethylene glycol co-polymers; monoglycerides; triglycerides; polyethylene glycols, modified food starch, acrylic polymers; mixtures of acrylic polymers with cellulose ethers; cellulose acetate phthalate; sepifilms, cyclodextrins; and mixtures thereof.
  • In various embodiments provided herein, the proton pump inhibitor is microencapsulated with one or more additives to enhance the processing or performance of microencapsulation. Such additives maybe pH modifier, plastersizer, antioxidant, or sweetener or flavor.
  • Provided herein are pharmaceutical formulations having enhanced shelf-lives comprising, at least one acid labile proton pump inhibitor and at least one antacid; wherein an initial serum concentration of the proton pump inhibitor is greater than about 0.1 μg/ml at any time within about 30 minutes after administration of the pharmaceutical formulation, wherein some or all of the proton pump inhibitor is dry coated.
  • In other embodiments, the at least one antacid comprises at least one soluble antacid. In some embodiments, the soluble antacid is sodium bicarbonate. In various embodiments, the at least one buffer is selected from sodium bicarbonate, calcium carbonate, sodium carbonate, magnesium oxide, magnesium hydroxide, magnesium carbonate, aluminum hydroxide, and mixtures thereof.
  • Provided herein are methods of extending the shelf-life of pharmaceutical formulations comprising microencapsulating at least one acid labile proton pump inhibitor with a material that enhances the shelf-life; and combining the microencapsulated acid labile proton pump inhibitor with at least one antacid. Also provided herein are methods of masking the taste of a pharmaceutical formulation comprising microencapsulating at least one acid labile proton pump inhibitor with a taste-masking material; and combining the microencapsulated acid labile proton pump inhibitor with an antacid.
  • Also provided herein are methods of making a pharmaceutical formulation with and extended shelf-life by dry coating the proton pump inhibitor. In some embodiments, the proton pump inhibitor is combined with some or all of the antacid to form a slug or sheet of material. This intermediate product is then broken into granular material with is combined with other components present in the pharmaceutical formulation. In other embodiments, the dry coated proton pump inhibitor is combined with additional antacid.
  • In various embodiments of the present invention, the pharmaceutical formulations may further comprise one or more excipients selected from parietal cell activators, organic solvents, erosion facilitators, diffusion facilitators, antioxidants, flavoring agents and carrier materials selected from binders, suspending agents, disintegration agents, filling agents, surfactants, solubilizers, stabilizers, lubricants, wetting agents, diluents, anti-adherents, and antifoaming agents.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention is directed to pharmaceutical formulations exhibiting enhanced shelf-life stability and/or improved taste masking properties useful for the treatment of a disease, condition or disorder. Methods of treatment using the pharmaceutical formulations of the present invention are also described.
  • It has been discovered that pharmaceutical compositions comprising (1) an acid labile proton pump inhibitor which is microencapsulated with a material that enhances the shelf-life of the pharmaceutical composition together with (2) one or more antacid, provide superior performance by enhancing shelf-life stability of the pharmaceutical formulation during manufacturing and storage.
  • It has also been discovered that pharmaceutical compositions comprising (1) an acid labile proton pump inhibitor which has been dry coated and (2) one or more antacids, provide superior performance by enhancing shelf-life stability of the pharmaceutical formulation during manufacturing and storage. In some embodiments, the proton pump inhibitor is dry coated with some or all of the antacid. In other embodiments, the dry coated proton pump inhibitor is combined with a second antacid which can be the same antacid as used to dry coat the proton pump inhibitor or a different antacid. In some embodiments, the dry coated material is combined with one or more pharmaceutical excipients. In yet other embodiments, the proton pump inhibitor is dry coated with a material comprising antacid, sweetener(s), lubricant, and binder.
  • Certain taste-masking materials have also been discovered which, when used in the pharmaceutical formulations provide (1) more palatable forms of the drug by blocking the contact of the unpleasant taste of the pharmaceutical agent from the contact of the taste receptor, thereby increasing patient compliance; and/or (2) require lower amounts of traditional flavoring agents.
  • To more readily facilitate an understanding of the invention and its preferred embodiments, the meanings of terms used herein will become apparent from the context of this specification in view of common usage of various terms and the explicit definitions of other terms provided in the glossary below or in the ensuing description.
  • GLOSSARY
  • As used herein, the terms “comprising,” “including,” and “such as” are used in their open, non-limiting sense.
  • The term “about” is used synonymously with the term “approximately.” Illustratively, the use of the term “about” indicates that values slightly outside the cited values, i.e. dosages comprising plus or minus 0.1% to 10%, which are also effective and safe. As one of ordinary skill in the art would understand, such dosages are thus encompassed by the scope of the claims reciting the terms “about” and “approximately.”
  • The phrase “acid-labile pharmaceutical agent” refers to any pharmacologically active drug subject to acid catalyzed degradation.
  • “Aftertaste” is a measurement of all sensation remaining after swallowing. Aftertaste can be measured, e.g., from 30 seconds after swallowing, 1 minutes after swallowing, 2 minutes after swallowing, 3 minutes after swallowing, 4 minutes after swallowing, 5 minutes after swallowing, and the like.
  • “Amplitude” is the initial overall perception of the flavors balance and fullness. The amplitude scale is 0-none, 1-low, 2-moderate, and 3-high.
  • “Anti-adherents,” “glidants,” or “anti-adhesion” agents prevent components of the formulation from aggregating or sticking and improve flow characteristics of a material. Such compounds include, e.g., colloidal silicon dioxide such as Cab-o-sil®; tribasic calcium phosphate, talc, corn starch, DL-leucine, sodium lauryl sulfate, magnesium stearate, calcium stearate, sodium stearate, kaolin, and micronized amorphous silicon dioxide (Syloid®) and the like.
  • “Antifoaming agents” reduce foaming during processing which can result in coagulation of aqueous dispersions, bubbles in the finished film, or generally impair processing. Exemplary anti-foaming agents include silicon emulsions or sorbitan sesquoleate.
  • “Antioxidants” include, e.g., butylated hydroxytoluene (BHT), sodium ascorbate, and tocopherol.
  • “Binders” impart cohesive qualities and include, e.g., alginic acid and salts thereof; cellulose derivatives such as carboxymethylcellulose, methylcellulose (e.g., Methocel®), hydroxypropylmethylcellulose, hydroxyethylcellulose, hydroxypropylcellulose (e.g., Klucel®), ethylcellulose (e.g., Ethocel®), and microcrystalline cellulose (e.g., Avicel®); microcrystalline dextrose; amylose; magnesium aluminum silicate; polysaccharide acids; bentonites; gelatin; polyvinylpyrrolidone/vinyl acetate copolymer; crospovidone; povidone; starch; pregelatinized starch; tragacanth, dextrin, a sugar, such as sucrose (e.g., Dipac®), glucose, dextrose, molasses, mannitol, sorbitol, xylitol (e.g., Xylitab®), and lactose; a natural or synthetic gum such as acacia, tragacanth, ghatti gum, mucilage of isapol husks, polyvinylpyrrolidone (e.g., Polyvidone® CL, Kollidon® CL, Polyplasdone® XL-10), larch arabogalactan, Veegum®, polyethylene glycol, waxes, sodium alginate, and the like.
  • “Bioavailability” refers to the extent to which an active moiety, e.g., drug, prodrug, or metabolite, is absorbed into the general circulation and becomes available at the site of drug action in the body. Thus, a proton pump inhibitor administered through IV is 100% bioavailable. “Oral bioavailability” refers to the extent to with the proton pump inhibitor is absorbed into the general circulation and becomes available at the site of the drug action in the body when the pharmaceutical formulation is taken orally.
  • “Bioequivalence” or “bioequivalent” means that the area under the serum concentration time curve (AUC) and the peak serum concentration (Cmax) are each within 80% and 125% with a 90% confidence interval.
  • “Carrier materials” include any commonly used excipients in pharmaceutics and should be selected on the basis of compatibility with the proton pump inhibitor and the release profile properties of the desired dosage form. Exemplary carrier materials include, e.g., binders, suspending agents, disintegration agents, filling agents, surfactants, solubilizers, stabilizers, lubricants, wetting agents, diluents, and the like. “Pharmaceutically compatible carrier materials” may comprise, e.g., acacia, gelatin, colloidal silicon dioxide, calcium glycerophosphate, calcium lactate, maltodextrin, glycerine, magnesium silicate, sodium caseinate, soy lecithin, sodium chloride, tricalcium phosphate, dipotassium phosphate, sodium stearoyl lactylate, carrageenan, monoglyceride, diglyceride, pregelatinized starch, and the like. See, e.g., Remington: The Science and Practice of Pharmacy, Nineteenth Ed (Easton, Pa.: Mack Publishing Company, 1995); Hoover, John E., Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, Pa. 1975; Liberman, H. A. and Lachman, L., Eds., Pharmaceutical Dosage Forms, Marcel Decker, New York, N.Y., 1980; and Pharmaceutical Dosage. Forms and Drug Delivery Systems, Seventh Ed. (Lippincott Williams & Wilkins 1999).
  • “Character notes” include, e.g., aromatics, basis tastes, and feeling factors. The intensity of the character note can be scaled from 0-none, 1-slight, 2-moderate, or 3-strong.
  • A “derivative” is a compound that is produced from another compound of similar structure by the replacement of substitution of an atom, molecule or group by another suitable atom, molecule or group. For example, one or more hydrogen atom of a compound may be substituted by one or more alkyl, acyl, amino, hydroxyl, halo, haloalkyl, aryl, heteroaryl, cycloaolkyl, heterocycloalkyl, or heteroalkyl group to produce a derivative of that compound.
  • “Diffusion facilitators” and “dispersing agents” include materials that control the diffusion of an aqueous fluid through a coating. Exemplary diffusion facilitators/dispersing agents include, e.g., hydrophilic polymers, electrolytes, Tween® 60 or 80, PEG and the like. Combinations of one or more erosion facilitator with one or more diffusion facilitator can also be used in the present invention.
  • “Diluents” increase bulk of the composition to facilitate compression. Such compounds include e.g., lactose; starch; mannitol; sorbitol; dextrose; microcrystalline cellulose such as Avicel®; dibasic calcium phosphate; dicalcium phosphate dihydrate; tricalcium phosphate; calcium phosphate; anhydrous lactose; spray-dried lactose; pregelatinzed starch; compressible sugar, such as Di-Pac® (Amstar); mannitol; hydroxypropylmethylcellulose; sucrose-based diluents; confectioner's sugar; monobasic calcium sulfate monohydrate; calcium sulfate dihydrate; calcium lactate trihydrate; dextrates; hydrolyzed cereal solids; amylose; powdered cellulose; calcium carbonate; glycine; kaolin; mannitol; sodium chloride; inositol; bentonite; and the like.
  • The term “disintegrate” includes both the dissolution and dispersion of the dosage form when contacted with gastrointestinal fluid.
  • “Disintegration agents” facilitate the breakup or disintegration of a substance. Examples of disintegration agents include a starch, e.g., a natural starch such as corn starch or potato starch, a pregelatinized starch such as National 1551 or Amijel®, or sodium starch glycolate such as Promogel® or Explotab®; a cellulose such as a wood product, methylcrystalline cellulose, e.g., Avicel®, Avicel® PH101, Avicel® PH102, Avicel® PH105, Elcema® P100, Emcocel®, Vivacel®, Ming Tia®, and Solka-Floc®, methylcellulose, croscarmellose, or a cross-linked cellulose, such as cross-linked sodium carboxymethylcellulose (Ac-Di-Sol®), cross-linked carboxymethylcellulose, or cross-linked croscarmellose; a cross-linked starch such as sodium starch glycolate; a cross-linked polymer such as crospovidone; a cross-linked polyvinylpyrrolidone; alginate such as alginic acid or a salt of alginic acid such as sodium alginate; a clay such as Veegum® HV (magnesium aluminum silicate); a gum such as agar, guar, locust bean, Karaya, pectin, or tragacanth; sodium starch glycolate; bentonite; a natural sponge; a surfactant; a resin such as a cation-exchange resin; citrus pulp; sodium lauryl sulfate; sodium lauryl sulfate in combination starch; and the like.
  • “Drug absorption” or “absorption” refers to the process of movement from the site of administration of a drug toward the systemic circulation, e.g., into the bloodstream of a subject.
  • “Dry coating” is a method of coating the proton pump inhibitor with one or more other components without using water or other solvents.
  • “Dry granulation” is a method of converting powder particles into granules using the application of pressure without the use of a liquid.
  • An “enteric coating” is a substance that remains substantially intact in the stomach but dissolves and releases the drug once the small intestine is reached. Generally, the enteric coating comprises a polymeric material that prevents release in the low pH environment of the stomach but that ionizes at a slightly higher pH, typically a pH of 4 or 5, and thus dissolves sufficiently in the small intestines to gradually release the active agent therein.
  • The “enteric form of the proton pump inhibitor” is intended to mean that some or most of the proton pump inhibitor has been enterically coated to ensure that at least some of the drug is released in the proximal region of the small intestine (duodenum), rather than the acidic environment of the stomach.
  • “Erosion facilitators” include materials that control the erosion of a particular material in gastrointestinal fluid. Erosion facilitators are generally known to those of ordinary skill in the art. Exemplary erosion facilitators include, e.g., hydrophilic polymers, electrolytes, proteins, peptides, and amino acids.
  • “Filling agents” include compounds such as lactose, calcium carbonate, calcium phosphate, dibasic calcium phosphate, calcium sulfate, microcrystalline cellulose, cellulose powder, dextrose, dextrates, dextran, starches, pregelatinized starch, sucrose, xylitol, lactitol, mannitol, sorbitol, sodium chloride, polyethylene glycol, and the like.
  • “Flavoring agents” or “sweeteners” useful in the pharmaceutical compositions of the present invention include, e.g., acacia syrup, acesulfame K, alitame, anise, apple, aspartame, banana, Bavarian cream, berry, black currant, butterscotch, calcium citrate, camphor, caramel, cherry, cherry cream, chocolate, cinnamon, bubble gum, citrus, citrus punch, citrus cream, cotton candy, cocoa, cola, cool cherry, cool citrus, cyclamate, cylamate, dextrose, eucalyptus, eugenol, fructose, fruit punch, ginger, glycyrrhetinate, glycyrrhiza (licorice) syrup, grape, grapefruit, honey, isomalt, lemon, lime, lemon cream, monoammonium glyrrhizinate (MagnaSweet®), maltol, mannitol, maple, marshmallow, menthol, mint cream, mixed berry, neohesperidine DC, neotame, orange, pear, peach, peppermint, peppermint cream, Prosweet® Powder, raspberry, root beer, rum, saccharin, safrole, sorbitol, spearmint, spearmint cream, strawberry, strawberry cream, stevia, sucralose, sucrose, sodium saccharin, saccharin, aspartame, acesulfame potassium, mannitol, talin, sylitol, sucralose, sorbitol, Swiss cream, tagatose, tangerine, thamnatin, tutti fruitti, vanilla, walnut, watermelon, wild cherry, wintergreen, xylitol, or any combination of these flavoring ingredients, e.g., anise-menthol, cherry-anise, cinnamon-orange, cherry-cinnamon, chocolate-mint, honey-lemon, lemon-lime, lemon-mint, menthol-eucalyptus, orange-cream, vanilla-mint, and mixtures thereof.
  • “Gastrointestinal fluid” is the fluid of stomach secretions of a subject or the saliva of a subject after oral administration of a composition of the present invention, or the equivalent thereof. An “equivalent of stomach secretion” includes, e.g., an in vitro fluid having similar content and/or pH as stomach secretions such as a 1% sodium dodecyl sulfate solution or 0.1N HCl solution in water.
  • “Half-life” refers to the time required for the plasma drug concentration or the amount in the body to decrease by 50% from its maximum concentration.
  • “Lubricants” are compounds that prevent, reduce or inhibit adhesion or friction of materials. Exemplary lubricants include, e.g., stearic acid; calcium hydroxide; talc; sodium stearyl fumerate; a hydrocarbon such as mineral oil, or hydrogenated vegetable oil such as hydrogenated soybean oil (Sterotex®); higher fatty acids and their alkali-metal and alkaline earth metal salts, such as aluminum, calcium, magnesium, zinc, stearic acid, sodium stearates, glycerol, talc, waxes, Stearowet®, boric acid, sodium benzoate, sodium acetate, sodium chloride, leucine, a polyethylene glycol or a methoxypolyethylene glycol such as Carbowax™, sodium oleate, glyceryl behenate, polyethylene glycol, magnesium or sodium lauryl sulfate, colloidal silica such as Syloid™, Carb-O-Sil®, a starch such as corn starch, silicone oil, a surfactant, and the like.
  • A “measurable serum concentration” or “measurable plasma concentration” describes the blood serum or blood plasma concentration, typically measured in mg, μg, or ng of therapeutic agent per ml, dl, or l of blood serum, of a therapeutic agent that is absorbed into the bloodstream after administration. One of ordinary skill in the art would be able to measure the serum concentration or plasma concentration of a proton pump inhibitor or other therapeutic agent. See, e.g., Gonzalez H. et al., J. Chromatogr. B. Analyt. Technol. Biomed. Life Sci., vol. 780, pp 459-65, (Nov. 25, 2002).
  • “Parietal cell activators” or “activators” stimulate the parietal cells and enhance the pharmaceutical activity of the proton pump inhibitor. Parietal cell activators include, e.g., chocolate; alkaline substances such as sodium bicarbonate; calcium such as calcium carbonate, calcium gluconate, calcium hydroxide, calcium acetate and calcium glycerophosphate; peppermint oil; spearmint oil; coffee; tea and colas (even if decaffeinated); caffeine; theophylline; theobromine; amino acids (particularly aromatic amino acids such as phenylalanine and tryptophan); and combinations thereof.
  • “Pharmacodynamics” refers to the factors that determine the biologic response observed relative to the concentration of drug at a site of action.
  • “Pharmacokinetics” refers to the factors that determine the attainment and maintenance of the appropriate concentration of drug at a site of action.
  • “Plasma concentration” refers to the concentration of a substance in blood plasma or blood serum of a subject. It is understood that the plasma concentration of a therapeutic agent may vary many-fold between subjects, due to variability with respect to metabolism of therapeutic agents. In accordance with one aspect of the present invention, the plasma concentration of a proton pump inhibitors and/or other therapeutic agent may vary from subject to subject. Likewise, values such as maximum plasma concentration (Cmax) or time to reach maximum serum concentration (Tmax), or area under the serum concentration time curve (AUC) may vary from subject to subject. Due to this variability, the amount necessary to constitute “a therapeutically effective amount” of proton pump inhibitor or other therapeutic agent, may vary from subject to subject. It is understood that when mean plasma concentrations are disclosed for a population of subjects, these mean values may include substantial variation.
  • “Plasticizers” are compounds used to soften the microencapsulation material or film coatings to make them less brittle. Suitable plasticizers include, e.g., polyethylene glycols such as PEG 300, PEG 400, PEG 600, PEG 1450, PEG 3350, and PEG 800, stearic acid, propylene glycol, oleic acid, and triacetin.
  • “Prevent” or “prevention” when used in the context of a gastric acid related disorder means no gastrointestinal disorder or disease development if none had occurred, or no further gastrointestinal disorder or disease development if there had already been development of the gastrointestinal disorder or disease. Also considered is the ability of one to prevent some or all of the symptoms associated with the gastrointestinal disorder or disease.
  • A “prodrug” refers to a drug or compound in which the pharmacological action results from conversion by metabolic processes within the body. Prodrugs are generally drug precursors that, following administration to a subject and subsequent absorption, are converted to an active, or a more active species via some process, such as conversion by a metabolic pathway. Some prodrugs have a chemical group present on the prodrug that renders it less active and/or confers solubility or some other property to the drug. Once the chemical group has been cleaved and/or modified from the prodrug the active drug is generated. Prodrugs may be designed as reversible drug derivatives, for use as modifiers to enhance drug transport to site-specific tissues. The design of prodrugs to date has been to increase the effective water solubility of the therapeutic compound for targeting to regions where water is the principal solvent. See, e.g., Fedorak et al., Am. J. Physiol., 269:G210-218 (1995); McLoed et al., Gastroenterol, 106:405-413 (1994); Hochhaus et al., Biomed. Chrom., 6:283-286 (1992); J. Larsen and H. Bundgaard, Int. J. Pharmaceutics, 37, 87 (1987); J. Larsen et al., Int. J. Pharmaceutics, 47, 103 (1988); Sinkula et al., J. Pharm. Sci., 64:181-210 (1975); T. Higuchi and V. Stella, Pro-drugs as Novel Delivery Systems, Vol. 14 of the A.C.S. Symposium Series; and Edward B. Roche, Bioreversible Carriers in Drug Design, American Pharmaceutical Association and Pergamon Press, 1987.
  • “Proton pump inhibitor product” refers to a product sold on the market. Proton pump inhibitor products include, for example, Priolosec®, Nexium®, Prevacid®, Protonix®, and Aciphex®.
  • “Serum concentration” refers to the concentration of a substance such as a therapeutic agent, in blood plasma or blood serum of a subject. It is understood that the serum concentration of a therapeutic agent may vary many-fold between subjects, due to variability with respect to metabolism of therapeutic agents. In accordance with one aspect of the present invention, the serum concentration of a proton pump inhibitors and/or prokinetic agent may vary from subject to subject. Likewise, values such as maximum serum concentration (Cmax) or time to reach maximum serum concentration (Tmax), or total area under the serum concentration time curve (AUC) may vary from subject to subject. Due to this variability, the amount necessary to constitute “a therapeutically effective amount” of proton pump inhibitor, prokinetic agent, or other therapeutic agent, may vary from subject to subject. It is understood that when mean serum concentrations are disclosed for a population of subjects, these mean values may include substantial variation.
  • “Solubilizers” include compounds such as citric acid, succinic acid, fumaric acid, malic acid, tartaric acid, maleic acid, glutaric acid, sodium bicarbonate, sodium carbonate and the like.
  • “Stabilizers” include compounds such as any antioxidation agents, buffers, acids, and the like.
  • “Suspending agents” or “thickening agents” include compounds such as polyvinylpyrrolidone, e.g., polyvinylpyrrolidone K12, polyvinylpyrrolidone K17, polyvinylpyrrolidone K25, or polyvinylpyrrolidone K30; polyethylene glycol, e.g., the polyethylene glycol can have a molecular weight of about 300 to about 6000, or about 3350 to about 4000, or about 7000 to about 5400; sodium carboxymethylcellulose; methylcellulose; hydroxypropylmethylcellulose; polysorbate-80; hydroxyethylcellulose; sodium alginate; gums, such as, e.g., gum tragacanth and gum acacia; guar gum; xanthans, including xanthan gum; sugars; cellulosics, such as, e.g., sodium carboxymethylcellulose, methylcellulose, sodium carboxymethylcellulose, hydroxypropylmethylcellulose, hydroxyethylcellulose; polysorbate-80; sodium alginate; polyethoxylated sorbitan monolaurate; polyethoxylated sorbitan monolaurate; povidone and the like.
  • “Surfactants” include compounds such as sodium lauryl sulfate, sorbitan monooleate, polyoxyethylene sorbitan monooleate, polysorbates, polaxomers, bile salts, glyceryl monostearate, copolymers of ethylene oxide and propylene oxide, e.g., Pluronic® (BASF); and the like.
  • A “therapeutically effective amount” or “effective amount” is that amount of a pharmaceutical agent to achieve a pharmacological effect. The term “therapeutically effective amount” includes, for example, a prophylactically effective amount. An “effective amount” of a proton pump inhibitor is an amount effective to achieve a desired pharmacologic effect or therapeutic improvement without undue adverse side effects. For example, an effective amount of a proton pump inhibitor refers to an amount of proton pump inhibitor that reduces acid secretion, or raises gastrointestinal fluid pH, or reduces gastrointestinal bleeding, or reduces the need for blood transfusion, or improves survival rate, or provides for a more rapid recovery from a gastric acid related disorder. The effective amount of a pharmaceutical agent will be selected by those skilled in the art depending on the particular patient and the disease level. It is understood that “an effect amount” or “a therapeutically effective amount” can vary from subject to subject, due to variation in metabolism of therapeutic agents such as proton pump inhibitors and/or prokinetic agents, age, weight, general condition of the subject, the condition being treated, the severity of the condition being treated, and the judgment of the prescribing physician.
  • “Total intensity of aroma” is the overall immediate impression of the strength of the aroma and includes both aromatics and nose feel sensations.
  • “Total intensity of flavor” is the overall immediate impression of the strength of the flavor including aromatics, basic tastes and mouth feel sensations.
  • “Treat” or “treatment” as used in the context of a gastric acid related disorder refers to any treatment of a disorder or disease associated with a gastrointestinal disorder, such as preventing the disorder or disease from occurring in a subject which may be predisposed to the disorder or disease, but has not yet been diagnosed as having the disorder or disease; inhibiting the disorder or disease, e.g., arresting the development of the disorder or disease, relieving the disorder or disease, causing regression of the disorder or disease, relieving a condition caused by the disease or disorder, or stopping the symptoms of the disease or disorder. Thus, as used herein, the term “treat” is used synonymously with the term “prevent.”
  • “Wetting agents” include compounds such as oleic acid, glyceryl monostearate, sorbitan monooleate, sorbitan monolaurate, triethanolamine oleate, polyoxyethylene sorbitan monooleate, polyoxyethylene sorbitan monolaurate, sodium oleate, sodium lauryl sulfate, and the like.
  • Proton Pump Inhibitors
  • The terms “proton pump inhibitor,” “PPI,” and “proton pump inhibiting agent” can be used interchangeably to describe any acid labile pharmaceutical agent possessing pharmacological activity as an inhibitor of H+/K+-ATPase. A proton pump inhibitor may, if desired, be in the form of free base, free acid, salt, ester, hydrate, anhydrate, amide, enantiomer, isomer, tautomer, prodrug, polymorph, derivative, or the like, provided that the free base, salt, ester, hydrate, amide, enantiomer, isomer, tautomer, prodrug, or any other pharmacologically suitable derivative is therapeutically active.
  • In various embodiments, the proton pump inhibitor can be a substituted bicyclic aryl-imidazole, wherein the aryl group can be, e.g., a pyridine, a phenyl, or a pyrimidine group and is attached to the 4- and 5-positions of the imidazole ring. Proton pump inhibitors comprising a substituted bicyclic aryl-imidazoles include, but are not limited to, omeprazole, hydroxyomeprazole, esomeprazole, lansoprazole, pantoprazole, rabeprazole, dontoprazole, habeprazole, perprazole, tenatoprazole, ransoprazole, pariprazole, leminoprazole, or a free base, free acid, salt, hydrate, ester, amide, enantiomer, isomer, tautomer, polymorph, prodrug, or derivative thereof. See, e.g., The Merck Index, Merck & Co. Rahway, N.J. (2001).
  • Other proton pump inhibitors include but are not limited to: soraprazan (Altana); ilaprazole (U.S. Pat. No. 5,703,097) (Il-Yang); AZD-0865 (AstraZeneca); YH-1885 (PCT Publication WO 96/05177) (SB-641257) (2-pyrimidinamine, 4-(3,4-dihydro-1-methyl-2(1H)-isoquinolinyl)-N-(4-fluorophenyl)-5,6-dimethyl-monohydrochloride)(YuHan); BY-112 (Altana); SPI-447 (Imidazo(1,2-a)thieno(3,2-c)pyridin-3-amine,5-methyl-2-(2-methyl-3-thienyl) (Shinnippon); 3-hydroxymethyl-2-methyl-9-phenyl-7H-8,9-dihydro-pyrano(2,3-c)-imidazo(1,2-a)pyridine (PCT Publication WO 95/27714) (AstraZeneca); Pharmaprojects No. 4950 (3-hydroxymethyl-2-methyl-9-phenyl-7H-8,9-dihydro-pyrano(2,3-c)-imidazo(1,2-a)pyridine) (AstraZeneca, ceased) WO 95/27714; Pharmaprojects No. 4891 (EP 700899) (Aventis); Pharmaprojects No. 4697 (PCT Publication WO 95/32959) (AstraZeneca); H-335/25 (AstraZeneca); T-330 (Saitama 335) (Pharmacological Research Lab); Pharmaprojects No. 3177 (Roche); BY-574 (Altana); Pharmaprojects No. 2870 (Pfizer); AU-1421 (EP 264883) (Merck); AU-2064 (Merck); AY-28200 (Wyeth); Pharmaprojects No. 2126 (Aventis); WY-26769 (Wyeth); pumaprazole (PCT Publication WO 96/05199) (Altana); YH-1238 (YuHan); Pharmaprojects No. 5648 (PCT Publication WO 97/32854) (Dainippon); BY-686 (Altana); YM-020 (Yamanouchi); GYKI-34655 (Ivax); FPL-65372 (Aventis); Pharmaprojects No. 3264 (EP 509974) (AstraZeneca); nepaprazole (Toa Eiyo); RN-11203 (Nycomed Pharma); OPC-22575; pumilacidin A (BMS); saviprazole (EP 234485) (Aventis); SKandF-95601 (GSK, discontinued); Pharmaprojects No. 2522 (EP 204215) (Pfizer); S-3337 (Aventis); RS-13232A (Roche); AU-1363 (Merck); SKandF-96067 (EP 259174) (Altana); SUN 8176 (Daiichi Phama); Ro-18-5362 (Roche); ufiprazole (EP 74341) (AstraZeneca); and Bay-p-1455 (Bayer); or a free base, free acid, salt, hydrate, ester, amide, enantiomer, isomer, tautomer, polymorph, prodrug, or derivative of these compounds.
  • Still other proton pump inhibitors contemplated by the present invention include those described in the following U.S. Pat. Nos. 4,628,098; 4,689,333; 4,786,505; 4,853,230; 4,965,269; 5,021,433; 5,026,560; 5,045,321; 5,093,132; 5,430,042; 5,433,959; 5,576,025; 5,639,478; 5,703,110; 5,705,517; 5,708,017; 5,731,006; 5,824,339; 5,855,914; 5,879,708; 5,948,773; 6,017,560; 6,123,962; 6,187,340; 6,296,875; 6,319,904; 6,328,994; 4,255,431; 4,508,905; 4,636,499; 4,738,974; 5,690,960; 5,714,504; 5,753,265; 5,817,338; 6,093,734; 6,013,281; 6,136,344; 6,183,776; 6,328,994; 6,479,075; 6,559,167.
  • Other substituted bicyclic aryl-imidazole compounds as well as their salts, hydrates, esters, amides, enantiomers, isomers, tautomers, polymorphs, prodrugs, and derivatives may be prepared using standard procedures known to those skilled in the art of synthetic organic chemistry. See, e.g., March, Advanced Organic Chemistry: Reactions, Mechanisms and Structure, 4th Ed. (New York: Wiley-Interscience, 1992); Leonard et al., Advanced Practical Organic Chemistry (1992); Howarth et al., Core Organic Chemistry (1998); and Weisermel et al., Industrial Organic Chemistry (2002).
  • “Pharmaceutically acceptable salts,” or “salts,” include, e.g., the salt of a proton pump inhibitor prepared from formic, acetic, propionic, succinic, glycolic, gluconic, lactic, malic, tartaric, citric, ascorbic, glucuronic, maleic, fumaric, pyruvic, aspartic, glutamic, benzoic, anthranilic, mesylic, stearic, salicylic, p-hydroxybenzoic, phenylacetic, mandelic, embonic, methanesulfonic, ethanesulfonic, benzenesulfonic, pantothenic, toluenesulfonic, 2-hydroxyethanesulfonic, sulfanilic, cyclohexylaminosulfonic, algenic, β-hydroxybutyric, galactaric and galacturonic acids.
  • In one embodiment, acid addition salts are prepared from the free base using conventional methodology involving reaction of the free base with a suitable acid. Suitable acids for preparing acid addition salts include both organic acids, e.g., acetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, malic acid, malonic acid, succinic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid, and the like, as well as inorganic acids, e.g., hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like.
  • In other embodiments, an acid addition salt is reconverted to the free base by treatment with a suitable base. In a further embodiment, the acid addition salts of the proton pump inhibitors are halide salts, which are prepared using hydrochloric or hydrobromic acids. In still other embodiments, the basic salts are alkali metal salts, e.g., sodium salt.
  • Salt forms of proton pump inhibiting agents include, but are not limited to: a sodium salt form such as esomeprazole sodium, omeprazole sodium, rabeprazole sodium, pantoprazole sodium; or a magnesium salt form such as esomeprazole magnesium or omeprazole magnesium, described in U.S. Pat. No. 5,900,424; a calcium salt form; or a potassium salt form such as the potassium salt of esomeprazole, described in U.S. patent application Ser. No. 02/0198239 and U.S. Pat. No. 6,511,996. Other salts of esomeprazole are described in U.S. Pat. No. 4,738,974 and U.S. Pat. No. 6,369,085. Salt forms of pantoprazole and lansoprazole are discussed in U.S. Pat. Nos. 4,758,579 and 4,628,098, respectively.
  • In one embodiment, preparation of esters involves functionalizing hydroxyl and/or carboxyl groups that may be present within the molecular structure of the drug. In one embodiment, the esters are acyl-substituted derivatives of free alcohol groups, e.g., moieties derived from carboxylic acids of the formula RCOOR1 where R1 is a lower alkyl group. Esters can be reconverted to the free acids, if desired, by using conventional procedures such as hydrogenolysis or hydrolysis.
  • “Amides” may be prepared using techniques known to those skilled in the art or described in the pertinent literature. For example, amides may be prepared from esters, using suitable amine reactants, or they may be prepared from an anhydride or an acid chloride by reaction with an amine group such as ammonia or a lower alkyl amine.
  • “Tautomers” of substituted bicyclic aryl-imidazoles include, e.g., tautomers of omeprazole such as those described in U.S. Pat. Nos. 6,262,085; 6,262,086; 6,268,385; 6,312,723; 6,316,020; 6,326,384; 6,369,087; and 6,444,689; and U.S. Patent Publication No. 02/0156103.
  • An exemplary “isomer” of a substituted bicyclic aryl-imidazole is the isomer of omeprazole including but not limited to isomers described in: Oishi et al., Acta Cryst. (1989), C45, 1921-1923; U.S. Pat. No. 6,150,380; U.S. Patent Publication No. 02/0156284; and PCT Publication No. WO 02/085889.
  • Exemplary “polymorphs” include, but are not limited to, those described in PCT Publication No. WO 92/08716, and U.S. Pat. Nos. 4,045,563; 4,182,766; 4,508,905; 4,628,098; 4,636,499; 4,689,333; 4,758,579; 4,783,974; 4,786,505; 4,808,596; 4,853,230; 5,026,560; 5,013,743; 5,035,899; 5,045,321; 5,045,552; 5,093,132; 5,093,342; 5,433,959; 5,464,632; 5,536,735; 5,576,025; 5,599,794; 5,629,305; 5,639,478; 5,690,960; 5,703,110; 5,705,517; 5,714,504; 5,731,006; 5,879,708; 5,900,424; 5,948,773; 5,997,903; 6,017,560; 6,123,962; 6,147,103; 6,150,380; 6,166,213; 6,191,148; 5,187,340; 6,268,385; 6,262,086; 6,262,085; 6,296,875; 6,316,020; 6,328,994; 6,326,384; 6,369,085; 6,369,087; 6,380,234; 6,428,810; 6,444,689; and 6,462,0577.
  • Micronized Proton Pump Inhibitor
  • Particle size of the proton pump inhibitor can affect the solid dosage form in numerous ways. Because decreased particle size increases in surface area (S), the particle size reduction provides an increase in the rate of dissolution (dM/dt) as expressed in the Noyes-Whitney equation below:

  • dM/dt=DS/H(Cs−C)
  • M=mass of drug dissolved; t=time; D=diffusion coefficient of drug; S=effective surface area of drug particles; H=stationary layer thickness; Cs=concentration of solution at saturation; and C=concentration of solution at time t.
  • Because omeprazole, as well as other proton pump inhibitors, has poor water solubility, to aid the rapid absorption of the drug product, various embodiments of the present invention use micronized proton pump inhibitor in the microencapsulation. In some embodiments, the average particle size of at least about 90% the micronized proton pump inhibitor is less than about 200 μm, 150 μm, 100 μm, 80 μm, 60 μm, 40 μm, or less than about 35 μm, or less than about 30 μm, or less than about 25 μm, or less than about 20 μm, or less than about 15 μm, or less than about 10 μm. In other embodiments, at least 80% of the micronized proton pump inhibitor has an average particle size of less than about 200 μm, 150 μm, 100 μm, 80 μm, 60 μm, 40 μm, or less than about 35 μm, or less than about 30 μm, or less than about 25 μm, or less than about 20 μm, or less than about 15 μm, or less than about 10 μm. In still other embodiments, at least 70% of the micronized proton pump inhibitor has an average particle size of less than about 200 μm, 150 μm, 100 μm, 80 μm, 60 μm, 40 μm, or less than about 35 μm, or less than about 30 μm, or less than about 25 μm, or less than about 20 μm, or less than about 15 μm, or less than about 10 μm.
  • In other embodiments, the micronized proton pump inhibitor is of a size that allows greater than 50% of the proton pump inhibitor to be released within about 1 hour or 50 minutes, 40 minutes, 30 minutes, 20 minutes or 10 minutes of dissolution testing. In other embodiments, the micronized proton pump inhibitor allows greater than 25% of the proton pump inhibitor to be released within about 45 minutes, 30 minutes, or 15 minutes of dissolution testing.
  • Compositions are provided wherein the micronized proton pump inhibitor is of a size which allows greater than 75% of the proton pump inhibitor to be released within about 1.5 hours, or within about 1.25 hours, or within about 1 hour, or within about 50 minutes, or within about 40 minutes, or within about 30 minutes, or within about 20 minutes, or within about 10 minutes, or within about 5 minutes of dissolution testing. In another embodiment of the invention, the micronized proton pump inhibitor is of a size which allows greater than 90% of the proton pump inhibitor to be released within about 1.5 hours, or within about 1.25 hours, or within about 1 hour, or within about 50 minutes, or within about 40 minutes, or within about 30 minutes, or within about 20 minutes, or within about 10 minutes, or within about 5 minutes of dissolution testing. See U.S. application Ser. No. 10/893,092, filed Jul. 16, 2004, which claims priority to U.S. Provisional Application No. 60/488,324 filed Jul. 18, 2003, and any subsequent application claiming priority to these applications, all of which are incorporated by reference in their entirety.
  • Particle Size of Ingredients
  • The particle size of the proton pump inhibitor, antacid and excipients is an important factor which can effect bioavailability, blend uniformity, segregation, and flow properties. In general, smaller particle sizes of a drug increases the bioabsorption rate of the drug with substantially poor water solubility by increasing the surface area. The particle size of the drug and excipients can also affect the suspension properties of the pharmaceutical formulation. For example, smaller particles are less likely to settle and therefore form better suspensions.
  • In various embodiments, the average particle size of the dry powder (which can be administered directly, as a powder for suspension, or used in a solid dosage form) is less than about 500 microns in diameter, or less than about 450 microns in diameter, or less than about 400 microns in diameter, or less than about 350 microns in diameter, or less than about 300 microns in diameter, or less than about 250 microns in diameter, or less than about 200 microns in diameter, or less than about 150 microns in diameter, or less than about 100 microns in diameter, or less than about 75 microns in diameter, or less than about 50 microns in diameter, or less than about 25 microns in diameter, or less than about 15 microns in diameter. In other embodiments, the average particle size of the aggregates is between about 25 microns in diameter to about 300 microns in diameter. In still other embodiments, the average particle size of the aggregates is between about 25 microns in diameter to about 150 microns in diameter. And, in still further embodiments, the average particle size of the aggregates is between about 25 microns in diameter to about 100 microns in diameter. The term “average particle size” is intended to describe the average diameter of the particles and/or agglomerates used in the pharmaceutical formulation.
  • In another embodiment, the average particle size of the insoluble excipients is between about 5 μm to about 500 μm, or less than about 400 μm, or less than about 300 μm, or less than about 200 μm, or less than about 150 μm, or less than about 100 μm, or less than about 90 μm, or less than about 80 μm, or less than about 70 μm, or less than about 60 μm, or less than about 50 μm, or less than about 40 μm, or less than about 30 μm, or less than about 25 μm, or less than about 20 μm, or less than about 15 μm, or less than about 10 μm, or less than about 5 μm.
  • In other embodiments of the present invention, at least about 80% of the particles have a particle size of less than about 300 μm, or less than about 250 μm, or less than about 200 μm, or less than about 150 μm, or less than about 100 μm, or less than about 500 μm. In another embodiment, at least about 85% of the dry powder particles have a particle size of less than about 300 μm, or less than about 250 μm, or less than about 200 μm, or less than about 150 μm, or less than about 100 μm, or less than about 50 μm. In still other embodiments of the present invention, at least about 90% of the dry powder particles have a particle size of less than about 300 μm, or less than about 250 μm, or less than about 200 μm, or less than about 150 μm, or less than about 100 μm, or less than about 50 μm. In yet another embodiment, at least about 95% of the dry powder particles have a particle size of less than about 300 μm, or less than about 250 μm, or less than about 200 μm, or less than about 150 μm, or less than about 100 μm, or less than about 50 μm.
  • In other embodiments, the average particle size of the insoluble material is between about 5 μm to about 250 μm in diameter. In other embodiments, the average particle size of the insoluble excipients is between about 5 μm to about 100 μm, or between about 5 μm to about 80 μm, or between about 5 μm to about 50 μm in diameter.
  • In another embodiment, the particle size of other excipients is chosen to be about the same as the particle size of the antacid. In yet another embodiment, the particle size of the insoluble excipients is chosen to be about the same as the particle size of the proton pump inhibitor.
  • Several factors can be considered in choosing both the proper excipient and its quantity. For example, the excipient should be pharmaceutically acceptable. Also, in some examples, rapid dissolution and neutralization of gastric acid to maintain the gastric pH at about 6.5 for at least one hour. The excipients which will be in contact with the proton pump inhibitor, if any, should also be chemically compatible with the proton pump inhibitor. “Chemically compatible” is intended to mean that the material does not lead to more than 10% degradation of the proton pump inhibitor when stored at room temperature for at least about 1 year.
  • Parietal cell activators are administered in an amount sufficient to produce the desired stimulatory effect without causing untoward side effects to patients. In one embodiment, the parietal cell activator is administered in an amount of about 5 mg to about 2.5 grams per 20 mg dose of the proton pump inhibitor.
  • Antacids
  • The pharmaceutical composition of the invention comprises one or more antacids. A class of antacids useful in the present invention include, e.g., antacids possessing pharmacological activity as a weak base or a strong base. In one embodiment, the antacid, when formulated or delivered with an proton pump inhibiting agent, functions to substantially prevent or inhibit the acid degradation of the proton pump inhibitor by gastrointestinal fluid for a period of time, e.g., for a period of time sufficient to preserve the bioavailability of the proton pump inhibitor administered. The antacid can be delivered before, during, and/or after delivery of the Proton Pump Inhibitor. In one aspect of the present invention, the antacid includes a salt of a Group IA metal, including, e.g., a bicarbonate salt of a Group IA metal (alkali metal), a carbonate salt of a Group IA metal, an alkali earth metal antacid (Group IIA metal), an aluminum antacid, a calcium antacid, or a magnesium antacid.
  • Other antacids suitable for the present invention include, e.g., alkali metal (a Group IA metal including, but not limited to, lithium, sodium, potassium, rubidium, cesium, and francium) or alkaline earth metal (Group IIA metal including, but not limited to, beryllium, magnesium, calcium, strontium, barium, radium) carbonates, phosphates, bicarbonates, citrates, borates, acetates, phthalates, tartrate, succinates and the like, such as sodium or potassium phosphate, citrate, borate, acetate, bicarbonate and carbonate.
  • In various embodiments, an antacid includes, e.g., an amino acid, an alkali salt of an amino acid, aluminum hydroxide, aluminum hydroxide/magnesium carbonate/calcium carbonate co-precipitate, aluminum magnesium hydroxide, aluminum hydroxide/magnesium hydroxide co-precipitate, aluminum hydroxide/sodium bicarbonate co-precipitate, aluminum glycinate, calcium acetate, calcium bicarbonate, calcium borate, calcium carbonate, calcium citrate, calcium gluconate, calcium glycerophosphate, calcium hydroxide, calcium lactate, calcium phthalate, calcium phosphate, calcium succinate, calcium tartrate, dibasic sodium phosphate, dipotassium hydrogen phosphate, dipotassium phosphate, disodium hydrogen phosphate, disodium succinate, dry aluminum hydroxide gel, L-arginine, magnesium acetate, magnesium aluminate, magnesium borate, magnesium bicarbonate, magnesium carbonate, magnesium citrate, magnesium gluconate, magnesium hydroxide, magnesium lactate, magnesium metasilicate aluminate, magnesium oxide, magnesium phthalate, magnesium phosphate, magnesium silicate, magnesium succinate, magnesium tartrate, potassium acetate, potassium carbonate, potassium bicarbonate, potassium borate, potassium citrate, potassium metaphosphate, potassium phthalate, potassium phosphate, potassium polyphosphate, potassium pyrophosphate, potassium succinate, potassium tartrate, sodium acetate, sodium bicarbonate, sodium borate, sodium carbonate, sodium citrate, sodium gluconate, sodium hydrogen phosphate, sodium hydroxide, sodium lactate, sodium phthalate, sodium phosphate, sodium polyphosphate, sodium pyrophosphate, sodium sesquicarbonate, sodium succinate, sodium tartrate, sodium tripolyphosphate, synthetic hydrotalcite, tetrapotassium pyrophosphate, tetrasodium pyrophosphate, tripotassium phosphate, trisodium phosphate, and trometamol. (Based in part upon the list provided in The Merck Index, Merck & Co. Rahway, N.J. (2001)). In addition, due to the ability of proteins or protein hydrolysates to react with stomach acids, they too can serve as antacids in the present invention. Furthermore, combinations of the above mentioned antacids can be used in the pharmaceutical formulations described herein.
  • The antacids useful in the present invention also include antacids or combinations of antacids that interact with HCl (or other acids in the environment of interest) faster than the proton pump inhibitor interacts with the same acids. When placed in a liquid phase, such as water, these antacids produce and maintain a pH greater than the pKa of the proton pump inhibitor. In various embodiments, the antacid is selected from sodium bicarbonate, sodium carbonate, calcium carbonate, magnesium oxide, magnesium hydroxide, magnesium carbonate, aluminum hydroxide, and mixtures thereof. In other embodiments, the antacid is present in the pharmaceutical formulations of the present invention in an amount greater than about 5 mEq of antacid. In other embodiments, the antacid is present in the pharmaceutical formulations of the present invention in an amount greater than about 7 mEq of antacid. In other embodiments, the antacid is present in the pharmaceutical formulations of the present invention in an amount greater than about 10 mEq of antacid. In other embodiments, the antacid is present in the pharmaceutical formulations of the present invention in an amount greater than about 15 mEq of antacid. In other embodiments, the antacid is present in the pharmaceutical formulations of the present invention in an amount greater than about 20 mEq of antacid.
  • In another embodiment, the antacid comprises sodium bicarbonate in about 0.1 mEq/mg proton pump inhibitor to about 5 mEq/mg proton pump inhibitor. In yet another embodiment, the antacid comprises a mixture of sodium bicarbonate and magnesium hydroxide, wherein the sodium bicarbonate and magnesium hydroxide are each present in about 0.1 mEq/mg proton pump inhibitor to about 5 mEq/mg proton pump inhibitor. In still another embodiment, the antacid comprises a mixture of sodium bicarbonate, calcium carbonate, and magnesium hydroxide, wherein the sodium bicarbonate, calcium carbonate, and magnesium hydroxide are each present in about 0.1 mEq/mg proton pump inhibitor to about 5 mEq/mg of the proton pump inhibitor. In another embodiment, the antacid comprises a mixture of sodium bicarbonate and magnesium oxide, wherein the sodium bicarbonate and magnesium hydroxide are each present in about 0.1 mEq/mg proton pump inhibitor to about 5 mEq/mg proton pump inhibitor.
  • In various other embodiments of the present invention, the antacid is present in an amount of about 0.1 mEq/mg to about 5 mEq/mg of the proton pump inhibitor, or about 0.5 mEq/mg to about 3 mEq/mg of the proton pump inhibitor, or about 0.6 mEq/mg to about 2.5 mEq/mg of the proton pump inhibitor, or about 0.7 mEq/mg to about 2.0 mEq/mg of the proton pump inhibitor, or about 0.8 mEq/mg to about 1.8 mEq/mg of the proton pump inhibitor, or about 1.0 mEq/mg to about 1.5 mEq/mg of the proton pump inhibitor, or at least 0.5 mEq/mg of the proton pump inhibitor.
  • In other embodiments, the antacid is present in the pharmaceutical formulations of the present invention in an amount from about 5 to about 50 mEq of antacid. In other embodiments, the antacid is present in the pharmaceutical formulations of the present invention in an amount from about 5 to about 40 mEq of antacid. In other embodiments, the antacid is present in the pharmaceutical formulations of the present invention in an amount from about 10 to about 30 mEq of antacid. In other embodiments, the antacid is present in the pharmaceutical formulations of the present invention in an amount from about 10 to about 20 mEq of antacid. In other embodiments, the antacid is present in the pharmaceutical formulations of the present invention in an amount from about 5 to about 15 mEq of antacid.
  • In another embodiment, the antacid is present in the pharmaceutical formulations of the present invention in an amount of about 0.1 mEq to about 15 mEq/mg of proton pump inhibitor, or about 0.1 mEq/mg of proton pump inhibitor, or about 0.5 mEq/mg of proton pump inhibitor, or about 1 mEq/mg of proton pump inhibitor, or about 2 mEq/mg of proton pump inhibitor, or about 2.5 mEq/mg of proton pump inhibitor, or about 3 mEq/mg of proton pump inhibitor, or about 3.5 mEq/mg of proton pump inhibitor, or about 4 mEq/mg of proton pump inhibitor, or about 4.5 mEq/mg of proton pump inhibitor, or about 5 mEq/mg of proton pump inhibitor, or about 6 mEq/mg of proton pump inhibitor, or about 7 mEq/mg of proton pump inhibitor, or about 8 mEq/mg of proton pump inhibitor, or about 9 mEq/mg of proton pump inhibitor, or about 10 mEq/mg of proton pump inhibitor, or about 11 mEq/mg of proton pump inhibitor, or about 12 mEq/mg of proton pump inhibitor, or about 13 mEq/mg of proton pump inhibitor, or about 14 mEq/mg of proton pump inhibitor, or about 15 mEq/mg of proton pump inhibitor.
  • In one embodiment, the antacid is present in the pharmaceutical formulations of the present invention in an amount of about 1 mEq to about 160 mEq per dose, or about 1 mEq, or about 5 mEq, or about 10 mEq, or about 15 mEq, or about 20 mEq, or about 25 mEq, or about 30 mEq, or about 35 mEq, or about 40 mEq, or about 45 mEq, or about 50 mEq, or about 60 mEq, or about 70 mEq, or about 80 mEq, or about 90 mEq, or about 100 mEq, or about 110 mEq, or about 120 mEq, or about 130 mEq, or about 140 mEq, or about 150 mEq, or about 160 mEq per dose.
  • In another embodiment, the antacid is present in an amount of more than about 5 times, or more than about 10 times, or more than about 20 times, or more than about 30 times, or more than about 40 times, or more than about 50 times, or more than about 60 times, or more than about 70 times, or more than about 80 times, or more than about 90 times, or more than about 100 times the amount of the proton pump inhibiting agent on a weight to weight basis in the composition.
  • In another embodiment, the amount of antacid present in the pharmaceutical formulation is between 200 and 3500 mg. In other embodiments, the amount of antacid present in the pharmaceutical formulation is about 200 mgs, or about 300 mgs, or about 400 mgs, or about 500 mgs, or about 600 mgs, or about 700 mgs, or about 800 mgs, or about 900 mgs, or about 1000 mgs, or about 1100 mgs, or about 1200 mgs, or about 1300 mgs, or about 1400 mgs, or about 1500 mgs, or about 1600 mgs, or about 1700 mgs, or about 1800 mgs, or about 1900 mgs, or about 2000 mgs, or about 2100 mgs, or about 2200 mgs, or about 2300 mgs, or about 2400 mgs, or about 2500 mgs, or about 2600 mgs, or about 2700 mgs, or about 2800 mgs, or about 2900 mgs, or about 3000 mgs, or about 3200 mgs, or about 3500 mgs.
  • In some embodiments, if the at least one buffering agent is a combination of two or more buffering agents, the combination comprises at least two non-amino acid buffering agents, wherein the combination of at least two non-amino acid buffering agents comprises substantially no aluminum hydroxide-sodium bicarbonate co-precipitate. In other embodiments, if the pharmaceutical composition comprises an amino acid buffering agent, the total amount of buffering agent present in the pharmaceutical composition is less than about 5 mEq, or less than about 4 mEq, or less than about 3 mEq. The phrase “amino acid buffering agent” as used herein includes amino acids, amino acid salts, and amino acid alkali salts. including: glycine, alanine, threonine, isoleucine, valine, phenylalanine, glutamic acid, asparagininic acid, lysine, aluminum glycinate and/or lysine glutamic acid salt, glycine hydrochloride, L-alanine, DL-alanine, L-threonine, DL-threonine, L-isoleucine, L-valine, L-phenylalanine, L-glutamic acid, L-glutamic acid hydrochloride, L-glutamic acid sodium salt, L-asparaginic acid, L-asparaginic acid sodium salt, L-lysine and L-lysine-L-glutamic acid salt. The term “non-amino acid buffering agent” herein includes buffering agents as defined hereinabove but does not include amino acid buffering agents.
  • In other embodiments, the pharmaceutical composition comprises substantially no or no poly[phosphoryl/sulfon]-ated carbohydrate and is in the form of a solid dosage unit. In still another related embodiment, if such a composition comprises a poly[phosphoryl/sulfon]-ated carbohydrate (e.g. sucralfate or sucrose octasulfate), the weight ratio of poly[phosphoryl/sulfon]-ated carbohydrate to buffering agent is less than 1:5 (0.2), less than 1:10 (0.1) or less than 1:20 (0.05). Alternatively, the poly[phosphoryl/sulfon]-ated carbohydrate is present in the composition, if at all, in an amount less than 50 mg, less than 25 mg, less than 10 mg or less than 5 mg.
  • Also provided herein are pharmaceutical formulations comprising at least one soluble antacid. For example, in one embodiment, the antacid is sodium bicarbonate and is present in about 0.1 mEq/mg proton pump inhibitor to about 5 mEq/mg proton pump inhibitor. In another embodiment, the antacid is a mixture of sodium bicarbonate and magnesium hydroxide, wherein the sodium bicarbonate and magnesium hydroxide are each present in about 0.1 mEq/mg proton pump inhibitor to about 5 mEq/mg proton pump inhibitor. Yet in another embodiment, the antacid is a mixture of sodium bicarbonate and magnesium oxide, wherein the sodium bicarbonate and magnesium oxide are each present in about 0.1 mEq/mg proton pump inhibitor to about 5 mEq/mg proton pump inhibitor. The term “soluble antacid” as used herein refers to an antacid that has a solubility of at least 500 mg/mL, or 300 mg/mL, or 200 mg/mL, or 100 mg/mL, or 50 mg/mL in the gastrointestinal fluid. In some embodiments, the soluble antacid is sodium bicarbonate.
  • Particle Size of Antacids
  • Particle size of the buffer, especially that an insoluble buffer can affect the onset of in-vivo neutralization of the stomach acid. Since decreased particle size increases in surface area, the particle size reduction provides an increase in the rate of acid neutralization, leading to superior protection of PPI from gastric acid degradation. On the other hand, extremely fine particle size of buffer will result in the powder mixture that is difficult to manufacture in commercial scale due to their poor flow and difficulties in processing (i.e., compression and encapsulation).
  • In some embodiments of the present invention, the antacid is a specific particle size. For example, in some embodiments, the average particle size is no greater than 20 μm, or no greater than 30 μm, or no greater than 40 μm, or no greater than 50 μm, or no greater than 60 μm, or no greater than 70 μm, or no greater than 80 μm, or no greater than 90 μm or no greater than 100 μm in diameter. In various embodiments, at least about 70% of the antacid is no greater than 20 μm, or no greater than 30 μm, or no greater than 40 μm, or no greater than 50 μm, or no greater than 60 μm, or no greater than 70 μm, or no greater than 80 μm, or no greater than 90 μm or no greater than 100 μm in diameter. In other embodiments, at least about 85% of the antacid is no greater than 20 μm, or no greater than 30 μm, or no greater than 40 μm, or no greater than 50 μm, or no greater than 60 μm, or no greater than 70 μm, or no greater than 80 μm, or no greater than 90 μm or no greater than 100 μm in diameter.
  • In various embodiments of the present invention, the antacid is micronized. In some embodiments, particle size of at least 90% of antacid (D90) is less than about 300 μm, or less than about 250 μm, or less than about 200 μm, or less than about 150 μm, or less than about 100 μm. In other embodiments, at least 75% of the antacid (D75) has particle size of less than about 300 μm, or less than about 250 μm, or less than about 200 μm, or less than about 150 μm, or less than about 100 μm. In still other embodiments, at least 50% of the antacid (D50) has particle size of less than about 300 μm, or less than about 250 μm, or less than about 200 μm, or less than about 150 μm, or less than about 100 μm.
  • Spray dried antacid can also facilitate the speed of neutralization by fast reacting with acid upon contact. Sprayed dried antacid typically has spherical particle shape which aids with achieving homogeneous blend during manufacturing process. In one embodiment the antacid is spray dried with at least 15% of coating material such as maltodextrin or starch. In still other embodiment the antacid is spray dried with at least 10% of coating material such as maltodextrin or starch. Yet another embodiment the antacid is spray dried with at least 5% of coating material such as maltodextrin or starch. In still other embodiments, the antacid is spray dried with between about 1% to about 10% of a coating material. In yet other embodiments, the antacid is spray dried with about 5% of a coating material such as maltodextrin or starch.
  • Shelf-Life Enhancing Materials
  • Materials useful for enhancing the shelf-life of the pharmaceutical formulations of the present invention include materials compatible with the proton pump inhibitor of the pharmaceutical formulations which sufficiently isolate the proton pump inhibitor from other non-compatible excipients. Materials compatible with the proton pump inhibitors of the present invention are those that enhance the shelf-life of the proton pump inhibitor, i.e., by slowing or stopping degradation of the proton pump inhibitor.
  • A pharmaceutical formulation of the present invention may have an enhanced shelf-life stability if, e.g., the microencapsulated proton pump inhibitor has less than about 0.5% degradation after one month of storage at room temperature, or less than about 1% degradation after one month at room temperature, or less than about 1.5% degradation after one month of storage at room temperature, or less than about 2% degradation after one month storage at room temperature, or less than about 2.5% degradation after one month of storage at room temperature, or less than about 3% degradation after one month of storage at room temperature.
  • In other embodiments, a pharmaceutical formulation of the present invention may have an enhanced shelf-life stability if the pharmaceutical formulation contains less than about 5% total impurities after about 3 years of storage, or after about 2.5 years of storage, or about 2 years of storage, or about 1.5 years of storage, or about 1 year of storage, or after 11 months of storage, or after 10 months of storage, or after 9 months of storage, or after 8 months of storage, or after 7 months of storage, or after 6 months of storage, or after 5 months of storage, or after 4 months of storage, or after 3 months of storage, or after 2 months of storage, or after 1 month of storage.
  • In further embodiments, pharmaceutical formulations of the present invention may have enhanced shelf-life stability if the pharmaceutical formulation contains less degradation of the proton pump inhibitor than proton pump inhibitor in the same formulation which is not microencapsulated or dry coated, or “bare”. For example, if bare proton pump inhibitor in the pharmaceutical formulation degrades at room temperature by more than about 2% after one month of storage and the microencapsulated or dry coated material degrades at room temperature by less than about 2% after one month of storage, then the proton pump inhibitor has been microencapsulated or dry coated with a compatible material that enhances the shelf-life of the pharmaceutical formulation.
  • In some embodiments, the material useful for enhancing the shelf-life of the pharmaceutical formulations increases the shelf-life stability of the pharmaceutical formulation for at least about 5 days at room temperature, or at least about 10 days at room temperature, or at least about 15 days at room temperature, or at least about 20 days at room temperature, or at least about 25 days at room temperature, or at least about 30 days at room temperature or at least about 2 months at room temperature, or at least about 3 months at room temperature, or at least about 4 months at room temperature, or at least about 5 months at room temperature, or at least about 6 months at room temperature, or at least about 7 months at room temperature, or at least about 8 months at room temperature, or at least about 9 months at room temperature, or at least about 10 months at room temperature, or at least about 11 months at room temperature, or at least about one year at room temperature, or at least about 1.5 years at room temperature, or at least about 2 years at room temperature, or at least about 2.5 years at room temperature, or about 3 years at room temperature.
  • Microencapsulating Materials
  • Exemplary microencapsulation materials useful for enhancing the shelf-life of pharmaceutical formulations comprising a proton pump inhibitor include, e.g., cellulose hydroxypropyl ethers (HPC) such as EF Klucel®, Nisso HPC and PrimaFlo HP22; low-substituted hydroxypropyl ethers (L-HPC); cellulose hydroxypropyl methyl ethers (HPMC) such as Seppifilm-LC, Pharmacoat®, Metolose SR, Opadry YS, PrimaFlo, MP3295A, Benecel MP824, and Benecel MP843; methylcellulose polymers such as Methocel® and Metolose®; Ethylcelluloses (EC) and mixtures thereof such as E461, Ethocel®, Aqualon®-EC, Surelease; Polyvinyl alcohol (PVA) such as Opadry AMB; hydroxyethylcelluloses such as Natrosol®; carboxymethylcelluloses and salts of carboxymethylcelluloses (CMC) such as Aqualon®-CMC; polyvinyl alcohol and polyethylene glycol co-polymers such as Kollicoat IR®; monoglycerides (Myverol), triglycerides (KLX), polyethylene glycols, modified food starch, acrylic polymers and mixtures of acrylic polymers with cellulose ethers such as Eudragit® EPO, Eudragit® RD100, and Eudragit® E100; cellulose acetate phthalate; sepifilms such as mixtures of HPMC and stearic acid, cyclodextrins, and mixtures of these materials. In other embodiments, the microencapsulation material is selected from hydroxypropylcellulose and cellulose ethers. In still other embodiments, the microencapsulation material is selected from Klucel EF, Klucel EXF, Methocel E5, Methocel E15, and Methocel A15. In other embodiments, the material that enhances the shelf-life has a viscosity of 100-800 cps at 10% solution; or a viscosity of 200-600 cps at 10% solution; or a viscosity of 300-400 cps at 10% solution.
  • In various embodiments, a buffering agent such as sodium bicarbonate is incorporated into the microencapsulation material. In other embodiments, an antioxidant such as BHT or BHA is incorporated into the microencapsulation material. In still other embodiments, plasticizers such as polyethylene glycols, e.g., PEG 300, PEG 400, PEG 600, PEG 1450, PEG 3350, and PEG 800, stearic acid, propylene glycol, oleic acid, and triacetin are incorporated into the microencapsulation material. In other embodiments, the microencapsulating material useful for enhancing the shelf-life of the pharmaceutical formulations is from the USP or the National Formulary (NF).
  • In further embodiments, one or more other compatible materials are present in the microencapsulation material. Exemplary materials include, e.g., parietal cell activators, organic solvents, erosion facilitators, diffusion facilitators, anti-adherents, anti-foaming agents, antioxidants, sweetening agents, flavoring agents, and carrier materials such as binders, suspending agents, disintegration agents, filing agents, surfactants, solubilizers, stabilizers, lubricants, wetting agents, and diluents.
  • In some embodiments of the present invention, the final formulation of the pharmaceutical formulation will be in the form of a tablet and at least about 50%, or at least about 55%, or at least about 60%, or at least about 65%, or at least about 70%, or at least about 75%, or at least about 80%, or at least about 85% or at least about 90%, or at least about 92%, or at least about 95%, or at least about 98%, or at least about 99% of the microspheres survive the tableting process, wherein microspheres that have survived the tableting process are those which provide the desired properties described herein.
  • In other embodiments, the final formulation of the pharmaceutical formulation is in the form of a powder for oral suspension and the microencapsulation material surrounding the proton pump inhibitor will sufficiently dissolve in water, with or without stirring, in less than 1 hour, or less than 50 minutes, or less than 40 minutes, or less than 30 minutes, or less than 25 minutes, or less than 20 minutes, or less than 15 minutes, or less than 10 minutes or less than 5 minutes, or less than 1 minute. Sufficiently dissolves means that at least about 50% of the encapsulation material has dissolved.
  • In various embodiments the microencapsulating material useful for enhancing the shelf-life of the pharmaceutical formulation sufficiently disintegrates to release the proton pump inhibitor into the gastrointestinal fluid of the stomach within less than about 1.5 hours, or within about 10 minutes, or within about 20 minutes, or within about 30 minutes, or within about or within about 40 minutes, or within about 50 minutes, or within about 1 hour, or within about 1.25 hours, or within about 1.5 hours after exposure to the gastrointestinal fluid. Sufficiently disintegrates means that at least about 50% of the microencapsulation material has dissolved.
  • In various embodiments, the average particle sizes of the microencapsulated drugs range from submicron to less than about 1,000 microns in diameter, or less than about 900 microns in diameter, or less than about 800 microns in diameter, or less than about 700 microns in diameter, or less than about 600 microns in diameter, or less than about 500 microns in diameter, or less than about 450 microns in diameter, or less than about 400 microns in diameter, or less than about 350 microns in diameter, or less than about 300 microns in diameter, or less than about 250 microns in diameter, or less than about 200 microns in diameter, or less than about 150 microns in diameter, or less than about 100 microns in diameter, or less than about 75 microns in diameter, or less than about 50 microns in diameter, or less than about 25 microns in diameter, or less than about 15 microns in diameter. In other embodiments, the average particle size of the aggregates is between about 25 microns in diameter to about 300 microns in diameter. In still other embodiments, the average particle size of the aggregates is between about 100 microns in diameter to about 200 microns in diameter. And in still further embodiments, the average particle size of the aggregates is between about 25 microns in diameter to about 100 microns in diameter. The term “average particle size” is intended to describe the average diameter of the particles and/or agglomerates used in the pharmaceutical formulation.
  • Taste-Masking Materials
  • Proton pump inhibitors are inherently bitter tasting and in one embodiment of the present invention, these bitter proton pump inhibitors are microencapsulated with a taste-masking material. Materials useful Tor masking the taste of pharmaceutical formulations include those materials capable of microencapsulating the proton pump inhibitor, thereby protecting the senses from its bitter taste. Taste-masking materials of the present invention provide superior pharmaceutical formulations by e.g., creating a more palatable pharmaceutical formulation as compared to pharmaceutical formulations and/or by creating a dosage form requiring less of the traditional flavoring or tastemasking agents.
  • The “flavor leadership” criteria used to develop a palatable product include (1) immediate impact of identifying flavor, (2) rapid development of balanced, full flavor, (3) compatible mouth feel factors, (4) no “off” flavors, and (5) short aftertaste. See, e.g., Worthington, A Matter of Taste, Pharmaceutical Executive (April 2001). The pharmaceutical formulations of the present invention improve upon one or more of these criteria.
  • There are a number of known methods to determine the effect of a taste-masking material such as discrimination tests for testing differences between samples and for ranking a series of samples in order of a specific characteristic; scaling tests used for scoring the specific product attributes such as flavor and appearance; expert tasters used to both quantitatively and qualitatively evaluate a specific sample; affective tests for either measuring the response between two products, measuring the degree of like or dislike of a product or specific attribute, or determine the appropriateness of a specific attribute; and descriptive methods used in flavor profiling to provide objective description of a product are all methods used in the field.
  • Different sensory qualities of a pharmaceutical formulation such as aroma, flavor, character notes, and aftertaste can be measured using tests know in the art. See, e.g., Roy et al., Modifying Bitterness: Mechanism, Ingredients, and Applications (1997). For example, aftertaste of a product can be measured by using a time vs. intensity sensory measurement. And recently, modem assays have been developed to alert a processor of formulations to the bitter taste of certain substances. Using information known to one of ordinary skill in the art, one would readily be able to determine whether one or more sensory qualities of a pharmaceutical formulation of the present invention have been improved by the use of the taste-masking material.
  • Taste of a pharmaceutical formulation is important for both increasing patient compliance as well as for competing with other marketed products used for similar diseases, conditions and disorders. Taste, especially bitterness, is particularly important in pharmaceutical formulations for children since, because they cannot weigh the positive benefit of getting better against the immediate negative impact of the bitter taste in their mouth, they are more likely to refuse a drug that tastes bad. Thus, for pharmaceutical formulations for children, it becomes even more important to mask the bitter taste.
  • Microencapsulation of the proton pump inhibitor can (1) lower the amount of flavoring agents necessary to create a palatable product and/or (2) mask the bitter taste of the proton pump inhibitor by separating the drug from the taste receptors.
  • Taste-masking materials include, e.g., cellulose hydroxypropyl ethers (HPC) such as Klucel®, Nisswo HPC and PrimaFlo HP22; low-substituted hydroxypropyl ethers (L-HPC); cellulose hydroxypropyl methyl ethers (HPMC) such as Seppifilm-LC, Pharmacoat®, Metolose SR, Opadry YS, PrimaFlo, MP3295A, Benecel MP824, and Benecel MP843; methylcellulose polymers such as Methocel® and Metolose®; Ethylcelluloses (EC) and mixtures thereof such as E461, Ethocel®, Aqualon®-EC, Surelease; Polyvinyl alcohol (PVA) such as Opadry AMB; hydroxyethylcelluloses such as Natrosol®; carboxymethylcelluloses and salts of carboxymethylcelluloses (CMC) such as Aualon®-CMC; polyvinyl alcohol and polyethylene glycol co-polymers such as Kollicoat IR®; monoglycerides (Myverol), triglycerides (KLX), polyethylene glycols, modified food starch, acrylic polymers and mixtures of acrylic polymers with cellulose ethers such as Eudragit® EPO, Eudragit® RD100, and Eudragit® E100; cellulose acetate phthalate; sepifilms such as mixtures of HPMC and stearic acid, cyclodextrins, and mixtures of these materials.
  • In other embodiments of the present invention, additional taste-masking materials contemplated are those described in U.S. Pat. Nos. 4,851,226, 5,075,114, and 5,876,759. For further examples of taste-masking materials, see, e.g., Remington: The Science and Practice of Pharmacy, Nineteenth Ed. (Easton, Pa.: Mack Publishing Company, 1995); Hoover, John E., Remington's Pharmaceutical Sciences (Mack Publishing Co., Easton, Pa. 1975); Liberman, H. A. and Lachman, L., Eds., Pharmaceutical Dosage Forms (Marcel Decker, New York, N.Y., 1980); and Pharmaceutical Dosage Forms and Drug Delivery Systems, Seventh Ed. (Lippincott Williams & Wilkins 1999).
  • In various embodiments, a pH modifier such as sodium carbonate or sodium bicarbonate is incorporated into the microencapsulation material. In other embodiments, an antioxidant such as BHT or BHA is incorporated into the microencapsulation material. In yet another embodiment, sucrose or sucralose is incorporated into the taste masking material. In still other embodiments, plasticizers such as polyethylene glycol and/or stearic acid are incorporated into the microencapsulation material.
  • In further embodiments, one or more other compatible materials are present in the microencapsulation material. Exemplary materials include, e.g., parietal cell activators, organic solvents, erosion facilitators, diffusion facilitators, anti-adherents, anti-foaming agents, antioxidants, flavoring agents, and carrier materials such as binders, suspending agents, disintegration agents, filing agents, surfactants, solubilizers, stabilizers, lubricants, wetting agents, diluents.
  • In addition to microencapsulating the proton pump inhibitors with a taste-masking material or a material that enhances the shelf-life of the formulation as described herein, the pharmaceutical formulations of the present invention may also comprise one or more flavoring agents.
  • “Flavoring agents” or “sweeteners” useful in the pharmaceutical formulations of the present invention include, e.g., acacia syrup, acesulfame K, alitame, anise, apple, aspartame, banana, Bavarian cream, berry, black currant, butterscotch, calcium citrate, camphor, caramel, cherry, cherry cream, chocolate, cinnamon, bubble gum, citrus, citrus punch, citrus cream, cotton candy, cocoa, cola, cool cherry, cool citrus, cyclamate, cylamate, dextrose, eucalyptus, eugenol, fructose, fruit punch, ginger, glycyrrhetinate, glycyrrhiza (licorice) syrup, grape, grapefruit, honey, isomalt, lemon, lime, lemon cream, monoammonium glyrrhizinate (MagnaSweet®), maltol, mannitol, maple, marshmallow, menthol, mint cream, mixed berry, neohesperidine DC, neotame, orange, pear, peach, peppermint, peppermint cream, Prosweet® Powder, raspberry, root beer, rum, saccharin, safrole, sorbitol, spearmint, spearmint cream, strawberry, strawberry cream, stevia, sucralose, sucrose, sodium saccharin, saccharin, aspartame, neotame, acesulfame potassium, mannitol, talin, xylitol, sucralose, sorbitol, swiss cream, tagatose, tangerine, thaumatin, tutti fruitti, vanilla, walnut, watermelon, wild cherry, wintergreen, xylitol, or any combination of these flavoring ingredients, e.g., anise-menthol, cherry-anise, cinnamon-orange, cherry-cinnamon, chocolate-mint, honey-lemon, lemon-lime, lemon-mint, menthol-eucalyptus, orange-cream, vanilla-mint, and mixtures thereof. In other embodiments, sodium chloride is incorporated into the pharmaceutical formulation.
  • Based on the proton pump inhibitor, antacid, and excipients, as well as the amounts of each one, one of skilled in the art would be able to determine the best combination of flavors to provide the optimally flavored product for consumer demand and compliance. See, e.g., Roy et al., Modifying Bitterness: Mechanism, Ingredients, and Applications (1997).
  • In one embodiment, one or more flavoring agents are mixed with the taste-masking material prior to microencapsulating the proton pump inhibitor and, as such, are part of the taste-masking material. In other embodiments, the flavoring agent is mixed with the non-compatible excipients during the formulation process and is therefore not in contact with the proton pump inhibitor, and not part of the microencapsulation material.
  • In another embodiment, an antacid, such as sodium bicarbonate, is also microencapsulated with one or more taste-masking materials.
  • In another embodiment, the weight fraction of the taste masking material is, e.g., about 98% or less, about 95% or less, about 90% or less, about 85% or less, about 80% or less, about 75% or less, about 70% or less, about 65% or less, about 60% or less, about 55% or less, about 50% or less, about 45% or less, about 40% or less, about 35% or less, about 30% or less, about 25% or less, about 20% or less, about 15% or less, about 10% or less, about 5% or less, about 2%, or about 1% or less of the total weight of the pharmaceutical composition.
  • In other embodiments of the present invention, the amount of flavoring agent necessary to create a palatable product, as compared to a pharmaceutical formulation comprising non-microencapsulated proton pump inhibitor, is decreased by 5% or less, or by 5% to 10%, or by 10% to 20%, or by 20% to 30%, or by 30% to 40%, or by 40% to 50%, or by 50% to 60%, or by 60% to 70%, or by 70% to 80%, or by 80% to 90%, or by 90% to 95%, or by greater than 95%. In still other embodiments, no flavoring agent is necessary to create a more palatable pharmaceutical formulation as compared to a similar pharmaceutical formulation comprising non-microencapsulated proton pump inhibitor.
  • In various embodiments of the invention, the total amount of flavoring agent present in the pharmaceutical formulation is less than 20 grams, or less than 15 grams, or less than 10 grams, or less than 8 grams, or less than 5 grams, or less than 4 grams, or less than 3.5 grams, or less than 3 grams, or less than 2.5 grams or less than 2 grams, or less than 1.5 grams, or less than 1 gram, or less than 500 mg, or less than 250 mg, or less than 150 mg, or less than 100 mg, or less than 50 mg.
  • In particular embodiments, the formulation comprises a microencapsulated omeprazole having approximately 37% omeprazole, the remainder being the immediate release coating designed to protect the micronized omeprazole from degradation by flavor components and other excipients. The shelf life of a chewable product is thus improved. Additionally the flavor of the product can be enhanced, as the immediate release coating provides protection against a wide variety of acidic excipients.
  • Methods of Microencapsulation
  • The proton pump inhibitor may be microencapsulated by methods known by one of ordinary skill in the art. Such known methods include, e.g., spray drying processes, spinning disk processes, hot melt processes, spray chilling methods, fluidized bed, electrostatic deposition, centrifugal extrusion, rotational suspension separation, polymerization at liquid-gas or solid-gas interface, pressure extrusion, or spraying solvent extraction bath. In addition to these, several chemical techniques, e.g., complex coacervation, solvent evaporation, polymer-polymer incompatibility, interfacial polymerization in liquid media, in situ polymerization, in-liquid drying, and desolvation in liquid media could also be used. Furthermore, other methods such as dry granulation (i.e., roller compaction and slugging), extrusion/spheronization, or nano particle coating may also be used.
  • The spinning disk method allows for: 1) an increased production rate due to higher feed rates and use of higher solids loading in feed solution, 2) the production of more spherical particles, 3) the production of a more even coating, and 4) limited clogging of the spray nozzle during the process.
  • Spray drying is often more readily available for scale-up to a commercial scale. In various embodiments, the material used in the spray-dry encapsulation process is emulsified or dispersed into the core material in a concentrated form, e.g., 10-60% solids. In some embodiments of the present invention, the solid loading is between about 10-20%, or between about 10-40%, or between about 40-60%. The microencapsulation material is, in one embodiment, is emulsified until about 1 to 3 μm droplets are obtained. In other embodiments, the microencapsulation material is emulsified until about 1 to 200 μm droplets are obtained, or until about 1 to 100 μm droplets are obtained. In still other embodiments, the median droplet size of the microencapsulation material is between about 1 to about 300 μm, or between about 1 to about 200 μm, or between about 50 to about 150 μm. Once a dispersion of proton pump inhibitor and encapsulation material are obtained, the emulsion is fed as droplets into the heated chamber of the pray drier. In some embodiments, the droplets are sprayed into the chamber or spun off a rotating disk. The microspheres are then dried in the heated chamber and fall to the bottom of the spray drying chamber where they are harvested.
  • Coacervation involves microencapsulation of materials such as active pharmaceutical ingredients and involves a three part process of particle or droplet formation, coacerate wall formation, and capsule isolation. This method can produce very small particle size microcapsules (10-70 microns).
  • Extrusion/spheronization is another method that involves wet massing of active pharmaceutical ingredients, followed by the extrusion of the wet mass through a perforated plate to produce short cylindrical rods. These rods are subsequently placed into a rapidly rotating spheronizer to shape the cylindrical rods into uniform spheres. The spheres are subsequently dried using a fluid bed drier and then coated with a functional coating using a fluid bed equipped with a Wurster insert and spray nozzle. This method produces smooth, uniform spheres that are ideal for receiving a functional coating. Drug loadings as high as 80% are possible (depending on drug characteristics).
  • In some embodiments of the present invention, the microspheres have irregular geometries. In other embodiments, the microspheres are aggregates of smaller particles. In various embodiments, the drug loading of the proton pump inhibitor in the microspheres is greater than 1%, greater than 2.5%, greater than 5%, greater than 10%, greater than 15%, greater than 20%, greater than 25%, greater than 30%, greater than 35%, greater than 40%, greater than 45%, greater than 50%, greater than 55%, greater than 60%, greater than 65%, greater than 70%, greater than 75%, greater than 80% weight percent of the proton pump inhibitor to the microencapsulated drug. In other embodiments, the drug loading of the proton pump inhibitor in the microspheres is between about 5-60 wt %, or about 5-50 wt-%, or about 10-40 wt-%.
  • Methods of Dry Coating
  • In addition to microencapsulation, the stability of the proton pump inhibitors used in the present invention may be increased by alternative methods such as dry coating and nano-particle coating. Dry coating involves the formation of granules of coated proton pump inhibitor which are then mixed with other components. Dry granulation is achieved by forming dense compacts which are subsequently milled to a desired particle size and then blended with other components of the pharmaceutical composition. Dry granulation and nano-particle coating can provide enhanced stability and tastemasking characteristics to active pharmaceutical by diluting and isolating such components in a granulated matrix of compatible ingredients that can enhance the shelf life of proton pump inhibitor products as well as tastemask the bitterness if sweetener or flavors are used in coating material.
  • Typical technique for dry granulation is to use slugging or roller compaction. During slugging process, the dry powders are compressed using a conventional tablet machine, or more usually, a large heavy duty rotary press. The resulting compacts or “slug” are then milled to a desired particle size. Roller compaction is an alternative gentler method, the powder mix being squeezed between two rollers to form a compressed sheet. The sheet normally is weak and brittle and breaks immediately into flakes. These flakes need gentler treatment to break them into granules, and this can be usually be achieved by screening alone. Parikh, D. M., Handbook of Pharmaceutical Granulation Technology, (Marcel Dekker ed. 1997).
  • Nano particle coating is another method that involves a nano particle deposited onto the drug core using Physical Vapour Deposition (PVD) methods. See, e.g., Lu, Y. Chen, S. C., Advanced Drug Delivery Review: Micro and Nano-Fabrication of Biodegradable Polymers for Drug Delivery 56, 1621-33 (2004) and Mark W. Horn et al., Blending of Nanoscale and Microscale in Uniform Large-Area Sculptured Thin-Film Architectures NANOTECHNOLOGY 15, 303-310 (2004). This method can coat a 10-20 micro drug core with various thickness of metal, or metal salt (e.g., SB, MgO, MgOH, CaCO3, etc.).
  • In various embodiments, the average particle sizes of the dry coated proton pump inhibitor ranges from submicron to less than about 1,000 microns in diameter, or less than about 900 microns in diameter, or less than about 800 microns in diameter, or less than about 700 microns in diameter, or less than about 600 microns in diameter, or less than about 500 microns in diameter, or less than about 450 microns in diameter, or less than about 400 microns in diameter, or less than about 350 microns in diameter, or less than about 300 microns in diameter, or less than about 250 microns in diameter, or less than about 200 microns in diameter, or less than about 150 microns in diameter, or less than about 100 microns in diameter, or less than about 75 microns in diameter, or less than about 50 microns in diameter, or less than about 25 microns in diameter, or less than about 15 microns in diameter. In other embodiments, the average particle size of the aggregates is between about 25 microns in diameter to about 300 microns in diameter. In still other embodiments, the average particle size of the aggregates is between about 100 microns in diameter to about 200 microns in diameter. And in still further embodiments, the average particle size of the aggregates is between about 25 microns in diameter to about 100 microns in diameter. The term “average particle size” is intended to describe the average diameter of the particles and/or agglomerates used in the pharmaceutical formulation.
  • In some embodiments, the dry coated proton pump inhibitor granules are less than about 2000 microns, or less than about 1500 microns, or less than about 1000 microns. In some embodiments, the average particle size of the dry coated proton pump inhibitor granules is between about 100 to about 2000 microns, or between about 100 to about 1000 microns, or between about 200 to about 800 microns, or between about 300 to about 600 microns.
  • In other embodiments, the dry coated proton pump inhibitor granules comprise antacid, binder, lubricant and/or sweeteners. In some embodiments, the antacid is sodium bicarbonate. In other embodiments, the binder is hydroxypropyl cellulose. In still other embodiments, the sweetener is sucralose and/or xylitab. In yet other embodiments, the lubricant is magnesium stearate.
  • In various embodiments, the dry coated proton pump inhibitor is combined with additional antacid. In some embodiments, the additional antacid is the same antacid as used in the material used to dry coat the proton pump inhibitor. In other embodiments, the antacid is a different antacid. In still other embodiments, the antacid is a combination of two or more antacids.
  • In yet other embodiments, one or more pharmaceutically acceptable excipients are mixed with the dry coated proton pump inhibitor to form the pharmaceutical composition. In some embodiments the additional pharmaceutical excipients include one or more flavors.
  • In further embodiments, one or more other compatible materials are present in the dry coating material. Exemplary materials include, e.g., parietal cell activators, organic solvents, erosion facilitators, diffusion facilitators, anti-adherents, anti-foaming agents, antioxidants, flavoring agents, and carrier materials such as binders, suspending agents, disintegration agents, filing agents, surfactants, solubilizers, stabilizers, lubricants, wetting agents, diluents. In some embodiments, the additional compatible materials are binders, lubricants and sweeteners.
  • In some embodiments, one or more sweeteners are incorporated into the material used to dry coat the proton pump inhibitor. In other embodiments, one or more flavoring agents are incorporated into the material used to dry coat the proton pump inhibitor. In still other embodiments, the material used to dry coat the proton pump inhibitor comprises a sweetener and/or a flavoring agent. In some embodiments, this dry coated proton pump inhibitor is then mixed with additional sweeteners and/or flavoring agents. “Flavoring agents” or “sweeteners” useful in the pharmaceutical formulations of the present invention include, e.g., acacia syrup, acesulfame K, alitame, anise, apple, aspartame, banana, Bavarian cream, berry, black currant, butterscotch, calcium citrate, camphor, caramel, cherry, cherry cream, chocolate, cinnamon, bubble gum, citrus, citrus punch, citrus cream, cotton candy, cocoa, cola, cool cherry, cool citrus, cyclamate, cylamate, dextrose, eucalyptus, eugenol, fructose, fruit punch, ginger, glycyrrhetinate, glycyrrhiza (licorice) syrup, grape, grapefruit, honey, isomalt, lemon, lime, lemon cream, monoammonium glyrrhizinate (MagnaSweet®), maltol, mannitol, maple, marshmallow, menthol, mint cream, mixed berry, neohesperidine DC, neotame, orange, pear, peach, peppermint, peppermint cream, Prosweet® Powder, raspberry, root beer, rum, saccharin, safrole, sorbitol, spearmint, spearmint cream, strawberry, strawberry cream, stevia, sucralose, sucrose, sodium saccharin, saccharin, aspartame, neotame, acesulfame potassium, mannitol, talin, xylitol, sucralose, sorbitol, swiss cream, tagatose, tangerine, thaumatin, tutti fruitti, vanilla, walnut, watermelon, wild cherry, wintergreen, xylitol, or any combination of these flavoring ingredients, e.g., anise-menthol, cherry-anise, cinnamon-orange, cherry-cinnamon, chocolate-mint, honey-lemon, lemon-lime, lemon-mint, menthol-eucalyptus, orange-cream, vanilla-mint, and mixtures thereof. In other embodiments, sodium chloride is incorporated into the pharmaceutical formulation.
  • In some embodiments, the weight percent of the proton pump inhibitor in the dry coated granules is about 2-70%. In some embodiments, the weight percent of the proton pump inhibitor in the dry coated granules is about 5-50%, or about 5-30%. In yet other embodiments, the weight percent of the proton pump inhibitor in the granules is about 5%, or about 7%, or about 10%, or about 15%, or about 20%, or about 25%, or about 30%, or about 35%, or about 40%.
  • It should be noted that the compositions and methods described herein as containing microencapsulated proton pump inhibitors can, in addition to or in the alternative, contain dry coated proton pump inhibitors.
  • Kinetic Stomach Model
  • The acid neutralizing capacity and pH profile of various antacid combinations can be evaluated by using an in-vitro stomach model. Several of these simulated dynamic models are known in the art. See, e.g., Smyth et al., Correlation of In-Vivo Methodology for Evaluation of Antacids, J. Pharm. Sci. Vol. 65, 1045 (1976); Hobert, Fordham et al., In-Vivo Evaluation of Liquid Antacids, New England Journal of Med. 288, 923 (1973); Johnson et al., The Chemical Testing of Antacids, Gut 5, 585 (1964); Clain et al., In-Vitro Neutralizing Capacity of Commercially Available Antacid Mixtures and Their Role in the Treatment of Peptic Ulcer, S. Afr. Med. J., 57, 158 (1980); Rossett et al., In-Vitro Evaluation of Efficacy of More Frequently Used Antacids with Particular Attention to Tablets, Gastroentrology, 26, 490; Decktor et al., Comparative Effects of Liquid Antacids on Esophageal and Gastric pH in Patients with Heartburn, Am J. of Therapeutics, 2, 481 (1995); Charles Fuchs, Antacids: Their Function, Formulation and Evaluation, Drug and Cosmetic Industry, 49, 692; Stewart M. Beekman, Preparation and Properties of New Gastric Antacids I, Aluminum Hydroxide-Magnesium Carbonate Dried Gels, J. Am. Pharm. Assoc., 49, 191 (1960). For example, a modified Fuch's model where the continuous influx of 0.5 mEq of acid is added to initial 5.0 mEq of acid to simulate a fasting state of stomach can be used with the present invention.
  • In various embodiments of the present invention, the antacid increases the gastric pH to at least about 3.5 for no more than about 90 minutes as measured by a simulated stomach model such as Fuch's kinetic in-vitro pH model. In other embodiments, the antacid increases the pH to at least about 3.5 for no more than about 60 minutes. In still other embodiments, the antacid increases the pH to at least about 3.5 for no more than 45 minutes. Depending on the buffer system used (i.e., type of antacid and amount) some embodiments of the present invention, the antacid increases the gastric pH to at least about 3.5 for no more than about 30 minutes as measured by a simulated stomach model such as Fuchs' kinetic in-vitro pH model. In other embodiments, the antacid increases the gastric pH to at least about 3.5 for less than about 25 minutes as measured by a simulated stomach model such as Fuch's kinetic in-vitro pH model. In yet other embodiments, the antacid increases the gastric pH to at least about 3.5 for less than about 20 minutes, or less than about 15 minutes, or less than about 10 minutes as measured by a stimulated stomach model such as Fuch's kinetic in-vitro pH model.
  • In each of these embodiments, the antacid protects at least some of the proton pump inhibitor and a therapeutically effective amount of the proton pump inhibitor is delivered to the subject.
  • Dosage
  • The proton pump inhibiting agent is administered and dosed in accordance with good medical practice, taking into account the clinical condition of the individual patient, the site and method of administration, scheduling of administration, and other factors known to medical practitioners. In human therapy, it is important to provide a dosage form that delivers the required therapeutic amount of the drug in vivo, and renders the drug bioavailable in a rapid manner. In addition to the dosage forms described herein, the dosage forms described by Phillips et al. in U.S. Pat. Nos. 5,840,737, 6,489,346, 6,699,885, and 6,645,988 are incorporated herein by reference.
  • The percent of intact drug that is absorbed into the bloodstream is not narrowly critical, as long as a therapeutic-disorder-effective amount, e.g., a gastrointestinal-disorder-effective amount of a proton pump inhibiting agent, is absorbed following administration of the pharmaceutical composition to a subject. It is understood that the amount of proton pump inhibiting agent and/or antacid that is administered to a subject is dependent on, e.g., the sex, general health, diet, and/or body weight of the subject.
  • Illustratively, administration of a substituted bicyclic aryl-imidazole to a young child or a small animal, such as a dog, a relatively low amount of the proton pump inhibitor, e.g., about 1 mg to about 30 mg, will often provide blood serum concentrations consistent with therapeutic effectiveness. Where the subject is an adult human or a large animal, such as a horse, achievement of a therapeutically effective blood serum concentration will require larger dosage units, e.g., about 10 mg, about 15 mg, about 20 mg, about 30 mg, about 40 mg, about 80 mg, or about 120 mg dose for an adult human, or about 150 mg, or about 200 mg, or about 400 mg, or about 800 mg, or about 1000 mg dose, or about 1500 mg dose, or about 2000 mg dose, or about 2500 mg dose, or about 3000 mg dose, or about 3200 mg dose, or about 3500 mg dose for an adult horse.
  • In various other embodiments of the present invention, the amount of proton pump inhibitor administered to a subject is, e.g., about 1-2 mg/Kg of body weight, or about 0.5 mg/Kg of body weight, or about 1 mg/Kg of body weight, or about 1.5 mg/Kg of body weight, or about 2 mg/Kg of body weight.
  • Treatment dosages generally may be titrated to optimize safety and efficacy. Typically, dosage-effect relationships from in vitro and/or in vivo tests initially can provide useful guidance on the proper doses for subject administration. Studies in animal models generally may be used for guidance regarding effective dosages for treatment of gastrointestinal disorders or diseases in accordance with the present invention. In terms of treatment protocols, it should be appreciated that the dosage to be administered will depend on several factors, including the particular agent that is administered, the route chosen for administration, the condition of the particular subject.
  • In various embodiments, unit dosage forms for humans contain about 1 mg to about 120 mg, or about 1 mg, or about 5 mg, or about 10 mg, or about 15 mg, or about 20 mg, or about 30 mg, or about 40 mg, or about 50 mg, or about 60 mg, or about 70 mg, or about 80, mg, or about 90 mg, or about 100 mg, or about 110 mg, or about 120 mg of a proton pump inhibitor.
  • In a further embodiment of the present invention, the pharmaceutical formulation is administered in an amount to achieve a measurable serum concentration of a non-acid degraded proton pump inhibiting agent greater than about 100 ng/ml within about 30 minutes after administration of the pharmaceutical formulation. In another embodiment of the present invention, the pharmaceutical formulation is administered to the subject in an amount to achieve a measurable serum concentration of a non-acid degraded or non-acid reacted proton pump inhibiting agent greater than about 100 ng/ml within about 15 minutes after administration of the pharmaceutical formulation. In yet another embodiment, the pharmaceutical formulation is administered to the subject in an amount to achieve a measurable serum concentration of a non-acid degraded or non-acid reacted proton pump inhibiting agent greater than about 100 ng/ml within about 10 minutes after administration of the pharmaceutical formulation.
  • In another embodiment of the present invention, the composition is administered to the subject in an amount to achieve a measurable serum concentration of the proton pump inhibiting agent greater than about 150 ng/ml within about 15 minutes and to maintain a serum concentration of the proton pump inhibiting agent of greater than about 150 ng/ml from about 15 minutes to about 1 hour after administration of the composition. In yet another embodiment of the present invention, the composition is administered to the subject in an amount to achieve a measurable serum concentration of the proton pump inhibiting agent greater than about 250 ng/ml within about minutes and to maintain a serum concentration of the proton pump inhibiting agent of greater than about 150 ng/ml from about 15 minutes to about 1 hour after administration of the composition. In another embodiment of the present invention, the composition is administered to the subject in an amount to achieve a measurable serum concentration of the proton pump inhibiting agent greater than about 350 ng/ml within about 15 minutes and to maintain a serum concentration of the proton pump inhibiting agent of greater than about 150 ng/ml from about 15 minutes to about 1 hour after administration of the composition. In another embodiment of the present invention, the composition is administered to the subject in an amount to achieve a measurable serum concentration of the proton pump inhibiting agent greater than about 450 ng/ml within about 15 minutes and to maintain a serum concentration of the proton pump inhibiting agent of greater than about 150 ng/m1 from about 15 minutes to about 1 hour after administration of the composition.
  • In another embodiment of the present invention, the composition is administered to the subject in an amount to achieve a measurable serum concentration of the proton pump inhibiting agent greater than about 150 ng/ml within about 30 minutes and to maintain a serum concentration of the proton pump inhibiting agent of greater than about 150 ng/ml from about 30 minutes to about 1 hour after administration of the composition. In yet another embodiment of the present invention, the composition is administered to the subject in an amount to achieve a measurable serum concentration of the proton pump inhibiting agent greater than about 250 ng/ml within about 30 minutes and to maintain a serum concentration of the proton pump inhibiting agent of greater than about 150 ng/ml from about 30 minutes to about 1 hour after administration of the composition. In another embodiment of the present invention, the composition is administered to the subject in an amount to achieve a measurable serum concentration of the proton pump inhibiting agent greater than about 350 ng/ml within about 30 minutes and to maintain a serum concentration of the proton pump inhibiting agent of greater than about 150 ng/ml from about 30 minutes to about 1 hour after administration of the composition. In another embodiment of the present invention, the composition is administered to the subject in an amount to achieve a measurable serum concentration of the proton pump inhibiting agent greater than about 450 ng/ml within about 30 minutes and to maintain a serum concentration of the proton pump inhibiting agent of greater than about 150 ng/ml from about 30 minutes to about 1 hour after administration of the composition.
  • In still another embodiment of the present invention, the composition is administered to the subject in an amount to achieve a measurable serum concentration of a non-acid degraded or non-acid reacted proton pump inhibiting agent greater than about 500 ng/ml within about 1 hour after administration of the composition. In yet another embodiment of the present invention, the composition is administered to the subject in an amount to achieve a measurable serum concentration of a non-acid degraded or non-acid reacted proton pump inhibiting agent greater than about 300 ng/ml within about 45 minutes after administration of the composition.
  • In another embodiment of the present invention, the composition is administered to the subject in an amount sufficient to achieve a maximum serum concentration (Cmax) at a time (Tmax) that is within about 90, 70, 60, 50, 40, 30 or 20 minutes after administration of the composition according to the present invention.
  • In still another embodiment of the invention, the composition is administered to the subject in an amount sufficient to achieve a maximum serum concentration (Cmax) at a time (Tmax) that is between about 10 and about 90 minutes, between about 10 to about 60 minutes, between about 15 to about 60 minutes or between about 20 to about 60 minutes after administration of the composition according to the present invention. In some specific embodiments, the values of Cmax and Tmax are averages over a test population. In other specific embodiments, the values of Cmax and Tmax are the values for an individual.
  • In still another embodiment, the composition is administered in an amount sufficient to achieve a maximum serum concentration (Cmax) of from about 400 to about 3000 ng/mL, from about 400 to about 2500 ng/mL, from about 400 to about 2000 ng/mL, from about 400 to about 1500 ng/mL, from about 1000 to about 1500 ng/mL, from about 400 to about 1000 ng/mL or from about 400 to about 700 ng/mL. In some specific embodiments, the values of Cmax and Tmax are averages over a test population. In other specific embodiments, the values of Cmax and Tmax are the values for an individual.
  • In a further embodiment, the composition is administered in an amount sufficient to achieve a maximum serum concentration (Cmax) of greater than 400 ng/mL, greater than 600 ng/mL, greater than 1000 ng/mL. In some specific embodiments, the values of Cmax and Tmax are averages over a test population. In other specific embodiments, the values of Cmax and Tmax are the values for an individual.
  • Contemplated compositions of the present invention provide a therapeutic effect as proton pump inhibiting agent medications over an interval of about 5 minutes to about 24 hours after administration, enabling, for example, once-a-day, twice-a-day, three times a day, etc. administration if desired. Generally speaking, one will desire to administer an amount of the compound that is effective to achieve a serum level commensurate with the concentrations found to be effective in vivo for a period of time effective to elicit a therapeutic effect. Determination of these parameters is well within the skill of the art. These considerations are well known in the art and are described in standard textbooks.
  • In one embodiment of the present invention, the composition is administered to a subject in a gastrointestinal-disorder-effective amount, that is, the composition is administered in an amount that achieves a therapeutically-effective dose of a proton pump inhibiting agent in the blood serum of a subject for a period of time to elicit a desired therapeutic effect. Illustratively, in a fasting adult human (fasting for generally at least 10 hours) the composition is administered to achieve a therapeutically-effective dose of a proton pump inhibiting agent in the blood serum of a subject within about 45 minutes after administration of the composition. In another embodiment of the present invention, a therapeutically-effective dose of the proton pump inhibiting agent is achieved in the blood serum of a subject within about 30 minutes from the time of administration of the composition to the subject. In yet another embodiment, a therapeutically-effective dose of the proton pump inhibiting agent is achieved in the blood serum of a subject within about 20 minutes from the time of administration to the subject. In still another embodiment of the present invention, a therapeutically-effective dose of the proton pump inhibiting agent is achieved in the blood serum of a subject at about 15 minutes from the time of administration of the composition to the subject.
  • In further embodiments, greater than about 98%; or greater than about 95%; or greater than about 90%; or greater than about 75%; or greater than about 50% of the drug absorbed into the bloodstream is in a non-acid degraded or a non-acid reacted form.
  • In other embodiments, the pharmaceutical formulations provide a release profile of the proton pump inhibitor, using USP dissolution methods, whereby greater than about 50% of the proton pump inhibitor is released from the composition within about 2 hours; or greater than 50% of the proton pump inhibitor is released from the composition within about 1.5 hours; or greater than 50% of the proton pump inhibitor is released from the composition within about 1 hour after exposure to gastrointestinal fluid. In another embodiment, greater than about 60% of the proton pump inhibitor is released from the composition within about 2 hours; or greater than 60% of the proton pump inhibitor is released from the composition within about 1.5 hours; or greater than 60% of the proton pump inhibitor is released from the composition within about 1 hour after exposure to gastrointestinal fluid. In yet another embodiment, greater than about 70% of the proton pump inhibitor is released from the composition within about 2 hours; or greater than 70% of the proton pump inhibitor is released from the composition within about 1.5 hours; or greater than 70% of the proton pump inhibitor is released from the composition within about 1 hour after exposure to gastrointestinal fluid.
  • Dosage Forms
  • The pharmaceutical formulations of the present invention contain desired amounts of microencapsulated proton pump inhibitor and/or dry coated proton pump inhibitor and antacid can be in the form of, e.g., a tablet; including a suspension tablet, a chewable tablet, an effervescent tablet or caplet; a pill; a powder such as a sterile packaged powder, a dispensable powder, and an effervescent powder; a capsule including both soft or hard gelatin capsules such as HPMC capsules; a lozenge; a sachet; a troche; pellets; granules; or aerosol. These pharmaceutical formulations of the present invention can be manufactured by conventional pharmacological techniques.
  • The amount and types of buffers, proton pump inhibitors, and other excipients useful in each of these dosage forms are described throughout the specification and examples. It should be recognized that where a combination of buffer, proton pump inhibitor and/or excipient, including specific amounts of these components, is described with one dosage form that the same combination could be used for any other suitable dosage form. Moreover, it should be understood that one of skill in the art would, with the teachings found within this application, be able to make any of the dosage forms listed above by combining the components (i.e., amounts and types of PPIs, buffers, and other excipients) described in the different sections of the specification.
  • Moreover, each of the dosage forms may comprise one or more additional materials such as a pharmaceutically compatible carrier, binder, filling agent, suspending agent, flavoring agent, sweetening agent, disintegrating agent, surfactant, preservative, lubricant, colorant, diluent, solubilizer, moistening agent, stabilizer, wetting agent, anti-adherent, parietal cell activator, anti-foaming agent, antioxidant, chelating agent, antifungal agent, antibacterial agent, or one or more combination thereof.
  • Parietal cell activators are administered in an amount sufficient to produce the desired stimulatory effect without causing untoward side effects to patients. In one embodiment, the parietal cell activator is administered in an amount of about 5 mg to about 2.5 grams per 20 mg dose of the proton pump inhibitor.
  • The pharmaceutical compositions of the present invention can be manufactured by conventional pharmacological techniques. Conventional pharmacological techniques include, e.g., one or a combination of methods: (1) dry mixing, (2) direct compression, (3) milling, (4) dry or non-aqueous granulation, (5) wet granulation, or (6) fusion. See, e.g., Lachman et al., The Theory and Practice of Industrial Pharmacy (1986). Other methods include, e.g., prilling, spray drying, pan coating, melt granulation, granulation, wurster coating, tangential coating, top spraying, extruding, coacervation and the like.
  • In one embodiment, the proton pump inhibitor is microencapsulated or dry coated prior to being formulated into one of the above forms. In another embodiment, some or all of the antacid is also microencapsulated prior to being further formulated into one of the above forms. In still other embodiments, using standard coating procedures, such as those described in Remington's Pharmaceutical Sciences, 20th Edition (2000), a film coating is provided around the pharmaceutical formulation.
  • Provided herein are pharmaceutical formulations wherein some or all of the proton pump inhibitor and some or all of the antacid are microencapsulated or dry coated. In some embodiments, only some of the proton pump inhibitor is microencapsulated or dry coated. In other embodiments, all of the proton pump inhibitor is microencapsulated or dry coated. In still other embodiments, only some of the antacid is microencapsulated or dry coated.
  • In other embodiments, one or more layers of the pharmaceutical formulation are plasticized. Illustratively, a plasticizer is generally a high boiling point solid or liquid. Suitable plasticizers can be added from about 0.01% to about 50% by weight (w/w) of the coating composition. Plasticizers include, e.g., diethyl phthalate, citrate esters, polyethylene glycol, glycerol, acetylated glycerides, triacetin, polypropylene glycol, polyethylene glycol, triethyl citrate, dibutyl sebacate, stearic acid, stearol, stearate, and castor oil.
  • Exemplary Solid Compositions
  • In some embodiments, the pharmaceutical compositions of the present invention contain desired amounts of proton pump inhibiting inhibitor and antacid and are in a solid dosage form. In other embodiments, the pharmaceutical compositions of the present invention contain desired amounts of proton pump inhibitor and antacid and are administered in the form of a capsule (including both soft or hard capsules, e.g., capsules made from animal-derived gelatin or plant-derived HPMC).
  • Solid compositions, e.g., tablets, chewable tablets, effervescent tablets, and capsules, are prepared by mixing the microencapsulated proton pump inhibitor or dry coated proton pump inhibitor with one or more antacid and pharmaceutical excipients to form a bulk blend composition. When using dry coated proton pump inhibitor, mixing with additional antacid is optional. When referring to these bulk blend compositions as homogeneous, it is meant that the microencapsulated or dry coated proton pump inhibitor and antacid are dispersed evenly throughout the composition so that the composition may be readily subdivided into equally effective unit dosage forms, such as tablets, pills, and capsules. The individual unit dosages may also comprise film coatings, which disintegrate upon oral ingestion or upon contact with diluent.
  • Compressed tablets are solid dosage forms prepared by compacting the bulk blend compositions described above. In various embodiments, compressed tablets of the present invention will comprise one or more flavoring agents. In other embodiments, the compressed tablets will comprise a film surrounding the final compressed tablet. In other embodiments, the compressed tablets comprise one or more excipients and/or flavoring agents.
  • A capsule may be prepared, e.g., by placing the bulk blend composition, described above, inside of a capsule. In some embodiments, the pharmaceutical compositions of the present invention contain desired amounts of proton pump inhibiting inhibitor and antacid and are in a solid dosage form. In other embodiments, the pharmaceutical compositions of the present invention contain desired amounts of proton pump inhibitor and antacid and are administered in the form of a capsule (including both soft and hard capsules, e.g., capsules made from animal-derived gelatin or plant-derived HPMC). The pharmaceutical compositions of the present invention can be manufactured by conventional pharmacological techniques.
  • A chewable tablet may be prepared by compacting bulk blend compositions, described above. In one embodiment, the chewable tablet comprises a material useful for enhancing the shelf-life of the pharmaceutical formulation. In some embodiments, the proton pump inhibitor is dry coated. In another embodiment, microencapsulated material has taste-masking properties. In various other embodiments, the chewable tablet comprises one or more flavoring agents and one ore more taste-masking materials. In yet other embodiments the chewable tablet comprises both a material useful for enhancing the shelf-life of the pharmaceutical formulation and one or more flavoring agents.
  • In various embodiments, the microencapsulated or dry coated proton pump inhibitor, antacid, and optionally one or more excipients are dry blended and compressed into a mass, such as a tablet, having a hardness sufficient to provide a pharmaceutical composition that substantially disintegrates within less than about 30 minutes, less than about 35 minutes, less than about 40 minutes, less than about 45 minutes, less than about 50 minutes, less than about 55 minutes, or less than about 60 minutes, after oral administration, thereby releasing the antacid and the proton pump inhibitor into the gastrointestinal fluid. When at least 50% of the pharmaceutical composition has disintegrated, the compressed mass has substantially disintegrated.
  • In specific embodiments, chewable tablets are prepared using micronized proton pump inhibitor that has been combined with various combinations of potential excipients. In particular embodiments, micronized omeprazole is combined with various colorants, flavorings and other taste masking excipients. In specific embodiments, acidity in particular colorants, flavorings or other taste masking excipients can give rise to instability of the proton pump inhibitor.
  • In order to provide enhanced stability, some embodiments provide for encapsulation of micronized proton pump inhibitor in an immediate release coating. The immediate release coating is designed to protect the micronized proton pump inhibitor from degradation by acidic excipients, such as flavor components.
  • Exemplary Powder Compositions
  • A powder for suspension may be prepared by combining microencapsulated proton pump inhibitor and one or more antacid. In various embodiments, the powder may comprise one or more pharmaceutical excipients. In some embodiments, the proton pump inhibitor is micronized. Other embodiments of the present invention also comprise a suspending agent and/or a wetting agent.
  • Effervescent powders are also prepared in accordance with the present invention. Effervescent salts have been used to disperse medicines in water for oral administration. Effervescent salts are granules or coarse powders containing a medicinal agent in a dry mixture, usually composed of sodium bicarbonate, citric acid and/or tartaric acid. When salts of the present invention are added to water, the acids and the base react to liberate carbon dioxide gas, thereby causing “effervescence.” Examples of effervescent salts include the following ingredients: sodium bicarbonate or a mixture of sodium bicarbonate and sodium carbonate, citric acid and/or tartaric acid. Any acid-base combination that results in the liberation of carbon dioxide can be used in place of the combination of sodium bicarbonate and citric and tartaric acids, as long as the ingredients were suitable for pharmaceutical use and result in a pH of about 6 or higher.
  • The method of preparation of the effervescent granules of the present invention employs three basic processes: wet granulation, dry granulation and fusion. The fusion method is used for the preparation of most commercial effervescent powders. It should be noted that, although these methods are intended for the preparation of granules, the formulations of effervescent salts of the present invention could also be prepared as tablets, according to known technology for tablet preparation.
  • Wet granulation is one of the oldest methods of granule preparation. The individual steps in the wet granulation process of tablet preparation include milling and sieving of the ingredients, dry powder mixing, wet massing, granulation, and final grinding. In various embodiments, the microencapsulated omeprazole is added to the other excipients of the pharmaceutical formulation after they have been wet granulated.
  • Dry granulation by slugging involves compressing a powder mixture into a rough tablet or “slug” on a heavy-duty rotary tablet press. The slugs are then broken up into granular particles by a grinding or milling operation, usually by passage through an oscillation granulator. The individual steps include mixing of the powders, compressing (slugging) and grinding (slug reduction or granulation). In a larger scale operation, roller compaction can be used instead of “slugging”. Roller compaction procedure is well-known to ones skilled in the art. No wet binder or moisture is involved in any of the dry granulation steps. In some embodiments, the microencapsulated omeprazole is dry granulated with other excipients in the pharmaceutical formulation. In other embodiments, the microencapsulated omeprazole is added to other excipients of the pharmaceutical formulation after they have been dry granulated.
  • Other Exemplary Compositions
  • Pharmaceutical compositions suitable for buccal (sublingual) administration include, e.g., lozenges in a flavored base, such as sucrose, acacia, tragacanth, and pastilles comprising microencapsulated proton pump inhibitor in an inert base such as gelatin, glycerin, sucrose, and acacia are also provided herein.
  • Many other types of release delivery systems are available and known to those of ordinary skill in the art. Examples of such delivery systems include, e.g., polymer-based systems, such as polylactic and polyglycolic acid, plyanhydrides and polycaprolactone; nonpolymer-based systems that are lipids, including sterols, such as cholesterol, cholesterol esters and fatty acids, or neutral fats, such as mono-, di- and triglycerides; hydrogel release systems; silastic systems; peptide-based systems; wax coatings; compressed tablets using conventional binders and excipients partially fused implants and the like. See, e.g., Liberman et al., Pharmaceutical Dosage Forms, 2 Ed., Vol. 1, pp. 209-214 (1990).
  • In some embodiments, the pharmaceutical composition comprises (a) microencapsulated proton pump inhibitor; and (b) at least one antacid; wherein the pharmaceutical composition is made by the process of (a) microencapsulating some or all of the proton pump inhibitor; and (b) dry blending the microencapsulated material with some or all of the at least one antacid. In other embodiments, the pharmaceutical composition comprises (a) microencapsulated proton pump inhibitor, and (b) at least one antacid, wherein the microencapsulated proton pump inhibitor is made by the process of spray drying the proton pump inhibitor with a microencapsulating material. In still other embodiments, the pharmaceutical composition comprises (a) microencapsulated proton pump inhibitor, and (b) at least one antacid, wherein the pharmaceutical composition is made by the process of (a) microencapsulating some or all of the proton pump inhibitor, and (b) blending the microencapsulated material with some or all of the at least one antacid.
  • Treatment
  • Initial treatment of a subject suffering from a disease, condition or disorder where treatment with an inhibitor of H+/K+-ATPase is indicated can begin with the dosages indicated above. Treatment is generally continued as necessary over a period of hours, days, or weeks to several months or years until the disease, condition or disorder has been controlled or eliminated. Subjects undergoing treatment with the compositions disclosed herein can be routinely monitored by any of the methods well known in the art to determine the effectiveness of therapy. Continuous analysis of such data permits modification of the treatment regimen during therapy so that optimal effective amounts of compounds of the present invention are administered at any point in time, and so that the duration of treatment can be determined as well. In this way, the treatment regimen/dosing schedule can be rationally modified over the course of therapy so that the lowest amount of an inhibitor of H+/K+-ATPase exhibiting satisfactory effectiveness is administered, and so that administration is continued only so long as is necessary to successfully treat the disease, condition or disorder.
  • In one embodiment, the pharmaceutical formulations are useful for treating a condition, disease or disorder where treatment with a proton pump inhibitor is indicated. In other embodiments, the treatment method comprises oral administration of one or more compositions of the present invention to a subject in need thereof in an amount effective at treating the condition, disease, disorder. In another embodiment, the disease, condition or disorder is a gastrointestinal disorder. The dosage regimen to prevent, give relief from, or ameliorate the disease, condition or disorder can be modified in accordance with a variety of factors. These factors include the type, age, weight, sex, diet, and medical condition of the subject and the severity of the disorder or disease. Thus, the dosage regimen actually employed can vary widely and therefore can deviate from the dosage regimens set forth herein.
  • In some embodiments, the pharmaceutical formulation is administered post meal. In further embodiments, the pharmaceutical formulation administered post meal is in the form of a chewable tablet.
  • The present invention also includes methods of treating, preventing, reversing, halting or slowing the progression of a gastrointestinal disorder once it becomes clinically evident, or treating the symptoms associated with, or related to the gastrointestinal disorder, by administering to the subject a composition of the present invention. The subject may already have a gastrointestinal disorder at the time of administration, or be at risk of developing a gastrointestinal disorder. The symptoms or conditions of a gastrointestinal disorder in a subject can be determined by one skilled in the art and are described in standard textbooks. The method comprises the oral administration a gastrointestinal-disorder-effective amount of one or more compositions of the present invention to a subject in need thereof.
  • Gastrointestinal disorders include, e.g., duodenal ulcer disease, gastrointestinal ulcer disease, gastroesophageal reflux disease, erosive esophagitis, poorly responsive symptomatic gastroesophageal reflux disease, pathological gastrointestinal hypersecretory disease, Zollinger Ellison Syndrome, and acid dyspepsia. In one embodiment of the present invention, the gastrointestinal disorder is heartburn.
  • Besides being useful for human treatment, the present invention is also useful for other subjects including veterinary animals, reptiles, birds, exotic animals and farm animals, including mammals, rodents, and the like. Mammals include primates, e.g., a monkey, or a lemur, horses, dogs, pigs, or cats. Rodents includes rats, mice, squirrels, or guinea pigs.
  • In various embodiments of the present invention, the compositions are designed to produce release of the proton pump inhibitor to the site of delivery (typically the stomach), while substantially preventing or inhibiting acid degradation of the proton pump inhibitor.
  • In various embodiments of the present invention, the compositions are designed to produce release of the proton pump inhibitor for immediate release in vitro dissolution profile, which is well accepted to by those in the arts to mean drug release of greater than 70% (Q70) in 45 minutes in a USP dissolution test.
  • In vitro dissolution rate contemplated by the present invention include drug release of greater than 70% (Q70) in 15 minutes in a media with a wide pH range of 1-8.
  • Combination Therapy
  • The present pharmaceutical compositions can also be used in combination (“combination therapy”) with another pharmaceutical agent that is indicated for treating or preventing a gastrointestinal disorder, such as, e.g., an anti-bacterial agent, an alginate, a prokinetic agent, a H2 antagonist, an antacid, or sucralfate, which are commonly administered to minimize the pain and/or complications related to this disorder.
  • Combination therapies contemplated by the present invention include administration of a pharmaceutical formulation of the present invention in conjunction with another pharmaceutically active agent that is indicated for treating or preventing a gastrointestinal disorder in a subject, as part of a specific treatment regimen intended to provide a beneficial effect from the co-action of these therapeutic agents for the treatment of a gastrointestinal disorder. The beneficial effect of the combination includes, but is not limited to, pharmacokinetic or pharmacodynamic co-action resulting from the combination of therapeutic agents. Administration of these therapeutic agents in combination typically is carried out over a defined time period (usually substantially simultaneously, minutes, hours, days, weeks, months or years depending upon the combination selected).
  • Combination therapies of the present invention are also intended to embrace administration of these therapeutic agents in a sequential manner, that is, where each therapeutic agent is administered at a different time, as Well as administration of these therapeutic agents, or at least two of the therapeutic agents, in a substantially simultaneous manner. Substantially simultaneous administration can be accomplished, e.g., by administering to the subject a single tablet or capsule having a fixed ratio of each therapeutic agent or in multiple, single capsules, or tablets for each of the therapeutic agents. Sequential or substantially simultaneous administration of each therapeutic agent can be effected by any appropriate route.
  • The composition of the present invention can be administered orally or nasogastrointestinal, while the other therapeutic agent of the combination can be administered by any appropriate route for that particular agent, including, but not limited to, an oral route, a percutaneous route, an intravenous route, an intramuscular route, or by direct absorption through mucous membrane tissues. For example, the composition of the present invention is administered orally or nasogastrointestinal and the therapeutic agent of the combination may be administered orally, or percutaneously. The sequence in which the therapeutic agents are administered is not narrowly critical. Combination therapy also can embrace the administration of the therapeutic agents as described above in further combination with other biologically active ingredients, such as, but not limited to, a pain reliever, such as a steroidal or nonsteroidal anti-inflammatory drug, or an agent for improving stomach motility, e.g., and with non-drug therapies, such as, but not limited to, surgery.
  • The therapeutic compounds which make up the combination therapy may be a combined dosage form or in separate dosage forms intended for substantially simultaneous administration. The therapeutic compounds that make up the combination therapy may also be administered sequentially, with either therapeutic compound being administered by a regimen calling for two step administration. Thus, a regimen may call for sequential administration of the therapeutic compounds with spaced-apart administration of the separate, active agents. The time period between the multiple administration steps may range from, e.g., a few minutes to several hours to days, depending upon the properties of each therapeutic compound such as potency, solubility, bioavailability, plasma half-life and kinetic profile of the therapeutic compound, as well as depending upon the effect of food ingestion and the age and condition of the subject. Circadian variation of the target molecule concentration may also determine the optimal dose interval.
  • The therapeutic compounds of the combined therapies contemplated by the present invention, whether administered simultaneously, substantially simultaneously, or sequentially, may involve a regimen calling for administration of one therapeutic compound by oral route and another therapeutic compound by an oral route, a percutaneous route, an intravenous route, an intramuscular route, or by direct absorption through mucous membrane tissues, for example. Whether the therapeutic compounds of the combined therapy are administered orally, by inhalation spray, rectally, topically, buccally, sublingually, or parenterally (e.g., subcutaneous, intramuscular, intravenous and intradermal injections, or infusion techniques), separately or together, each such therapeutic compound will be contained in a suitable pharmaceutical formulation of pharmaceutically-acceptable excipients, diluents or other formulations components.
  • In one embodiment, the pharmaceutical formulations of the present invention are administered with low strength enteric coated Aspirin. In another embodiment, the second active pharmaceutical, e.g., Aspirin or an NSAID, used in combination with the pharmaceutical formulations of the present invention, is enteric coated. In other embodiments, antacid present in the pharmaceutical formulations of the present invention increase the pH level of the gastrointestinal fluid, thereby allowing part or all of the enteric coating on the second active pharmaceutical to dissolve in the stomach.
  • For the sake of brevity, all patents and other references cited herein are incorporated by reference in their entirety.
  • EXAMPLES
  • The present invention is further illustrated by the following examples, which should not be construed as limiting in any way. The experimental procedures to generate the data shown are discussed in more detail below. For all formulations herein, multiple doses may be proportionally compounded as is known in the art. The coatings, layers and encapsulations are applied in conventional ways using equipment customary for these purposes.
  • The invention has been described in an illustrative manner, and it is to be understood that the terminology used is intended to be in the nature of description rather than of limitation.
  • Example 1A Microencapsulation Materials and Methods
  • Microencapsulation Process Using Spinning Disk Atomization
  • The basic operation for the spinning disk process used (microencapsulation by spray drying) was as follows: An encapsulation solution or suspension was prepared by dissolving the encapsulation (or shell) material in the appropriate solvent. Omeprazole (or the proton pump inhibitor) was dispersed in the coating solution and fed onto the center of the spinning disk or spray dryer which is pre-heated. One of three atomization methods can be used to make droplets of the feed solution/suspension. The microspheres were formed by removal of the solvent using heated airflow inside the drying chamber and collected as a powder using a cyclone separator.
  • Atomization of the feed solution/suspension can be achieved by major methods in a spray dryer, by rotary atomization using a spinning disc, by high pressure-nozzle, or by two fluid nozzles. A thin film was produced across the surface of the disk and atomization occurs as the coating material left the periphery of the disk in a rotary atomization process. The microspheres were formed by removal of the solvent using heated airflow inside the atomization chamber and collected as a free-flowing powder using a cyclone separator. High pressure nozzle utilizes a high pressure pump to feed the solution/suspension through a various size nozzle so droplets form when injected into the drying chamber. The pressure energy is converted to kinetic energy and the feed issues form the orifice as a high speed film that readily disintegrates into droplets. For the two fluid nozzle, the atomization is created due to high frictional shearing forces between the liquid surface and the air having a high velocity even at sonic velocities and sometimes rotated to obtain maximum atomization.
  • Spray Drying Microencapsulation Process
  • The basic operation for the microencapsulation by spray drying was as follows: An encapsulation solution or suspension was prepared by dissolving the shell material in the appropriate solvent. Proton pump inhibitor was dispersed in the coating solution and fed onto the spray dryer which is pre-heated. One of the three atomization method can be used to make droplet of the feed solution/suspension. The microspheres were formed by removal of the solvent using heated airflow inside the drying chamber and collected as a powder using a cyclone separator.
  • A spray dryer with attached fluid-bed dryer for sizing of dried particles and/or agglomeration if desired can be also used. Recycling of the super-fine particles from the cyclones back to the spray dryer inlet would allow the agglomeration to form desired particle size distribution.
  • The dissolution profiles of the microencapsulated omeprazole were determined by a method similar to the HPLC method outlined in Example 10, described below. The size of the microspheres was determined by using a microscopic optical method similar to the one outlined in Example 11.
  • TABLE 1A
    OME load
    (wt %)
    Theoretical/ % Omeprazole Released (wt %)
    Sample Analytical Material Method Size 5 min 30 min 45 min 2 hour
    4 25%/22% KLX Disk-hot 25-125 −1.1 10.3 22.2 36.5
    BHT (0.1% of KLX) melt micron 1.5 10.1 17.3 34.6
    5 25%/23% Methocel A15LV Spray dry 5-30 103.7 100.7 99.2 98.3
    PEG 3350 (5%) micron 113.1 103.7 102.7 101.0
    6 25%/26% Methocel A15LV Spray dry 5-30 77.5 85.6 85.0 86.1
    PEG 300 (5%) micron 120.3 87.3 90.4 86.3
    BHT (0.1%)
    7 25%/39% Methocel A15LV Spray dry 5-30 29.8 37.7 41.5 51.7
    Span 20 (5%) micron 33.8 30.7 28.2 38.0
    BHT (0.1%)
    8 25%/24% Methocel A15LV Spray dry 5-30 89.8 97.2 95.1 90.4
    BHT (0.1%) micron 93.1 82.5 83.9 84.8
    10 3%/2% Methocel A15LV Spray dry 5-30 94.4 104.7 102.7 97.9
    PEG 3350 (5%) micron 104.6 99.2 98.2 93.2
    BHT (0.1%)
    Sodium
    bicarbonate
    11 25%/20% Opadry YS-1-7003 Spray dry 5-30 91.4 103.1 100.1 94.8
    PEG 3350 (5%) micron 99.3 98.8 95.8 91.0
    BHT (0.1%)
    12 25%/27% Methocel K4M Spray dry 5-30 134.1 92.9 86.9 85.0
    PEG 3350 (10%) micron 73.2 88.4 85.3 84.2
    BHT 74.5 75.3 73.5
    78.7 77.2 74.1
    13 25%/26% Kollicoat IR Spray dry 5-30 99.1 94.7 94.2 91.9
    PEG 3350 (5%) micron 89.7 87.7 84.9 84.6
    BHT
    14 25%/21% Eudragit RD 100 Spray dry 5-30 111.5 72.6 76.9 73.0
    PEG 3350 (5%) micron 48.9 73.1 74.1 73.7
    BHT (0.1%)
    15 25%/26% Klucel (HPC) Spray dry 5-30 76.8 82.1 83.1
    PEG 3350 (5%) micron 69.6 71.7 73.1
    BHT (0.1%)
    16 25%/25% Ethocel #7 Disk- 25-125 5.4 9.3 13.8 23.8
    solvent micron 14 9.7 12.4 22.5
    17 25%/25% Ethocel (50%) Disk- 25-125 122.6 105.9 106.2 97.6
    Methocel E5 (50%) solvent micron 113.4 100.4 103.9 97.9
    18 25%/25% Ethocel (75%) Disk- 25-125 61.6 73.0 60.9
    Methocel (25%) solvent micron 44.3 53.8 67.9
    37.0 47.0 59.2
    40.5 47.6 61.1
    19 25%/25% Methocel Disk- 25-125 78.7 80.5 78.1
    solvent micron 84.8 84.8 78.7
    78.0 80.3 78.1
    79.0 75.2 77.0
    20 2.4%/5%   Ethocel Disk- 25-125 25.0 28.8 33.6
    Sodium solvent micron, 23.2 33.3 30.3
    Bicarbonate 19.3 20.6 27.4
    16.1 17.4 22.6
    21 25%/22% Ethocel Disk- 25-125 31.7 44.6 59.4
    PEG 3350 (5%) solvent micron 38.1 52.5 59.6
    22 25%/22% Ethocel (50%) Disk- 25-125 89.9 88.6 86.7
    Klucel EXAF (50%) solvent micron 84.5 88.4 85.0
    23 25%/22% Klucel Disk- 25-100 88.1 90.2 88.1
    solvent microns 83.2 82.9 82.3
    24 25%/22% Sepifilm LP Disk- 25-100 97.0 95.2 92.2
    solvent micron 90.3 89.8 90.1
    25 25%/23% Eudragit E100 Disk- 25-80 13.2 17.0 24.8
    solvent micron 8.2 12.1 20.2
    26 40%/35% Eudragit E100 Disk- 25-80 5.1 6.4 11.5
    solvent micron 13.1 16.4 23.5
    27 40%/38% Eudragit E100 Disk- 25-80 15.0 16.2 27.0
    Span 20 (5%) solvent micron 16.9 20.1 26.3
    28 40%/35% Eudragit E100 Disk- 25-80 16.3 19.5 28.8
    PEG 300 (5%) solvent micron 16.0 12.9 28.5
    29 25%/25% Eudragit EPO Disk- 25-80 15.3 17.8 25.6
    solvent micron 11.9 14.5 21.2
    30 40%/36% Eudragit EPO Disk- 25-90 15.2 17.8 27.1
    solvent micron 17.5 17.5 30.9
    31 25%/24% Opadry AMB Spray dry <30 105.8 104.0 77.5
    micron 105.8 103.8 98.6
    34 25%/23% Kollicoat IR Spray dry 99.4 94.0 83.4
    101.6 99.5 96.3
    35 25%/26% Kollicoat IR Spray dry <30 104.2 97.0 86.3
    Sodium micron 99.1 95.3 91.1
    bicarbonate
    38 25%/26% Klucel Spray dry <30 81.3 77.3 72.1
    Sodium micron 93.8 90.5 85.8
    bicarbonate
    39 25%/15% Klucel(60%) Spray dry <50 91.4 86.4 82.6
    Sucraolse (10%) micron 101.5 97.2 93.4
    Sodium
    bicarbonate (30%)
    40 50%/47% Eudragit EPO Disk- 20-75 10.2 14.0 23.5
    solvent microns 10.6 13.6 24.3
    41 60%/57% Eudragit EPO Disk- 20-90 13.9 17.5 35.7
    solvent microns 6.7 17.4 33.8
    42 40%/39% Eudragit EPO(67%) Disk- 20-85 17.0 20.2 34.3
    Sodium bicarb(33%) solvent microns 16.4 19.4 20.2
    43 48%/48% EudragitEPO(61.5%) Disk- 20-110 17.3 28.0 51.0
    PEG 300(11.5%) solvent microns 22.2 24.9 50.3
    PEG 3350 (3.8%) 27.8(pH 5) 1.9(pH 5) 0(pH 5)
    Sod bicarb(23.2%) 22.4(pH 5) 1.7(pH 5) 0(pH 5)
    59.2(pH 6) 41.4(pH 6)  25.2(pH 6)  
    55.8(pH 6) 39.5(pH 6)  23.9(pH 6)  
    44 70%/66% Eudragit EPO Disk- 20-100 21.7 27.1 43.6
    solvent microns 21.3 25.2 54.4
    27.8(pH 5) 0.9(pH 5) 0(pH 5)
    17.8(pH 5) 0.5(pH 5) 0(pH 5)
    59.1(pH 6) 39.0(pH 6)  23.6(pH 6)  
    31.6(pH 6) 38.4(pH 6)  22.8(pH 6)  
    45 25%/26% Opadry AMB Spray dry 90.0 84.1 79.8
    (No TiO2) 87.1 84.6 79.6
    46 25%/24% Opadry AMB Spray dry 56.2 85.0 81.6
    (No TiO2) 90.0 85.8 81.7
    47 25%/24% Opadry AMB Spray dry 93.4 90.0 86.8
    (No TiO2) 88.9 87.5 82.7
    BHT (0.1%)
    51 66%/ Eudragit EPO Disk- 20-100 21.7 27.1 43.6
    solvent microns 21.3 25.2 54.4
    52 24%/ Opadry AMB Spray Dry 5-30 93.4 90.0 86.8
    BHT (aqueous) microns 88.9 87.5 82.7
  • Example 1B Microencapsulation of Omeprazole with Klucel-EF-37 wt-%
  • A suspension containing Klucel EF, sodium bicarbonate and omeprazole was prepared (total solid content of 16.23%) and spray dried using a rotary atomizer. The pH of the suspension was 8.1. Spray rate was 35 Kg/hour and the resulting outlet temperature was 70-85° C. Atomizer speed was 22,000 rpm. The viscosity of the suspension was 680 cps and the pumping system had no difficulty in delivering the suspension to the atomizer. White, fine particles were collected. The median particle size of sample was approximately 80-110 μm. USP No. 2 in vitro dissolution test showed drug release of >90% in 15 minutes. The amounts of each component are shown below:
  • Actual Amount Calculated Wt. %
    Raw Material Weighed out (Kg) in Dried Sample
    Klucel ® EF, NF 50 61.61
    Omeprazole, USP 30 36.97
    Sodium Bicarbonate (NaHCO3), 1.15 1.42
    USP
    USP Purified Water* 418.85
    Total 500.00 100.00
  • Example 1C Microencapsulation of Omeprazole with Klucel-EF-28 wt-%
  • A suspension containing Klucel EF, sodium bicarbonate and omeprazole was prepared (total solid content of 16.23%) and spray dried using a rotary atomizer. The pH of the suspension was 8.1. The inlet temperature was 130° C. and the outlet temperature was 84-85° C. Atomizer speed was 27,000 rpm. The viscosity of the suspension was 680 cps and the pumping system had no difficulty in delivering the suspension to the atomizer. White, fine particles were collected. The median particle size of sample was approximately 35-40 μm USP No. 2 in vitro dissolution test showed drug release of >35% in 30 minutes. The amounts of each component are shown below:
  • Actual Amount Calculated Wt. %
    Raw Material Weighed out (Kg) in Dried Sample
    Klucel ® EF, NF 10.0 70.37
    Omeprazole, USP 4.0 28.15
    Sodium Bicarbonate (NaHCO3), 0.21 1.48
    USP
    USP Purified Water* 85.79
    Total 100.00 100.00
  • Example 1D Microencapsulation of Omeprazole with Methocel E5
  • The microencapsulation of omeprazole consisted of two main steps: 1) suspension preparation and 2) spray drying operation. Methocel E5 was weighed out and added into water extremely slowly until a clear, homogeneous. PEG400 was added to the Methocel E5 polymer solution. Omeprazole and sodium bicarbonate were added to the polymer solution with agitation until a milky white, homogeneous suspension was formed. The finished omeprazole suspension was agitated at a minimum speed to prevent settling. The omeprazole suspension was then sprayed using the parameter shown below.
  • Inlet Temperature 135° C.
    Outlet Temperature 85° C.
    Atomizer Rotational Speed 27,000 rpm

    White, fine particles were collected. The average particle size of sample was approximately 35-54 μm USP No. 2 in vitro dissolution test showed drug release of >70% in 15 minutes. The amount of each component is shown below:
  • Actual Amount Calculated Wt. %
    Raw Material Weighed out (Kg) in Dried Sample
    Methocel ® E5, NF 3.80 60.03
    Omeprazole, USP 1.60 25.28
    PolyEthylene Glycol 400 0.80 12.64
    (PEG400), NF
    Sodium Bicarbonate (NaHCO3), 0.13 2.05
    USP
    USP Purified Water 33.70
    Total 40.03 100.00
  • Example 1E Microencapsulation of Omeprazole Using High Pressure Nozzle Spray Drying
  • A suspension containing hydroxypropyl cellulose, sodium bicarbonate and omeprazole was prepared (total solid content of 15.0%) and spray dried using a high pressure nozzle. The pH of the suspension was 8.5. The inlet temperature was 140-150° C. and the outlet temperature was 79-85° C. Core and the Nozzle size (SK value) used in the spray drying was 72 and 17, respectively. The viscosity of the suspension was in the range of 600-700 cps and the pumping system had no difficulty in delivering the suspension to the atomizer. White, fine particles were collected. The median particle size of sample was approximately 60 μm. The amounts of each component are shown below:
  • Actual Amount Calculated Wt. %
    Raw Material Weighed out (Kg) in Dried Sample
    Klucel ®-EF, NF 70.00 61.6
    Omeprazole, USP 42.00 37.0
    Sodium Bicarbonate (NaHCO3), 1.61 1.4
    USP
    USP Purified Water 644.00
    Total 757.61 100.00
  • Example 2A Preparation of Microencapsulated Omeprazole
  • The chart below summarizes the wt %, the feed rates used, and the inlet/outlet temperatures for eleven different omeprazole microspheres.
  • TABLE 2A
    Omeprazole payload Inlet/
    in microcapsule Feed Outlet
    Method and (Theoretical/ Microencapsulation Wt % of Rate Temp
    Sample Solvent Analytical) Material material (g/min) (° C.)
    53 Spray dry* 25%/23.3% Omeprazole (25%) 5% 4.2 125/70
    Water Methocel A15 LV (70%)
    PEG 3350 (5%)
    54 Spray dry 25%/24.8% Omeprazole (25%) 5% 4.0 125/70
    Water Methocel A15 LV (74.9%)
    BHT (0.1%)
    55 Spray dry 25%/20.5% Omeprazole (25%) 5% 4.2 126/60
    Water Opadry YS-1-7003 (70.9%)
    PEG 3350 (5%)
    BHT (0.1%)
    56 Spray dry 25%/26.2% Omeprazole (25%) 10%  3.0 128/85
    Water Kollicoat IR (70.9%)
    PEG 3350 (5%)
    BHT (0.1%)
    57 Spray dry 25%/21.3% Omeprazole (25%) 5% 4.0 127/87
    Water Eudragit RD100 (70.9%)
    PEG 3350 (5%)
    BHT (0.1%)
    58 Spray dry 25%/26.0% Omeprazole (25%) 5% 4.2 126/83
    Water Klucel (70.9%)
    PEG 3350 (5%)
    BHT (0.1%)
    59 (not Spinning disk** Klucel 10%  90   /52
    made into 75% Methanol
    tablets) 25% Acetone
    60 Spray dry 25%/26.0% Omeprazole (25%) 5% 4.5 129/86
    Water Kollicoat (73%)
    Sodium Bicarb (2%)
    61 Spray dry 25%/26.3% Omeprazole (25%) 5% 4.5 122/84
    Water Klucel (73%)
    Sodium Bicarb (2%)
    62 Spinning disk 70%/66.1% Omeprazole (70%) 10%  90   /50
    75% Methanol Eudragit EPO (30%)
    25% Acetone
    63 Spray dry 25%/24.7% Omeprazole (25%) 10%  4.4 124/79
    Water Opadry AMB (74.9%)
    BHT (0.1%)
    *Used a concentric nozzle with 0.055 inch air opening and a 0.028 inch fluid opening.
    **Used a 3-inch stainless steel disk rotating at approximately 4,500 rpm.
  • The amount of encapsulated omeprazole used in each tablet batch varies based on the actual payload of each set of microcapsules to achieve the theoretical dose of 40 mg. The omeprazole was microencapsulated in a similar manner as that described in Example 1. All ingredients are mixed well to achieve a homogeneous blend.
  • Tablets containing omeprazole microspheres were prepared using a high-speed rotary tablet press (TBCB Pharmaceutical Equipment Group, Model ZPY15). Round, convex tablets with diameters of about 10 mm and an average weight of approximately 600 mg per tablet were prepared.
  • Chewable tablets were manufactured using the following materials: Encapsulated omeprazole (varied based on payload, to deliver 40 mg potency), sodium bicarbonate (1260 mg), calcium carbonate (790 mg), croscarmellose sodium (64 mg), Klucel (160 mg), Xylitab 100 (380 mg), microcrystalline cellulose (128 mg), sucralose (162 mg), peppermint flavor (34 mg), peach flavor (100 mg), masking powder (60 mg), FD&C Lake No. 40 Red (3 mg), and magnesium stearate (32 mg).
  • Example 2B Preparation of Microencapsulated PPIs
  • Table 2B below summarizes various drug wt %, shell materials, and in-vitro dissolution data for eighteen different proton pump inhibitor microspheres. The atomization method for the spray drying is also listed and is performed in a manner analogous to that listed in Example 1 above.
  • TABLE 2B
    Materials used in Chewable Tablets
    pH
    Modifier In Vitro
    No. PPI Shell Material (Buffer) Atomization Dissolution
    2B-2 Lansoprazole-30% HPC (Klucel-EF)-69.0% SC 1% High Pressure Q70 in 30 min
    2B-3 Pentoprazole- HPC (Klucel-EF)-54.0% SB 1% High Pressure Q70 in 30 min
    sodium 45%
    2B-4 Omeprazole-Mg HPC (Klucel-EF)-54.0% SB 1% High Pressure Q70 in 30 min
    45%
    2B-5 Rabeprazole-Na- HPC (Klucel-EF)-59% SB 1% Spinning disc Q70 in 30 min
    40%
    2B-7 Lansoprazole-30% HPMC(Methocel-E5)- SC 1% Spinning disc Q70 in 45 min
    64.0%
    PEG400 5%
    2B-8 Pentoprazole- HPMC (Methocel-E5)- SC 1% High Pressure Q70 in 30 min
    sodium 45% 44%
    PEG400 10%
    2B-9 Omeprazole-Mg HPMC (Methocel-E5)- SB 1% High Pressure Q70 in 30 min
    45% 54.9%
    PEG 400 10%
    2B-10 Rabeprazole-Na- HPMC (Methocel-E5)- SC 1% High Pressure Q70 in 15 min
    40% 51%
    PEG400 8%
    2B-11 Omeprazole-37% MC (Methocel-A15)-52% SB 3% Spinning disc Q70 in 30 min
    PEG400 10%
    2B-12 Lansoprazole-30% MC (Methocel-A15)-57% SC 1% Spinning disc Q70 in 30 min
    PEG400 12%
    2B-13 Pentoprazole- MC (Methocel-A15)-54% SC 1% High Pressure Q70 in 30 min
    sodium 45%
    2B-14 Omeprazole-Mg MC (Methocel-A15)-54% SB 1% High Pressure Q70 in 30 min
    45%
    2B-15 Rabeprazole-Na- MC (Methocel-A15)-59% SC 1% High Pressure Q70 in 30 min
    40%
    2B-16 Omeprazole-37% Klucel-EF 30% SB 3% High Pressure Q70 in 30 min
    MC (Methocel-A15)-22%
    PEG400 3%
    2B-17 Omeprazole-37% Klucel-EF 30% SB 3% High Pressure Q70 in 30 min
    HPMC (Methocel-E5)-
    22%
    PEG400 3%
    2B-18 Lansoprazole-40% Klucel-EF 30% SC 2% High Pressure Q70 in 30 min
    HPMC (Methocel-E5)-
    22%
    PEG400 4%
  • Example 2C Preparation of Lansoprazole Chewable Tablets
  • Lansoprazole chewable tablets are manufactured using the following materials: Microencapsulated lansoprazole (30% drug loading-Entry 2B-2 in Table 2B, 100 mg to deliver 30 mg drug potency), sodium bicarbonate (420 mg), direct-compression grade magnesium hydroxide (contains 5% starch, 526 mg to deliver 500 mg magnesium hydroxide), [total buffer=24.1 mEq], croscarmellose sodium (45 mg), Klucel (80 mg), Xylitab 300 (200 mg), sucralose (70 mg), fruit flavor crystals (52 mg), masking powder (30 mg), Lake FD & C Red #40 (2 mg), and magnesium stearate (20 mg).
  • The amount of microencapsulated lansoprazole used in each tablet batch is based on the actual payload of microcapsules to achieve the theoretical dose of 30 mg lansoprazole per tablet. Lansoprazole is microencapsulated in a similar manner as that described in Example 1 and 2. All ingredients are mixed well to achieve a homogenous bulk blend which is then compressed to round tablets.
  • Example 2D Preparation of Omeprazole Salt Caplets
  • Omeprazole magnesium tablets are manufactured using the following materials: Microencapsulated Omeprazole-Mg salt (45% drug loading-Entry 2B-14 in Table 2B, 89 mg to deliver 40 mg omeprazole), sodium bicarbonate (420 mg), direct-compression grade magnesium hydroxide (contains 5% starch, 337 mg to deliver 320 mg magnesium hydroxide), croscarmellose sodium (50 mg), Klucel-EXF (50 mg), and magnesium stearate (7 mg) [total 16 mEq in swallowable tablet or caplet].
  • The amount of microencapsulated omeprazole used in each tablet batch is based on the actual payload of microcapsules to achieve the theoretical dose of 40 mg omeprazole per tablet (44.5 mg of omeprazole magnesium salt trihydrate). Omeprazole magnesium salt is microencapsulated in a similar manner as that described in Example 1 and 2. All ingredients are mixed well to achieve a homogenous bulk blend which is then compressed to capsule shaped tablets.
  • Example 2E Preparation of Pantoprazole Chewable Tablets
  • Pantoprazole chewable tablets are manufactured using the following materials: Microencapsulated pentoprazole (45% drug loading-Entry 2B-8 in Table 2B, 89 mg to deliver 40 mg drug potency), sodium bicarbonate (336 mg), potassium bicarbonate (200 mg) direct-compression grade magnesium hydroxide (contains 5% starch, 430 mg to deliver 408 mg magnesium hydroxide) [total buffer=20 mEq], Klucel (70 mg), Xylitab 100 (200 mg), sucralose (70 mg), mint flavor crystals (52 mg), masking powder (30 mg), Lake FD & C Blue #2 (2 mg), and magnesium stearate (15 mg).
  • The amount of microencapsulated pantoprazole used in each tablet batch is based on the actual payload of microcapsules to achieve the theoretical dose of 40 mg pentoprazole per tablet. Pantoprazole is microencapsulated in a similar manner as that described in Example 1 and 2. All ingredients are mixed well to achieve a homogenous bulk blend which is then compressed to round tablets.
  • Example 2F Preparation of Rabeprazole Sodium Salt Caplets
  • Rabeprazole sodium caplets (capsule-shaped tablets) are manufactured using the following materials: Microencapsulated Rabeprazole sodium salt (40% drug loading-Entry 2B-10 Table 2B, 106 mg to deliver 40 mg rabeprazole), sodium bicarbonate (336 mg), Direct-compression grade magnesium hydroxide (contains 5% starch, 307 mg to deliver 292 mg magnesium hydroxide), magnesium oxide (101 mg), croscarmellose sodium (50 mg), Klucel-EXF (40 mg), and magnesium stearate (7 mg) [total 14mEq in swallowable tablet or caplet].
  • The amount of microencapsulated rabeprazole sodium used in each tablet batch is based on the actual payload of microcapsules to achieve the theoretical dose of 40 mg rabeprazole per tablet (42.5 mg of rabeprazole sodium salt). Rabeprazole sodium salt is microencapsulated in a similar manner as that described in Example 1 and 2. All ingredients are mixed well to achieve a homogenous bulk blend which is then compressed to tablets.
  • Example 2G Preparation of Capsules Containing Lansoprazole Capsule
  • The capsule product is manufactured using the following materials: Microencapsulated lansoprazole (40% drug loading-Entry 2B-18 in Table 2B, 75 mg, to deliver 30 mg potency), sodium bicarbonate (420 mg), magnesium hydroxide (218 mg), magnesium oxide (151 mg) (total buffer=23.3 mEq), croscarmellose sodium (50 mg), and magnesium stearate (5 mg).
  • The amount of encapsulated omeprazole used in each capsule batch varies based on the actual payload of each set of microcapsules to achieve the theoretical dose of 30 mg. Lansoprazole was microencapsulated in a similar manner as that described in Example 1 and 2. All ingredients are mixed well to achieve a homogenous bulk blend which is then filled into a capsule such as a size (00) hard gelatin capsule.
  • Example 2H Preparation of Capsules Containing Lansoprazole Microspheres
  • The capsule product is manufactured using the following materials: Microencapsulated lansoprazole (40% drug loading-Entry 2B-18 in Table 2B, 75 mg, to deliver 30 mg potency), sodium bicarbonate (600 mg), magnesium hydroxide (270 mg), [total buffer=18.4], croscarmellose sodium (50 mg), and magnesium stearate (7 mg).
  • The amount of encapsulated omeprazole magnesium salt used in each capsule batch varies based on the actual payload of each set of microcapsules to achieve the theoretical dose of 30 mg lansoprazole per capsule. Omeprazole magnesium salt was microencapsulated in a similar manner as that described in Example 1 and 2. All ingredients are mixed well to achieve a homogenous bulk blend which is then filled into a capsule such as a size zero (0) hard gelatin capsule.
  • Example 2I Preparation of Capsules Containing Omeprazole Magnesium Salt Microspheres
  • The capsule product is manufactured using the following materials: Microencapsulated omeprazole magnesium salt (45% drug loading-Entry 2B-4 in Table 2B, 88 mg to deliver 40 mg potency), sodium bicarbonate (504 mg), magnesium oxide (220 mg) [total buffer=17], croscarmellose sodium (40 mg), and magnesium stearate (7 mg).
  • The amount of encapsulated omeprazole used in each capsule batch varies based on the actual payload of each set of microcapsules to achieve the theoretical dose of 30 mg per capsule. Omeprazole magnesium salt was microencapsulated in a similar manner as that described in Example 1 and 2. All ingredients are mixed well to achieve a homogenous bulk blend which is then filled into a capsule such as a size zero (0) hard gelatin capsule.
  • Example 2J Preparation of Capsules Containing Pentoprazole Microspheres
  • The capsule product is manufactured using the following materials: Microencapsulated pentoprazole sodium salt (45% drug loading-Entry 2B-13 in Table 2B, 88 mg, to deliver 40 mg potency), sodium bicarbonate (336 mg) (total buffer=15), magnesium oxide (222 mg), croscarmellose sodium (35 mg), and magnesium stearate (7 mg).
  • The amount of encapsulated pentoprazole sodium salt used in each capsule batch varies based on the actual payload of each set of microcapsules to achieve the theoretical dose of 40 mg neutral pentoprazole per capsule. Pentoprazole sodium salt was microencapsulated in a similar manner as that described in Example 1 and 2. All ingredients are mixed well to achieve a homogenous bulk blend which is then filled into a capsule such as a size one hard gelatin capsule.
  • Example 3 Preparation of Chewable Tablets
  • Various chewable tablets are manufactured using the microencapsulate materials described in Examples 1 and 2. The amount of microencapsulated omeprazole used in each chewable tablet batch is based on the actual payload of each set of microcapsules to achieve the theoretical dose. All ingredients are mixed well to achieve a homogenous bulk blend which is then compressed to chewable tablets.
  • PPI Buffering Agent Sleep Aid Excipient
    20 mg per chewable tablet 20.6 mEq or 600 mg Mg(OH)2 0.125 triazolam mg per 170 mg Xylitab
    (microencapsulated) 5.0 mEq or 420 mg NaHCO3 tablet 30 mg Ac-Di-Sol
    25.6 mEq or 1020 mg total buffer 100 mg Klucel
    25 mg cherry flavor
    15 mg magnesium stearate
    3 mg Red #40 Lake
  • PPI Buffering Agent Sleep Aid Excipient
    40 mg per chewable tablet 24.0 mEq or 700 mg Mg(OH)2 5 mg zolpidem per tablet 170 mg Dipac sugar
    (microencapsulated) 7.2 mEq or 600 mg NaHCO 3 30 mg Ac-Di-Sol
    31.1 mEq or 1300 mg total buffer 120 mg Klucel
    27 mg grape flavor
    15 mg magnesium stearate
    1 mg Red #40 Lake
    1 mg Blue #2 Lake
  • PPI Buffering Agent Sleep Aid Excipient
    15 mg 17.1 mEq or 500 mg Mg(OH)2 5 mg zaleprone per tablet 170 mg Dipac sugar
    lansoprazole per 3.0 mEq or 250 mg NaHCO 3 30 mg Ac-Di-Sol
    chewable tablet 20.1 mEq or 750 mg total buffer 120 mg Klucel
    27 mg grape flavor
    15 mg magnesium stearate
    1 mg red #40 lake
    1 mg blue #2 lake
  • PPI Buffering Agent Sleep Aid Excipient
    30 mg lansoprazole 24.0 mEq or 700 mg Mg(OH)2 25 mg diphenhydramine per 170 mg Xylitab
    per chewable tablet 5 mEq or 420 mg NaHCO3 tablet 30 mg Ac-Di-Sol
    (microencapsulated) 29.0 mEq or 1120 mg total buffer 100 mg Klucel
    25 mg cherry flavor
    15 mg magnesium stearate
    3 mg Red #40 Lake
  • PPI Buffering Agent Prokinetic Agent Excipient
    20 mg 20.6 mEq or 600 mg Mg(OH)2 5 mg Mosapride per tablet 170 mg Xylitab
    microencapsulated 5.0 mEq or 420 mg NaHCO 3 30 mg Ac-Di-Sol
    omeprazole per 25.6 mEq or 1020 mg total buffer 100 mg Klucel
    chewable tablet 40 mg Sucralose
    25 mg cherry flavor
    15 mg magnesium stearate
    3 mg Red #40 Lake
  • PPI Buffering Agent Prokinetic Agent Excipient
    40 mg 24.0 mEq or 700 mg Mg(OH)2 10 mg Domperidone per 170 mg Dipac sugar
    microencapsulated 7.1 mEq or 600 mg NaHCO3 tablet 30 mg Ac-Di-Sol
    omeprazole per 31.1 mEq or 1300 mg total buffer 120 mg Klucel
    chewable tablet 27 mg grape flavor
    15 mg magnesium stearate
    1 mg Red #40 Lake
    1 mg Blue #2 Lake
  • PPI Buffering Agent Prokinetic Agent Excipient
    30 mg 24.0 mEq or 700 mg Mg(OH)2 20 mg Clebopride per 170 mg Xylitab
    microencapsulated 5.0 mEq or 420 mg NaHCO3 tablet 30 mg Ac-Di-Sol
    lansoprazole per 29.0 mEq or 1120 mg total buffer 100 mg Klucel
    chewable tablet 25 mg cherry flavor
    15 mg magnesium stearate
    3 mg Red #40 Lake
  • PPI Buffering Agent Prokinetic Agent Excipient
    60 mg 15 mEq or 750 mg Ca(OH)2 10 mg Norcisapride per 170 mg Xylitab
    microencapsulated 15 mEq or 1260 mg NaHCO3 tablet 30 mg Ac-Di-Sol
    omeprazole per 30 mEq or 2010 mg total buffer 100 mg Klucel
    chewable tablet 25 mg cherry flavor
    15 mg magnesium stearate
    3 mg Red #40 Lake
  • PPI Buffering Agent Prokinetic Agent Excipient
    40 mg 15 mEq or 750 mg Ca(OH)2 10 mg Cisapride per tablet 170 mg Xylitab
    microencapsulated 10 mEq or 840 mg NaHCO 3 30 mg Ac-Di-Sol
    omprazole per chewable 25 mEq or 1590 mg total buffer 100 mg Klucel
    tablet
    15 mg mint flavor
    15 mg magnesium stearate
  • PPI Buffering Agent Prokinetic Agent Excipient
    40 mg 24.0 mEq or 700 mg Mg(OH)2 10 mg Cisapride per tablet 60 mg sucralose
    microencapsulated 7.1 mEq or 600 mg NaHCO 3 60 mg Ac-Di-Sol
    omeprazole per bite- 31.1 mEq or 1300 mg total buffer 60 mg pregelatinized starch
    disintegration chewable
    30 mg Klucel
    tablet 27 mg grape flavor
    15 mg magnesium stearate
    1 mg Red #40 Lake
    1 mg Blue #2 Lake
  • PPI Buffering Agent Prokinetic Agent Excipient
    30 mg 17.1 mEq or 500 mg Mg(OH)2 2.5 mg Mosapride per 60 mg sucralose
    microencapsulated 5 mEq or 420 mg NaHCO3 tablet 60 mg Ac-Di-Sol
    lansoprazole per bite- 22.1 mEq or 920 mg total buffer 70 mg pregelatinized starch
    disintegration chewable
    30 mg Klucel
    tablet 25 mg cherry flavor
    15 mg magnesium stearate
    3 mg Red #40 Lake
  • Prokinetic
    PPI Buffering Agent Agent Excipient
    60 mg 15 mEq or 750 mg 10 mg 60 mg sucralose
    micro- Ca(OH)2 Domperidone 60 mg Ac-Di-Sol
    encapsulated 15 mEq or 1260 mg per tablet 60 mg pregela-
    omeprazole NaHCO3 tinized starch
    per bite- 30 mEq or 2010 mg 30 mg Klucel
    disinte- total buffer 25 mg cherry flavor
    gration 15 mg magnesium
    chewable stearate
    tablet
    3 mg Red #40 Lake
  • Prokinetic
    PPI Buffering Agent Agent Excipient
    40 mg 15 mEq or 750 mg 10 mg 60 mg sucralose
    micro- Ca(OH)2 Norcisapride 60 mg Ac-Di-Sol
    encapsulated 10 mEq or 840 mg 60 mg pregela-
    omprazole NaHCO3 tinized starch
    per bite- 25 mEq or 1590 mg 30 mg Klucel
    disinte- total buffer 15 mg mint flavor
    gration 15 mg magnesium
    chewable stearate
    tablet
  • PPI Antacid Excipient
    40 mg 5.1 mEq or 150 mg 20 mg Ac-Di-Sol
    micro- Mg(OH)2 40 mg Microcrystalline
    encapsulated 3.0 mEq or 250 mg cellulose (MCC, PH102)
    omeprazole NaHCO 3 7 mg magnesium
    per chewable 8.1 mEq or 400 mg stearate
    tablet total antacid
  • PPI Antacid Excipient
    15 mg 8.6 mEq or 250 mg 30 mg Ac-Di-Sol
    micro- Mg(OH)2 55 mg Plasdone
    encapsulated 2.4 mEq or 200 mg 8 mg magnesium
    lansoprazole NaHCO3 stearate
    per chewable 11.0 mEq or 450 mg
    tablet total antacid
  • PPI Antacid Excipient
    10 mg 3.4 mEq or 100 mg 15 mg Ac-Di-Sol
    micro- Mg(OH)2 40 mg Klucel
    encapsulated 3.0 mEq or 250 mg 6 mg magnesium
    omeprazole NaHCO3 stearate
    per c chewable 6.4 mEq or 350 mg
    tablet total antacid
  • PPI Antacid Excipient
    40 mg 5.1 mEq or 150 mg 20 mg Ac-Di-Sol
    micro- Mg(OH)2 50 mg Microcrystalline
    encapsulated 3.8 mEq or 315 mg Cellulose (MCC, PH102)
    omeprazole NaHCO3 10 mg magnesium
    per chewable 8.9 mEq or 465 mg stearate
    tablet total antacid
  • PPI Antacid Excipient
    20 mg 5.1 mEq or 150 mg 100 mg Xylitab
    micro- Mg(OH)2 30 mg Ac-Di-Sol
    encapsulated 3.8 mEq or 315 mg 80 mg Klucel
    omeprazole NaHCO
    3 20 mg Sucralose
    per chewable 8.9 mEq or 465 mg 10 mg cherry flavor
    tablet total antacid 10 mg magnesium
    stearate
    1 mg Red #40 Lake
  • PPI Antacid Excipient
    40 mg 7.5 mEq or 220 mg 100 mg Dipac sugar
    micro- Mg(OH)2 20 mg Ac-Di-Sol
    encapsulated 2.4 mEq or 200 mg 80 mg Klucel
    omeprazole NaHCO3 17 mg grape flavor
    per chewable 9.9 mEq or 420 mg 11 mg magnesium
    tablet total antacid stearate
    1 mg Red #40 Lake
    1 mg Blue #2 Lake
  • PPI Antacid Excipient
    30 mg 5.1 mEq or 150 mg 70 mg Destab Sugar
    micro- Mg(OH)2 30 mg Ac-Di-Sol
    encapsulated 3.8 mEq or 315 mg 100 mg Klucel
    lansoprazole NaHCO
    3 20 mg Asulfame-K
    per chewable 8.9 mEq or 465 mg 15 mg cherry flavor
    tablet total antacid 9 mg magnesium
    stearate
    1 mg Red #40 Lake
  • PPI Antacid Excipient
    60 mg 4.4 mEq or 220 mg 80 mg Xylitab
    micro- Ca(OH)2 30 mg Ac-Di-Sol
    encapsulated 3.6 mEq or 300 mg 100 mg Klucel
    omeprazole NaHCO3 35 mg Sucralose
    per chewable 8.0 mEq or 520 mg 10 mg cherry flavor
    tablet total antacid 9 mg magnesium
    stearate
    2 mg Red #40 Lake
  • PPI Antacid Excipient
    40 mg 7.0 mEq or 350 mg 70 mg Xylitab
    micro- Ca(OH)2 30 mg Ac-Di-Sol
    encapsulated 3.0 mEq or 250 mg 10 mg Sucralose
    omeprazole NaHCO 3 80 mg Klucel
    per chewable 10.0 mEq or 600 mg 10 mg mint flavor
    tablet total antacid 8 mg magnesium
    stearate
  • PPI Antacid Excipient
    40 mg 8.0 mEq or 400 mg 20 mg sucralose
    micro- Ca(OH)2 40 mg Ac-Di-Sol
    encapsulated 3.6 mEq or 300 mg 35 mg pregela-
    omeprazole NaHCO3 tinized starch
    per chewable 11.6 mEq or 700 mg 25 mg Klucel
    tablet total 15 mg cherry flavor
    8 mg magnesium
    stearate
    1 mg Red #40 Lake
  • PPI Antacid Excipient
    30 mg 5.1 mEq or 150 mg 27 mg sucralose
    micro- Mg(OH)2 40 mg Ac-Di-Sol
    encapsulated 3.8 mEq or 315 mg 35 mg pregela-
    lansoprazole NaHCO3 tinized starch
    per chewable 8.9 mEq or 465 mg 30 mg Microcrystalline
    tablet total antacid Cellulose (PH101)
    20 mg cherry flavor
    10 mg magnesium
    stearate
    2 mg Red #40 Lake
  • PPI Antacid Excipient
    60 mg 7.9 mEq or 230 mg 34 mg sucralose
    micro- Mg(OH)2 30 mg Ac-Di-Sol
    encapsulated 3.0 mEq or 250 mg 35 mg pregela-
    omeprazole NaHCO3 tinized starch
    per chewable 10.9 mEq or 480 mg 30 mg Klucel
    tablet total antacid 25 mg cherry flavor
    10 mg magnesium
    stearate
    2 mg Red #40 Lake
  • PPI Antacid Excipient
    40 mg 8.0 mEq or 400 mg 30 mg sucralose
    micro- Ca(OH)2 40 mg Ac-Di-Sol
    encapsulated 2.9 mEq or 240 mg 30 mg pregela-
    omeprazole NaHCO3 tinized starch
    per chewable 10.9 mEq or 640 mg 30 mg Klucel
    tablet total antacid 40 mg Xylitab
    7 mg mint flavor
    10 mg magnesium
    stearate
  • Example 4 Preparation of Caplets
  • Various caplets are manufactured using the microencapsulate materials described in Examples 1 and 2. The amount of microencapsulated omeprazole used in each caplet batch is based on the actual payload of each set of microcapsules to achieve the theoretical dose. All ingredients are mixed well to achieve a homogenous bulk blend which is then compressed to caplets.
  • Prokinetic
    PPI Buffering Agent Agent Excipient
    40 mg 17.1 mEq or 500 mg 20 mg 20 mg Ac-Di-Sol
    micro- Mg(OH)2 Clebopride 80 mg Klucel
    encapsulated 3.0 mEq or 250 mg per tablet 10 mg magnesium
    omeprazole NaHCO3 stearate
    per caplet 20.1 mEq or 750 mg
    total buffer
  • Prokinetic
    PPI Buffering Agent Agent Excipient
    15 mg 17.1 mEq or 500 mg 10 mg 20 mg Ac-Di-Sol
    micro- Mg(OH)2 Clebopride 80 mg Klucel
    encapsulated 3.0 mEq or 250 mg per tablet 10 mg magnesium
    lansoprazole NaHCO3 stearate
    per caplet 20.1 mEq or 750 mg
    total buffer
  • Prokinetic
    PPI Buffering Agent Agent Excipient
    10 mg 13.7 mEq or 400 mg 10 mg 20 mg Ac-Di-Sol
    micro- Mg(OH)2 Cisapride 80 mg Klucel
    encapsulated 3.0 mEq or 250 mg pre tablet 10 mg magnesium
    omeprazole NaHCO3 stearate
    per caplet 16.7 mEq or 650 mg
    total buffer
  • Prokinetic
    PPI Buffering Agent Agent Excipient
    40 mg 20.6 mEq or 600 mg 10 mg 20 mg Ac-Di-Sol
    micro- Mg(OH)2 Metoclo- 80 mg Klucel
    encapsulated 3.0 mEq or 250 mg pramide 10 mg magnesium
    omeprazole NaHCO3 per tablet stearate
    per caplet 23.6 mEq or 850 mg
    total buffer
  • PPI Buffering Agent NSAID Excipient
    40 mg 17.1 mEq or 500 mg 81 mg 20 mg Ac-Di-Sol
    micro- Mg(OH)2 aspirin 80 mg Klucel
    encapsulated 3.0 mEq or 250 mg per tablet 10 mg magnesium
    omeprazole NaHCO3 stearate
    per caplet 20.1 mEq or 750 mg
    total buffer
  • PPI Buffering Agent NSAID Excipient
    15 mg 17.1 mEq or 500 mg 75 mg 20 mg Ac-Di-Sol
    micro- Mg(OH)2 indo- 80 mg Klucel
    encapsulated 3.0 mEq or 250 mg methacin 10 mg magnesium
    lansoprazole NaHCO3 per tablet stearate
    per caplet 20.1 mEq or 750 mg
    total buffer
  • PPI Buffering Agent NSAID Excipient
    10 mg 13.7 mEq or 400 mg 200 mgs 20 mg Ac-Di-Sol
    micro- Mg(OH)2 Ibupro- 80 mg Klucel
    encapsulated 3.0 mEq or 250 mg pene 10 mg magnesium
    omeprazole NaHCO3 per tablet stearate
    per caplet 16.7 mEq or 650 mg
    total buffer
  • PPI Buffering Agent NSAID Excipient
    40 mg 20.6 mEq or 600 mg 100 mgs 20 mg Ac-Di-Sol
    micro- Mg(OH)2 aspirin 80 mg Klucel
    encapsulated 3.0 mEq or 250 mg per tablet 10 mg magnesium
    omeprazole NaHCO3 stearate
    per caplet 23.6 mEq or 850 mg
    total buffer
  • PPI Buffering Agent NSAID Excipient
    20 mg 20.6 mEq or 600 mg 25 mg 170 mg Xylitab
    micro- Mg(OH)2 rofecoxib 30 mg Ac-Di-Sol
    encapsulated 5.0 mEq or 420 mg per tablet 100 mg Klucel
    omeprazole NaHCO
    3 40 mg Sucralose
    per caplet 25.6 mEq or 1020 mg 25 mg cherry flavor
    total buffer 15 mg magnesium
    stearate
    3 mg Red #40 Lake
  • PPI Buffering Agent NSAID Excipient
    40 mg 24.0 mEq or 700 mg 100 mg 170 mg Dipac sugar
    micro- Mg(OH)2 diclofenac 30 mg Ac-Di-Sol
    encapsulated 7.1 mEq or 600 mg per tablet 120 mg Klucel
    omeprazole NaHCO3 27 mg grape flavor
    per caplet 31.1 mEq or 1300 mg 15 mg magnesium
    total buffer stearate
    1 mg Red #40 Lake
    1 mg Blue #2 Lake
  • PPI Buffering Agent NSAID Excipient
    30 mg 24.0 mEq or 700 mg 400 mg 170 mg Xylitab
    micro- Mg(OH)2 aspirin 30 mg Ac-Di-Sol
    encapsulated 5.0 mEq or 420 mg and 400 mg 100 mg Klucel
    lansoprazole NaHCO3 enteric 25 mg cherry flavor
    per caplet 29.0 mEq or 1120 mg coated 15 mg magnesium
    total buffer aspirin stearate
    per tablet 3 mg Red #40 Lake
  • PPI Buffering Agent NSAID Excipient
    60 mg 15 mEq or 750 mg 600 mg 170 mg Xylitab
    micro- Ca(OH)2 oxaprozin 30 mg Ac-Di-Sol
    encapsulated 15 mEq or 1260 mg per tablet 100 mg Klucel
    omeprazole NaHCO3 25 mg cherry flavor
    per caplet 30 mEq or 2010 mg 15 mg magnesium
    total buffer stearate
    3 mg Red #40 Lake
  • PPI Buffering Agent NSAID Excipient
    40 mg 15 mEq or 750 mg 100 mg 170 mg Xylitab
    micro- Ca(OH)2 aspirin 30 mg Ac-Di-Sol
    encapsulated 10 mEq or 840 mg per tablet 100 mg Klucel
    omprazole NaHCO
    3 15 mg mint flavor
    per caplet 25 mEq or 1590 mg 15 mg magnesium
    total buffer stearate
  • PPI Buffering Agent NSAID Excipient
    40 mg 24.0 mEq or 700 mg 100 mg 60 mg sucralose
    micro- Mg(OH)2 diclofenac 60 mg Ac-Di-Sol
    encapsulated 7.1 mEq or 600 mg per tablet 60 mg pregela-
    omeprazole NaHCO3 tinized starch
    per caplet 31.1 mEq or 1300 mg 30 mg Klucel
    total buffer 27 mg grape flavor
    15 mg magnesium
    stearate
    1 mg Red #40 Lake
    1 mg Blue #2 Lake
  • PPI Buffering Agent NSAID Excipient
    30 mg 17.1 mEq or 500 mg 200 mg 60 mg sucralose
    micro- Mg(OH)2 micro- 60 mg Ac-Di-Sol
    encapsulated 5 mEq or 420 mg encapsulated 70 mg pregela-
    lansoprazole NaHCO3 asprin tinized starch
    per caplet 22.1 mEq or 920 mg per tablet 30 mg Klucel
    total buffer 25 mg cherry flavor
    15 mg magnesium
    stearate
    3 mg Red #40 Lake
  • PPI Buffering Agent NSAID Excipient
    60 mg 15 mEq or 750 mg 100 mg 60 mg sucralose
    micro- Ca(OH)2 ketoprofen 60 mg Ac-Di-Sol
    encapsulated 15 mEq or 1260 mg per tablet 60 mg pregela-
    omeprazole NaHCO3 tinized starch
    per caplet 30 mEq or 2010 mg 30 mg Klucel
    total buffer 25 mg cherry flavor
    15 mg magnesium
    stearate
    3 mg Red #40 Lake
  • PPI Buffering Agent NSAID Excipient
    40 mg 15 mEq or 750 mg 100 mg 60 mg sucralose
    micro- Ca(OH)2 aspirin 60 mg Ac-Di-Sol
    encapsulated 10 mEq or 840 mg per tablet 60 mg pregela-
    omprazole NaHCO3 tinized starch
    per tablet 25 mEq or 1590 mg 30 mg Klucel
    total buffer 15 mg mint flavor
    15 mg magnesium
    stearate
  • Example 5 Preparation of Capsule Containing Microencapsulated Omeprazole
  • Various capsules are manufactured using the microencapsulate materials described in Examples 1 and 2. The amount of microencapsulated omeprazole used in each capsule batch is based on the actual payload of each set of microcapsules to achieve the theoretical dose. All ingredients are mixed well to achieve a homogenous bulk blend which is then filled into a hard gelatin capsule such as a size 00 hard gelatin capsule from Capsugel.
  • Prokinetic
    PPI Buffering Agent Agent Excipient
    15 mg 17.1 mEq or 500 mg 5 mg 30 mg Ac-Di-Sol
    micro- Mg(OH)2 Mosapride 15 mg Klucel
    encapsulated 3.0 mEq or 250 mg per capsule 7 mg magnesium
    lansoprazole NaHCO3 stearate
    per capsule 20.1 mEq or 750 mg
    total buffer
  • Prokinetic
    PPI Buffering Agent Agent Excipient
    40 mg 15.4 mEq or 450 mg 10 mg 30 mg Ac-Di-Sol
    micro- Mg(OH)2 Norcisapride 7 mg magnesium
    encapsulated 2.4 mEq or 200 mg per capsule stearate
    omeprazole NaHCO3
    per capsule 17.8 mEq or 650 mg
    total buffer
  • PPI Antacid Excipient
    40 mg 600 mg Mg(OH)2 50 mg Ac-Di-Sol
    micro- 200 mg NaHCO3 50 mg Klucel
    encapsulated 23.0 mEq or 800 mg 5 mg magnesium
    omeprazole total buffer stearate
    per capsule
  • PPI Antacid Excipient
    15 mg 17.1 mEq or 500 mg 50 mg Ac-Di-Sol
    micro- Mg(OH)2 15 mg Klucel
    encapsulated 3.0 mEq or 250 mg 7 mg magnesium
    lansoprazole NaHCO3 stearate
    per capsule 20.1 mEq or 750 mg 6.0% disintegrant
    total buffer
  • PPI Antacid Excipient
    20 mg 20.6 mEq or 600 mg 50 mg Ac-Di-Sol
    micro- Mg(OH)2 50 mg Klucel
    encapsulated 3.0 mEq or 250 mg 10 mg magnesium
    omeprazole NaHCO3 stearate
    per capsule 23.6 mEq or 850 mg 5.1% disintegrant
    total buffer
  • PPI Antacid Excipient
    15 mg 17.1 mEq or 500 mg 60 mg Ac-Di-Sol
    micro- Mg(OH)2 15 mg Klucel
    encapsulated 3.0 mEq or 250 mg 7 mg magnesium
    lansoprazole NaHCO3 stearate
    per capsule 20.1 mEq or 750 mg 7.1% disintegrant
    total buffer
  • PPI Antacid Excipient
    15 mg 6.9 mEq or 200 mg 35 mg Ac-Di-Sol
    micro- Mg(OH)2 20 mg Klucel
    encapsulated 2.6 mEq or 220 mg 6 mg magnesium
    lansoprazole NaHCO3 stearate
    per capsule 9.5 mEq or 420 mg 7.1% disintegrant
    Size
    1 capsule total buffer
  • PPI Antacid Excipient
    40 mg 15.4 mEq or 450 mg 30 mg Ac-Di-Sol
    micro- Mg(OH)2 7 mg magnesium
    encapsulated 2.4 mEq or 200 mg stearate
    omeprazole NaHCO3
    per capsule 17.8 mEq or 650 mg
    total buffer
  • PPI Antacid Excipient
    15 mg 6.9 mEq or 200 mg 35 mg Ac-Di-Sol
    micro- Mg(OH)2 20 mg Klucel
    encapsulated 2.6 mEq or 220 mg 6 mg magnesium
    lansoprazole NaHCO3 stearate
    per capsule 9.5 mEq or 420 mg Size 1 capsule
    total antacid
  • PPI Antacid Excipient
    10 mg 3.4 mEq or 100 mg 18 mg Ac-Di-Sol
    micro- Mg(OH)2 15 mg Microcrystalline
    encapsulated 3.0 mEq or 250 mg Cellulose (MCC, PH102)
    omeprazole NaHCO 3 7 mg magnesium
    per capsule 6.4 mEq or 350 mg stearate
    total antacid Size 2 capsule
  • PPI Antacid Excipient
    40 mg 3.4 mEq or 100 mg 20 mg Ac-Di-Sol
    micro- Mg(OH)2 5 mg magnesium
    encapsulated 2.4 mEq or 200 mg stearate
    omeprazole NaHCO3 Size 2 capsule
    per capsule 5.8 mEq or 300 mg
    total antacid
  • PPI Buffering Agent NSAID Excipient
    15 mg 17.1 mEq or 500 mg 75 mg 30 mg Ac-Di-Sol
    micro- Mg(OH)2 ketoprofen 15 mg Klucel
    encapsulated 3.0 mEq or 250 mg per capsule 7 mg magnesium
    lansoprazole NaHCO3 stearate
    per capsule 20.1 mEq or 750 mg
    total buffer
  • PPI Buffering Agent NSAID Excipient
    40 mg 15.4 mEq or 450 mg 100 mg enteric 30 mg Ac-Di-Sol
    micro- Mg(OH)2 coated aspirin 7 mg magnesium
    encapsulated 2.4 mEq or 200 mg per capsule stearate
    omeprazole NaHCO3
    per capsule 17.8 mEq or 650 mg
    total buffer
  • Example 6 Analytical Assay for Determining the Amount of Omeprazole Present in Tablets Containing Omeprazole Microspheres
  • The following procedure was used to determine the potency of omeprazole in the tablets. The tablet was accurately weighed and placed into 100 ml volumetric flask. To that, 1.0 ml of Nanopure water was added to wet and soften the tablet. The solution was allowed to stand for 30 minutes. After sitting, the sample was vortexed and sonicated for 30 minutes or until completely dissolved. 1.0 ml of chloroform was then added and the sample was vortexed and sonicated for an additional 15 minutes. The solution was then brought to volume with methanol and vortexed again to mix solution. 10 ml was then decanted into a 10 cc syringe fitted with a 0.45-micron filter. The material was pushed through the filter and the first several milliliters were discarded. The remaining mixture was then collected for HPLC injection. A 5-point calibration curve was prepared in methanol ranging from 15 to 300 μg/ml. The following chromatographic conditions were used: mobile phase: 75.5% Na2PO4, pH=8.0, 24.5% acetonitrile; flow rate: 1.0 mL/min; injection volume: 20 μL; detector: UV, 280 nm; column: waters symmetry shield RP8.
  • Example 7 Stability Study of Microencapsulated Omeprazole
  • Microspheres that exhibited dissolution results with greater than 80% omeprazole release after 2 hours were placed on stability. The microspheres were stored in opened vials at 25° C. All samples showed degradation after 4 weeks at elevated temperatures. The open vials stored at 25° C. were analyzed after 6-8 weeks for potency and for impurities using the Omeprazole EP method. The stability results are summarized in the table 7A below.
  • TABLE 7A
    Omeprazole
    Loading 4-Week Potency Values AUC
    Sample (Initial) (Omeprazole Loading) Purity*
    5 23.3 25.0(107% of initial)@25° C. 95.65
    6 26.0 24.9(95.8% of initial) @25° C. 99.90
    8 24.8 26.4(106.6% of initial)@25° C. 99.95
    10 2.2 2.3 (106% of initial) @25° C. 76.16
    11 20.5 22.6(110% of initial) @25° C. 100.0 
    13 26.2 23.8(90.8% of initial) @25° C. 99.54
    14 21.3 19.1(89.5% of initial) @25° C. 98.88
    15 26.0 22.8(87.8% of initial)@25° C. 99.70
    17 25.8 21.9(84.9% of initial) @25° C. 98.22
    (99.3@T0)
    23 22.2 20.7 (93.2% of initial) @25° C. 97.69
    35 26.0 21.7(83.6% of initial) @25° C. 97.88
    *AUC Purity = Area Under the Curve after 6-8 weeks at 25° C. in open container.
  • Example 8 Method for Determining Payload of Omeprazole Microspheres
  • The HPLC samples for the omeprazole assay of various microspheres were prepared as follows: 5 mg of the microsphere were accurately weighed into a screw cap culture tube. To that, 200 μL of chloroform was added. The microspheres were allowed to dissolve, sonicated and vortex for approximately one minute. Then, 10 ml of methanol was added and the sample was again vortexed for one minute. Once completed, an aliquot of the sample was removed for HPLC analysis.
  • A 5-point calibration curve was prepared in methanol ranging from 20 to 500 μg/mL to calculate payload. The chromatographic conditions were: Mobile phase: 75.5% Na2PO4 pH 8.0, 24.5% Acetonitrile; Flow Rate: 1.0 mL/min; Run Time: 15 min; Injection Volume: 20 μL; Detector: U.V., 280 nm; Column: Waters SymmetryShield RP8.
  • Example 9 Method for Determining the Amount of Impurities Present in the Microspheres
  • The HPLC samples for the omeprazole assay of various microspheres were prepared in the following manner: 5 mgs of the omeprazole microspheres were weighed into a screw cap culture tube. To that, 200 μL of chloroform were added. The microspheres were allowed to dissolve, sonicate and vortex for approximately one minute each. 10 mL of methanol was then added and the sample was again vortexed for 1 minute. Once complete, an aliquot was removed for HPLC analysis.
  • For standards, 100 μg/mL concentration of omeprazole in methanol for a marker was prepared. A 0.1 μg/mL concentration of omeprazole was then prepared to set one-half the minimal detection limit. Then, a 1 μg/mL concentration of omeprazole impurity D in methanol was prepared. The chromatographic conditions were: Mobile Phase: 75% Na2PO4 pH 7.6, 25% acetonitrile; Flow Rate: 1.0 mL/min; Run Time: 30 min; Injection Volume: 20 μL; Detector: U.V., 280 nm; Column: Waters SymmetryShield RP8.
  • Example 10 Method for Determining Dissolution of Omeprazole Microspheres
  • The omeprazole potency method was used for the dissolution testing. The HPLC samples for the omeprazole assay of various microspheres were prepared according to the following method. 5 mgs of the microspheres were accurately weighed into an 8 ounce amber bottle. To that, 100 ml of pH 7.4 monobasic phosphate buffer was added. The samples were placed in a 37° C. water bath and vigorously shaken until the end of the release study. Using an Eppendorf pipette, 100 μL was removed and the outside part of the tip was rinsed with 100 μL of buffer back into the sample bottle. The sample was then transferred into a limited insert for HPLC analysis using a 1 cc syringe fitted with a 45 micron filter. Samples were then taken at 30, 45, and 120 minutes.
  • A 6-point calibration curve was prepared in diluent (70% sodium phosphate pH 10.0/30% acetonitrile) ranging from 1 to 120 μg/mL to determine sample release rates. The chromatographic conditions were: Mobile phase: 75.5% Na2PO4 pH 8.0, 24.5% Acetonitrile; Flow Rate: 1.0 mL/min; Run Time: 15 min; Injection Volume: 20 μL; Detector: U.V., 280 nm; Column: Waters SymmetryShield RP8.
  • Example 11 Optical Microscopy
  • The omeprazole microspheres were observed using an Olympus BX60 optical microscope equipped with an Olympus DP 10 digital camera to determine their particle size and morphology characteristics. The microspheres were observed at either 100× or 200× magnification.
  • The microspheres prepared by spray drying were in the size range of 5 to 30 microns. The microspheres prepared by spinning disk-solvent process were in the size range of 25 to 100 microns. The microspheres prepared by spinning disk-hot melt process were in the size range of 30 to 125 microns.
  • Example 12 Thermal Gravimetric Analysis (TGA)
  • Thermal Gravimetric Analysis was performed on neat omeprazole (Two lots from Uquifa and USP Standard) and the omeprazole microspheres using a TA Instruments Model 2950 equipped with Thermal Solutions Instrument Software and Universal Analysis Data Software. The neat omeprazole samples showed very little weight loss up to 150° C. at which temperature a dramatic weight loss begins. This weight loss occurs at the melting point of omeprazole which is in the range of 150-160° C.
  • For the omeprazole microspheres, the percent weight loss up to 140° C. was recorded to determine the amount of volatiles present. Most samples exhibit a weight loss of less than 1% up to 140° C. except the samples that contained sodium bicarbonate which have a greater weight loss, from 7-32%. The following TGA run conditions were used: nitrogen atmosphere; Isothermal for 5 minutes at 25° C.; ramp 10° C./minute to 250° C.; platinum sample pan.
  • Example 13A Formulation of Placebo Antacid Tablet Used in Example 13G
  • Antacid tablet without any omeprazole was prepared using the components listed in the table below. All ingredients were blended homogeneously in 1 cubic feet V-blender. The resulting blend was compressed into a tablet using a Stokes 16 station rotary tablet press equipped with plain, round ¾″, FFBE punches.
  • Ingredients % Level Mg/tab
    Sodium Bicarbonate, USP #2 39.9 850
    Magnesium Hydroxide, spray dried (95% w/w) 29.6 632
    Hydroxypropyl Cellulose (Klucel ®) 5.1 110
    Croscarmellose Sodium (Ac-Di-Sol ®) 2.1 45
    Xylitab ® 100 12.7 270
    Sucralose ® 5.4 115
    Peach Durarome 2.5 53
    Masking Flavor 1.6 35
    Magnesium Stearate 1.0 22
    TOTALS: 100.0 2132
  • Example 13B Preparation of SAN-15A Used in Example 13G
  • Micronized, non-microencapsulated omeprazole chewable tablets were prepared using the components listed in the table. All ingredients were blended homogeneously in 1 cubic feet V-blender. The resulting blend was compressed into a tablet using a Stokes 16 station rotary tablet press equipped with plain, round ¾″, FFBE punches.
  • Ingredients % Level Mg/tab
    Omeprazole, USP (micronized) 1.9 40.8
    Sodium Bicarbonate, USP #2 39.1 850
    Magnesium Hydroxide, spray dried (95% w/w) 29.0 632
    Hydroxypropyl Cellulose (Klucel ®) 5.1 110
    Croscarmellose Sodium (Ac-Di-Sol ®) 2.1 45
    Xylitab ® 100 12.2 270
    Sucralose ® 5.3 115
    Peach Flavor 2.4 53
    Masking Flavor 1.6 35
    Magnesium Stearate 1.0 22
    FD&C Red Lake #40 0.1 3
    TOTALS: 100.0 2176
  • Example 13C Preparation of SAN-15B Used in Example 13G
  • Microencapsulated omeprazole powder (40 mg) prepared in Example 1C was co-administered with the antacid chewable tablet prepared in Example 13A (total ANC of 30.7 mEq). The powder and the placebo chewable tablet were chewed together in the mouth to simulate administration of 40 mg of microencapsulated omeprazole incorporated in the said placebo tablet.
  • Example 13D Preparation of SAN-15C Used in Example 13G
  • Microencapsulated omeprazole powder (40 mg) prepared in Example 1D was co-administered with an antacid chewable tablet prepared in Example 13A (total ANC of 30.7 mEq). The powder and the placebo chewable tablet were chewed together in the mouth to simulate administration of 40 mg of microencapsulated omeprazole incorporated in the said placebo tablet.
  • Example 13E Preparation of SAN-20D Used in Example 13G
  • Microencapsulated omeprazole powder prepared in Example 1C was blended homogeneously with other ingredients shown in the table. The powder blend was then compressed into a capsule shaped tablet using a Stokes rotary press.
  • SAN-20D
    Ingredients mg/cap %
    Microencapsulated Omeprazole (28% w/w) 142.86 14.1%
    Sodium Bicarbonate #2 280 27.7%
    Magnesium Hydroxide (95%) 473.68 46.9%
    Croscarmellose Sodium 50 4.9%
    Hydroxypropyl Cellulose 55 5.4%
    Magnesium Stearate 9 0.9%
    Total Fill Wt: 1010.5 100%
  • Example 13F Preparation of SAN-20E Used in Example 13G
  • Microencapsulated omeprazole powder prepared in Example 1C was blended homogeneously with other ingredients shown in the table. The powder blend was then compressed into a capsule shaped tablet using a Stokes rotary press.
  • SAN-20E
    Ingredients mg/cap %
    Microencapsulated Omeprazole (28% w/w) 142.86 13.7%
    Sodium Bicarbonate #2 280 26.9%
    Magnesium Hydroxide (95%) 473.68 45.4%
    Croscarmellose Sodium 82 7.9%
    Hydroxypropyl Cellulose 55 5.3%
    Magnesium Stearate 9 0.9%
    Total Fill Wt: 1042.5 100%
  • Example 13G Pharmacokinetic Study of Non-Enteric Coated Omeprazole 40 mg Chewable Tablets and Prilosec® Delayed-Release Capsules 40 mg
  • This trial was conducted as an open-label, single-dose, crossover trial, with each subject receiving up to twelve different oral omeprazole formulations, one in each of the twelve treatment periods. Each dose was followed by a minimum 7-day washout. Omeprazole was administered at a dose of 40 mg. The compositions administered are set forth in Examples 13A-13F and Table 13.G.1. All formulations were administered with about 120 ml (4 oz) of water after an overnight fast and 1 hour prior to a standardized, high-fat breakfast. Within a given treatment period, the same treatment was administered to all subjects. The non-enteric coated formulation study drugs were compared to Prilosec® which contained enteric-coated granules.
  • TABLE 13.G.1
    The pharmacokinetic release trial with omeprazole dosage forms.
    (All dosage forms contained 40 mg omeprazole)
    Period Study Material
     1 Prilosec (40 mg omeprazole)
     3 (SAN - 15A) SAN - 15A Tablet, 30.7 mEq (850 mg SB &
    600 mg MH) 40 mg “Non-micro-
    encapsulated” OME & MS95 MH
     5 (SAN - 15B) Placebo Antacid Tablet, 30.7 mEq (850 mg SB &
    600 mg MH) with 40 mg microencapsulated
    omeprazole with Klucel shell material
     7 (SAN - 15C) Placebo Antacid Tablet, 30.7 mEq (850 mg SB &
    600 mg MH) with 40 mg microencapsulated
    omeprazole with Methocel shell material
    10 (SAN - 20D) SAN - 20D Caplet, 18.8 mEq (280 mg SB & 450 mg
    MH) 40 mg microencapsulated omeprazole
    12 (SAN - 20E) SAN - 20E Caplet, 18.8 mEq (280 mg SB & 450 mg
    MH) 40 mg microencapsulated omeprazole
  • Volunteers were screened for up to 14 days before baseline measurements of blood plasma levels of omeprazole. In each period, a standardized high-fat breakfast was given in the clinic 1 hour after dosing of omeprazole. Blood samples for determination of plasma omeprazole concentrations were collected for 12 hours post treatment.
  • This trial was designed to assess the pharmacokinetics of immediate-release omeprazole chewable tablets and caplets versus the Prilosec® 40 mg delayed-release formulation. The duration of the trial for each subject was approximately 24 weeks, including up to 14 days for screening and a minimum 7 day wash-out period between omeprazole doses. Data from 12 healthy male subjects were expected to provide adequate power to assess pharmacokinetics and safety using descriptive statistics. The descriptive statistics were assessed using the pharmacokinetic parameters: Tmax, Cmax, AUC(0-t), and AUC(0-inf).
  • The treatments administered to subjects in this trial are listed in Table 13.G.1, above. In general, the treatment protocol entailed a 14 day assessment period, followed by a first period (Period 1) in which Prilosec® 40 mg delayed release capsule was administered to the subjects, after an overnight fast, and 1 hour prior to a standardized high-fat breakfast. Plasma sampling was conducted for 6 hour post-dose. Period 1 was followed by a 7-14 day washout period, during which the plasma levels of omeprazole were expected to decrease to a steady baseline. The other periods listed in Tablet 13.G.1 were conducted in a similar manner to Period 1, except substituting a dosage form according to the invention for the delayed-release formulation used in Period 1.
  • Pharmacokinetic Sampling, Analytical Methods, and Parameters
  • Blood samples (3 mL) were obtained by venipuncture within 30 minutes before each dose and at 0, 5, 10, 15, 20, 30, 45, 60, 90, 120, 180, 240, 300, 360 minutes (6 hours) after delivery of each dose during each trial period. Zero time was the time that the subject swallowed a capsule, caplet or chewable tablet of trial drug.
  • Plasma omeprazole concentrations were measured using a previously validated liquid chromatography mass spectrometry (LC-MSIMS) assay (MDS Pharma Services, Lincoln, Nebr.). The linear assay range was 5.0 to 750 ng/mL. The following pharmacokinetic parameters were measured for each subject:
      • Plasma omeprazole concentration at each sampling time
      • Peak omeprazole plasma concentration (Cmax) and the time at which Cmax is observed (Tmax) obtained directly from the data without interpolation
      • Terminal elimination rate constant (Kel) determined from a log-linear regression analysis of the terminal plasma omeprazole concentrations
      • Half-life of drug elimination (T %) calculated as 0.693/Kel
      • Area under the plasma drug time-concentration curve calculated from 0 time to last time point evaluated [AUC(0-t)] calculated using the trapezoidal rule with the plasma concentration at time t being the last measurable concentration
      • Area under the plasma drug time-concentration curve calculated from 0 time and extrapolated to infinity [AUC(0-inf)] calculated as AUC(0-t)+Ct/Kel, where Ct is the last measurable plasma concentration (at time t) and Kel is the terminal elimination rate constant defined above.
    Pharmacokinetic Analysis
  • For the analysis of data collected on Day 1 of each period (pre-meal dosing), an analysis of variance (ANOVA) model was used to test the bioequivalence of each of the tested drug formulations. The model included the following factors: treatment, period, sequence, and subject nested within sequence. Ninety percent confidence intervals (CIs) for treatment differences were calculated; the endpoints of these CIs were then reverse transformed to represent CIs about the percent mean ratios on the original scale. With respect to AUC(0-inf) and Cmax, equivalence was declared for each parameter if the bounds of the 90% CIs for the percent mean ratio, comparing a composition according to the invention (Periods 2-12) with Prilosec, were between 80% and 125%.
  • Pharmacokinetic Results
  • Pharmacokinetic results for this study are presented in Table 13.G.2 and FIGS. 3-10.
  • TABLE 13.G.2
    Intervals (Minutes)
    ng/mL Period Day 0 5 10 15 20 30 45 60
    Mean Prilosec (P1) 1 0 0 0 0 2 20 205 485
    Min 1 0 0 0 0 0 0 37 104
    Max 1 0 0 0 0 15 80 543 1474
    Mean Chew Tab (P3) (SAN-15A) 1 0 80 642 921 1001 1092 939 800
    Min 1 0 0 73 149 188 177 145 132
    Max 1 0 352 1321 1779 2073 2705 2571 2311
    Mean Chew Tab (P5) (SAN-15B) 1 0 53 473 820 956 958 897 856
    Min 1 0 0 36 330 428 379 227 167
    Max 1 0 150 1073 1770 2110 2327 2848 3097
    Mean Chew Tab (P7) (SAN-15C) 1 0 65 706 1396 1386 1230 1048 852
    Min 1 0 0 191 473 711 515 321 226
    Max 1 0 382 1172 2807 2306 2580 2358 2117
    Mean Caplet (P10) (SAN-20D) 1 0 56 184 269 199 213 235 322
    Min 1 0 0 5 23 28 49 48 56
    Max 1 0 440 1256 1518 744 595 469 1013
    Mean Caplet (P12) (SAN-20E) 1 0 5 43 81 116 327 540 583
    Min 1 0 0 8 38 64 46 36 50
    Max 1 0 30 98 135 199 1507 1687 1856
    (continued)
    Cmax Tmax
    ng/mL Period Day 90 120 180 240 300 360 N (ng/mL) (hr)
    Mean Prilosec (P1) 1 991 679 417 265 203 143 12 1061 1.38
    Min 1 241 127 37 8 8 0 12 273 1.00
    Max 1 2994 2146 1596 1093 1081 707 12 2994 1.50
    Mean Chew Tab (P3) 1 597 441 280 193 129 95 12 1201 0.54
    Min 1 71 42 9 0 0 0 12 196 0.50
    Max 1 2133 1701 1322 999 709 587 12 2705 1.50
    Mean Chew Tab (P5) 1 633 467 305 226 148 122 12 1192 0.50
    Min 1 70 29 14 7 0 0 12 428 0.17
    Max 1 2586 2058 1532 1279 788 744 12 3097 1.00
    Mean Chew Tab (P7) 1 610 436 295 217 161 112 11 1550 0.33
    Min 1 67 31 7 0 0 0 11 711 0.25
    Max 1 2244 1538 1279 1069 760 596 11 2807 0.50
    Mean Caplet (P10) 1 699 598 430 271 178 136 8 111 1.59
    Min 1 143 97 42 21 10 6 8 531 0.25
    Max 1 1440 1850 1404 1036 749 633 8 1850 3.00
    Mean Caplet (P12) 1 909 531 320 196 143 110 8 1083 1.59
    Min 1 93 111 28 10 0 0 8 159 0.75
    Max 1 1951 1315 1099 795 648 553 8 1951 3.00
  • Example 14A Preparation of SAN-15 20 mg Chewable Tablets
  • Microencapsulation of Omeprazole
  • Omeprazole was microencapsulated as described in Example 1B.
  • Preparation of Chewable Tablet
  • An omeprazole pre-blend containing microencapsulated omeprazole, antacid excipients and other formulation components was prepared. A flavor pre-blend containing sensory components was then prepared. The main blend was then prepared by combining the omeprazole and flavor pre-blends. Magnesium stearate was then added to the main blend and mixed to form a final blend. All blending operations were carried out in appropriately sized V-blenders. Blend uniformity was ensured by testing at various stages of blending. The final blend was then compressed on a high speed rotary tablet press to form the final tablets. The tablet press was a Stokes® instrumented tablet press using less than a full set of tablet tooling in order to conserve powder. A compression force of 50 kN using ¾″ round FFBE tooling gave an acceptable tablet harness and friability in all prototype batches. The amount of each component is listed below in Table 14.A.2.
  • TABLE 14.A.2
    Ingredient Quantity/20 mg Tablet
    Microencapsulated Omeprazole 55.6 mg*
    Sodium Bicarbonate 600 mg
    Magnesium Hydroxide (95% w/w) 736.8 mg**
    Hydroxypropyl Cellulose 90 mg
    Croscarmellose Sodium 33 mg
    Xylitol
    200 mg
    Sucralose
    80 mg
    Peach Durarome 52 mg
    Peppermint Durarome 10 mg
    Masking Flavor 27 mg
    Magnesium Stearate 17 mg
    Red #40 Lake Dye 2 mg
    Total Weight/Unit 1902 mg
    *Spray-dried omeprazole (37% w/w) includes a 2% omeprazole overage in the blend manufacture that helps ensure label claim amount of omeprazole in the final product
    **Spray-dried magnesium hydroxide (95% w/w with 5% starch) equivalent to 700 mg of active magnesium hydroxide
  • Example 14B Clinical Trial with SAN-15 (20 mg) Chewable Tablet
  • The primary objective was to show that SAN-15 20 mg Chewable Tablets are pharmacokinetically bioequivalent to Prilosec 20 mg with respect to area under the curve.
  • This was an open-label, randomized, 2-period crossover trial to evaluate the pharmacokinetics, pharmacodynamics, and safety of seven consecutive daily doses of SAN-15 chewable tablets (omeprazole immediate-release chewable tablets) 20 mg compared to seven consecutive daily doses of Prilosec Delayed-Release Capsules 20 mg in healthy subjects. A comparison of pharmacokinetic parameters for SAN-15 chewable tablets administered before versus after a meal was also conducted.
  • Volunteers were screened within 21 days before baseline measurements (e.g. gastric pH, vital signs). Gastric pH was recorded for 24 hours before the first dose of trial drug. In Period 1, subjects received SAN-15 chewable tablets 20 mg or Prilosec 20 mg, as randomized, 1 hour before breakfast for seven consecutive days. A standardized high-fat breakfast was given in the clinic 1 hour after dosing on Days 1 and 7, or 1 hour after water for baseline assessment. Standardized lunch and dinner were also given 5 and 10 hours post dose at Baseline and on Days 1 and 7 in the clinic. Blood samples to determine plasma omeprazole concentrations were collected for 12 hours and gastric pH was measured for 24 hours after the doses on Days 1 and 7. Subjects who had received SAN-15 chewable tablets 20 mg in Period 1 were given an eighth dose on Day 8 in Period 1, 1 hour after the start of the standardized high-fat breakfast. Blood samples were collected for 12 hours after the eighth dose. After a 10- to 14-day washout period, subjects returned for Period 2 and received the alternate treatment from that received in Period 1. Procedures in Period 2 were identical to those in Period 1 except that no eighth dose of SAN-15 chewable tablets 20 mg was given.
  • Number of Subjects:
  • Thirty-five subjects were dosed and 34 subjects completed the trial. Thirty-four subjects were included in the pharmacokinetic analyses and 29 subjects were included in the pharmacodynamic analyses for Days 1 and 7.
  • Treatments:
  • The treatments administered to subjects are listed in the table below:
  • Treatment Treatment Description
    SAN-15 SAN-15 chewable tablets (omeprazole immediate-release
    chewable tablets) 20 mg to be administered orally and
    chewed for 30 seconds, followed by drinking 120 mL
    water each morning after an overnight fast, 1 hour
    before starting breakfast.
    Prilosec Prilosec Capsules (omeprazole delayed-release capsules)
    20 mg to be administered orally with 120 mL water each
    morning after an overnight fast, 1 hour before starting
    breakfast.
  • Pharmacokinetic Sampling, Analytical Methods, and Parameters
  • Blood samples (3 mL) were obtained by venipuncture on Days 1 and 7 of both periods and Day 8 of Period 1 (for SAN-15 chewable tablets) within 30 minutes before each dose, and at 5, 10, 15, 20, 30, 45, 60, 90, 120, 150, 180, 210, 240, 300, 360, 420, 480, 540, 600, 660, and 720 minutes (12 hours) after each dose. Zero time was the time that the subject ingested a chewable tablet or swallowed a capsule of Prilosec.
  • Plasma omeprazole concentrations were measured using a validated liquid chromatography mass spectrometry (LC-MS/MS) assay (MDS Pharma Services, Lincoln, Nebr.). The linear assay range was 5.0 to 750 ng/mL. The following pharmacokinetic parameters were measured for each subject:
      • Plasma omeprazole concentration at each sampling time
      • Peak omeprazole plasma concentration (Cmax) and the time at which Cmax is observed (Tmax) obtained directly from the data without interpolation
      • Terminal elimination rate constant (kel) determined from a log-linear regression analysis of the terminal plasma omeprazole concentrations
      • Half-life of drug elimination (T ½) calculated as 0.693/Kel
      • Area under the plasma drug time-concentration curve from 1 time to last time point evaluated [AUC(0-0] calculated using the trapezoidal rule with the plasma concentration at time “t” being the last measurable concentration
      • Area under the plasma drug time-concentration curve from 1 time and extrapolated to infinity [AUC(0-inf)] calculated as AUC(0-t)+Ct/Kel, where Ct is the last measurable plasma concentration and Kel is the terminal elimination rate constant defined above.
        The Pharmacokinetic results are shown below in Tables 14.B. and 14.C.
  • TABLE 14B
    SAN-15 Chewable Tablets 20 mg
    Interval (Minutes)
    ng/mL Day 0 5 10 15 20 30 45 60 90 120 150
    Mean 1 0 48 223 423 487 455 351 298 221 116 77
    Min 1 0 0 21 36 80 94 91 55 22 8 0
    Max 1 0 336 646 1150 1420 1270 1280 1200 1100 618 499
    Mean 7 0 60 266 435 606 610 572 496 437 268 191
    Min 7 0 0 30 74 129 152 170 158 55 21 10
    Max 7 0 386 1120 1620 2010 1590 1510 1310 1230 696 548
    SAN-15 Chewable Tablets 20 mg (Continued)
    Interval (Minutes) Cmax Tmax
    ng/mL Day 180 210 240 300 360 420 480 540 600 660 720 N (ng/mL) (hr)
    Mean 1 55 40 28 15 9 6 4 2 1 1 1 34 594 0.48
    Min 1 0 0 0 0 0 0 0 0 0 0 0 34 117 0.17
    Max 1 421 347 276 179 131 87 62 40 26 17 12 34 1420 1.50
    Mean 7 144 108 83 49 29 16 11 6 4 2 22 34 769 0.71
    Min 7 6 0 0 0 0 0 0 0 0 0 00 34 198 0.17
    Max 7 478 404 361 262 196 139 104 75 53 43 31 34 2010 1.50
  • TABLE 14C
    Prilosec Delayed-Release Capsules 20 mg
    Interval (Minutes)
    ng/mL Day 0 5 10 15 20 30 45 60 90 120 150
    Mean 1 0 0 0 0 3 26 103 176 367 232 149
    Min 1 0 0 0 0 0 0 0 0 0 35 17
    Max 1 0 0 0 5 91 459 513 1180 1580 719 552
    Mean 7 0 0 0 1 5 100 232 325 506 360 273
    Min 7 0 0 0 0 0 0 0 0 85 68 48
    Max 7 0 0 0 11 83 1070 1180 1110 1740 1220 952
    Prilosec Delayed-Release Capsules 20 mg (Continued)
    Interval (Minutes) Cmax Tmax
    ng/mL Day 180 210 240 300 360 420 480 540 600 660 720 N (ng/mL) (hr)
    Mean 1 97 66 46 25 16 8 6 4 3 1 1 34 433 1.48
    Min 1 7 0 0 0 0 0 0 0 0 0 0 34 107 0.33
    Max 1 461 419 332 253 178 115 85 63 55 32 23 34 1580 2.50
    Mean 7 198 149 115 67 40 25 15 10 4 4 3 34 583 1.50
    Min 7 31 18 11 0 0 0 0 0 0 0 0 34 177 0.75
    Max 7 667 583 530 363 263 195 146 104 74 65 44 34 1740 3.00
  • Example 15A Preparation of SAN-15 40 mg Chewable Tablets
  • Microencapsulation of Omeprazole
  • Omeprazole was microencapsulated as described in Example 1B.
  • Preparation of Chewable Tablet
  • The procedure used to make SAN-15 40 mg Chewable Tablet was the same as the one described in Example 14A except that the following ingredients were used:
  • TABLE 15.A.2
    Ingredient Quantity/40 mg Tablet
    Microencapsulated Omeprazole 110*
    Sodium Bicarbonate 600 mg
    Magnesium Hydroxide (95% w/w) 736.8 mg**
    Hydroxypropyl Cellulose 90 mg
    Croscarmellose Sodium 33 mg
    Xylitol
    200 mg
    Sucralose 80 mg
    Peach Durarome 52 mg
    Peppermint Durarome 10 mg
    Masking Flavor 27 mg
    Magnesium Stearate 17 mg
    Red #40 Lake Dye 2 mg
    Total Weight/Unit 1902 mg
    *Spray-dried omeprazole (37% w/w) includes a 2% omeprazole overage in the blend manufacture that helps ensure label claim amount of omeparzole in the final product
    **Spray-dried magnesium hydroxide (95% w/w with 5% starch) equivalent to 700 mg of active magnesium hydroxide
  • Example 15B Clinical Trial with SAN-15 (40 mg) Chewable Tablet
  • Trial design, design rationale, treatments, timing of dose for each subject, pharmacokinetic sampling, analytical methods, and parameters, and pharmacodynamic parameters and methodology were the same for this trial as was in the above Example 14.B with the exception of the use of SAN-15 40 mg chewable tablets and Prilosec Delayed-Release Capsules 40 mg in this trial verses 20 mg of each in the above example. The Pharmacokinetic results are shown below in Tables 15.B. and 15.C.
  • TABLE 15B
    SAN-15 Chewable Tablets 40 mg
    Intervals (Minutes)
    ng/mL Day 0 5 10 15 20 30 45 60 90 120 150
    Mean 1 0 83 396 705 970 1062 907 727 575 349 244
    Min 1 0 0 13 81 164 175 240 140 51 19 11
    Max 1 0 885 1740 2170 2300 2470 2160 1940 1910 1600 1110
    Mean 7 0 82 458 736 1090 1443 1394 1316 1266 857 662
    Min 7 0 8 55 94 172 288 532 411 248 129 60
    Max 7 10 304 1740 2010 2230 2970 2530 2450 2450 1840 1420
    SAN-15 Chewable Tablets 40 mg (Continuation)
    Intervals (Minutes) Cmax Tmax
    ng/mL Day 180 210 240 300 360 420 480 540 600 660 720 N (ng/mL) (hr)
    Mean 1 189 141 120 83 45 32 24 18 12 9 7 35 1272 0.58
    Min 1 0 0 0 0 0 0 0 0 0 0 0 35 403 0.25
    Max 1 1180 881 1060 1040 445 414 399 329 237 197 163 35 2470 1.50
    Mean 7 519 419 333 219 150 98 65 39 29 19 14 35 1763 0.77
    Min 7 32 15 9 0 0 0 0 0 0 0 0 35 933 0.25
    Max 7 1330 1160 920 755 589 458 338 277 211 148 137 35 2970 1.50
  • TABLE 15C
    Prilosec Delayed-Release Capsules 40 mg
    Intervals (Minutes)
    ng/mL Day 0 5 10 15 20 30 45 60 90 120 150
    Mean 1 0 0 0 0 2 40 219 445 829 494 356
    Min 1 0 0 0 0 0 0 0 10 45 34 40
    Max 1 0 0 0 8 49 354 1200 1950 2210 1760 1650
    Mean 7 1 1 1 1 28 232 600 862 1296 1005 790
    Min 7 0 0 0 0 0 0 0 0 201 216 183
    Max 7 18 15 16 15 748 1970 2050 2360 2510 2060 1810
    Prilosec Delayed-Release Capsules 40 mg (Continuation)
    Intervals (Minutes) Cmax Tmax
    ng/mL Day 180 210 240 300 360 420 480 540 600 660 720 N (ng/mL) (hr)
    Mean 1 267 231 166 118 84 53 36 27 22 16 12 35 938 1.54
    Min 1 17 10 0 0 0 0 0 0 0 0 0 35 274 0.75
    Max 1 1400 1330 1140 882 985 624 476 420 397 332 260 35 2210 3.50
    Mean 7 615 507 408 282 189 129 85 60 42 28 20 35 1417 1.50
    Min 7 116 66 37 14 6 0 0 0 0 0 0 35 408 0.50
    Max 7 1370 1280 1060 855 600 505 390 327 278 200 218 35 2510 5.00
  • Example 16A Preparation of Immediate Release Chewable Tablet Using Dry-Coated Omeprazole
  • Dry-coated omeprazole are prepared by mixing the ingredients listed in table 16A-1 and dry-granulate the powder using roller compactor. Resulting granules are screened through a 10-mesh screen.
  • TABLE 16A-1
    Dry Granule Ingredient for dry-coated omeprazole (20% w/w)
    Mg/unit % Level Weight/batch
    Omeprazole, USP (micronized) 20 mg 20.0% 2.0 Kg
    Sodium Bicarbonate, USP #2 30 mg 30.0% 3.0 Kg
    Klucel (HPC) 10 mg 10.0% 1.0 Kg
    Xylitab 100 39.7 mg 39.7% 4.0 Kg
    Magnesium Stearate 0.3 mg  0.3% 0.03 Kg
    Total Dry Coated API Granules 100 mg  100% 10 Kg
  • The dry granules are then mixed with the components shown in table 16A-2 until a homogeneous blend is obtained. This blend is then compressed using a 18 mm diameter tooling in a Stokes rotary press.
  • TABLE 16-A-2
    Chewable Tablet using dry-coated omeprazole (40 mg dose)
    Mg/unit Weight/batch
    Omeprazole Dry Granule (20% w/w) 200 2.0 Kg
    Sodium Bicarbonate, USP #2 390 3.9 Kg
    Magnesium Hydroxide, 95% 745.3 7.45 Kg
    Klucel (HPC) 80 0.8 Kg
    Croscarmellose sodium 28 0.28 Kg
    Xylitab 100 151 1.51 Kg
    Sucralose 80 0.8 Kg
    Masking Flavor 30 0.3 Kg
    Strawberry flavor 60 0.6 Kg
    Magnesium Stearate 13 0.13 Kg
    Total 1777 17.77 Kg
  • Example 16B Preparation of Immediate Release Chewable Tablet Using Dry-Coated Omeprazole
  • Dry-coated omeprazole are prepared by mixing the ingredients listed in the table 16B-1 and dry-granulate the powder using roller compactor. Resulting granules are screened through a 10-mesh screen.
  • TABLE 16B-1
    Dry Granule Ingredient for dry-coated omeprazole (10% w/w)
    Mg/unit % Level
    Omeprazole 10 mg 10.0%
    Sodium Bicarbonate 25 mg 25.0%
    Klucel (HPC) 10 mg 10.0%
    Xylitab 100 54.7 mg 54.7%
    Magnesium Stearate 0.3 mg 0.3%
    Total Dry Coated API Granules 100 mg 100%
  • The dry granules are then mixed with the components shown in table 16B-2 until a homogeneous blend is obtained. This blend is then compressed using a 18 mm diameter tooling in a Stokes rotary press.
  • TABLE 16-B-2
    Chewable Tablet using dry-coated omeprazole (40 mg dose)
    Mg/unit Weight/batch
    Omeprazole Dry Granule (10% w/w) 400 2.0 Kg
    Sodium Bicarbonate, USP #2 380 1.9 Kg
    Magnesium Hydroxide, 95% 745.3 3.73 Kg
    Klucel (HPC) 60 0.3 Kg
    Croscarmellose sodium 28 0.14 Kg
    Xylitab 100 11 0.06 Kg
    Sucralose 80 0.4 Kg
    Masking Flavor 30 0.15 Kg
    Chocolate flavor 70 0.35 Kg
    Magnesium Stearate 13 0.07 Kg
    Total 1817 9.09 Kg
  • Example 16C Preparation of Immediate Release Caplet Using Dry-Coated Omeprazole
  • Dry-coated omeprazole are prepared by mixing the ingredients listed in the table 16C-1 and dry-granulate the powder using roller compactor. Resulting granules are screened through a 10-mesh screen.
  • TABLE 16C-1
    Dry Granule Ingredient for dry-coated omeprazole (10% w/w)
    Mg/unit % Level
    Omeprazole, USP (micronized) 10 mg 10%
    Sodium Bicarbonate 85 mg 85%
    Klucel (HPC) 5 mg 5%
    Magnesium Stearate 0.3 mg 0.3%
    Total Dry Coated API Granules 100.3 mg 100%
  • The dry granules prepared in Example 16C-1 are mixed with the ingredients shown in Table 16C-2 until a homogeneous blend is obtained. This blend is then compressed using oval-shaped tooling in a Stokes rotary press.
  • TABLE 16-C-2
    Caplet using dry-coated omeprazole (40 mg dose)
    Mg/unit Weight/batch
    Omeprazole Dry Granule (10% w/w) 400 4 Kg
    Sodium Bicarbonate, USP #2 160 1.6 Kg
    Magnesium Oxide, USP 280 2.8 Kg
    Klucel (HPC) 10 0.1 Kg
    Croscarmellose sodium 65 0.65 Kg
    Magnesium Stearate 8 0.08 Kg
    Total 923 9.23 Kg
  • Many modifications, equivalents, and variations of the present invention are possible in light of the above teachings, therefore, it is to be understood that within the scope of the appended claims, the invention may be practiced other than as specifically described.

Claims (39)

What is claimed is:
1. A pharmaceutical formulation comprising (a) at least one non-enteric coated acid-labile proton pump inhibitor that is dry coated, and (b) at least one antacid;
wherein an initial serum concentration of the proton pump inhibitor is greater than about 0.1 μg/ml within about 30 minutes after administration of the pharmaceutical formulation to a subject.
2. A pharmaceutical formulation according to claim 1, wherein at least some of the antacid is used to dry coat the proton pump inhibitor.
3. A pharmaceutical formulation according to claim 1, wherein part of the antacid is used to dry coat the proton pump inhibitor and part of the antacid is combined with the dry coated proton pump inhibitor.
4. A pharmaceutical formulation according to claim 3, wherein the antacid combined with the dry coated proton pump inhibitor comprises at least one antacid that is different from the antacid used to dry coat the proton pump inhibitor.
5. A pharmaceutical formulation according to claim 2, wherein the antacid used to dry coat the proton pump inhibitor comprises an alkaline metal salt or a Group IA metal selected from a bicarbonate salt of a Group IA metal, a carbonate salt of a Group IA metal.
6. A pharmaceutical formulation according to claim 5, wherein the material used to dry coat the proton pump inhibitor further comprises sodium bicarbonate or sodium carbonate.
7. A pharmaceutical formulation according to claim 2, wherein the material used to dry coat the proton pump inhibitor further comprises a binder.
8. A pharmaceutical formulation according to claim 6, wherein the material used to dry coat the proton pump inhibitor further comprises a sweetener selected from sucralose, xylitol or mixtures thereof.
9. A pharmaceutical formulation according to claim 1, wherein the proton pump inhibitor is a substituted bicyclic aryl-imidazole selected from the group consisting of omeprazole, hydroxyomeprazole, esomeprazole, tenatoprazole, lansoprazole, pantoprazole, rabeprazole, dontoprazole, habeprazole, perprazole, ransoprazole, pariprazole, leminoprazole; or a free base, free acid, salt, hydrate, ester, amide, enantiomer, isomer, tautomer, polymorph, or prodrug thereof.
10. A pharmaceutical formulation according to claim 1, wherein the proton pump inhibitor is selected from omeprazole, or a free base, free acid, salt, hydrate, polymorph, or prodrug thereof.
11. A pharmaceutical formulation according to claim 1, wherein the proton pump inhibitor is selected from lansoprazole, or a free base, free acid, salt, hydrate, polymorph, or prodrug thereof.
12. A pharmaceutical formulation according to claim 1, wherein the proton pump inhibitor is selected from esomeprazole, or a free base, free acid, salt, hydrate, polymorph, or prodrug thereof.
13. A pharmaceutical formulation according to claim 1 comprising about 5 mgs to about 200 mgs of the proton pump inhibitor.
14. A pharmaceutical formulation according to claim 1 comprising about 10 mgs, or about 15 mgs, or about 20 mgs, or about 30 mgs, or about 40 mgs, or about 60 mgs, or about 80 mgs of the proton pump inhibitor.
15. A pharmaceutical formulation according to claim 1, wherein the antacid is selected from sodium bicarbonate, sodium carbonate, calcium carbonate, magnesium oxide, potassium bicarbonate, magnesium hydroxide, magnesium carbonate, aluminum hydroxide, and mixtures thereof.
16. A pharmaceutical formulation according to claim 1, wherein the antacid comprises at least one soluble buffer.
17. A pharmaceutical formulation according to claim 16, wherein the soluble buffer is present in at least about 5 mEq.
18. A pharmaceutical formulation according to claim 1, wherein the antacid is present in an amount of between about 5 mEq to about 60 mEq.
19. A pharmaceutical formulation according to claim 1 comprising about 500 to about 2000 mg of antacid.
20. A pharmaceutical formulation according to claim 1, wherein the pharmaceutical formulation has less than 5% total new impurities after 1 year of storage at room temperature.
21. A pharmaceutical formulation according to claim 1 in the form of a capsule, a chewable tablet, a tablet, a caplet, or a capsule.
22. A pharmaceutical formulation according to claim 1, wherein the proton pump inhibitor is dry coated using a dry granulation technique.
23. A pharmaceutical formulation according to claim 1, wherein the proton pump inhibitor is dry coated using a nano particle coating technique.
24. A pharmaceutical formulation according to claim 1, wherein serum concentration of the proton pump inhibitor is greater than about 0.5 μg/ml within about 1 hour after administration of the pharmaceutical formulation to a subject.
25. A pharmaceutical formulation according the claim 1, wherein the average particle size of the dry blended proton pump inhibitor is between about 100 to about 1000 microns in diameter.
26. A pharmaceutical formulation according the claim 1, wherein the average particle size of the dry blended proton pump inhibitor is between about 100 to about 1000 microns in diameter.
27. A pharmaceutical formulation according the claim 1, wherein the average particle size of the dry blended proton pump inhibitor is between about 300 to about 700 microns in diameter.
28. A pharmaceutical formulation according to claim 1, wherein a maximum serum concentration is reached within about 45 minutes after administration of the pharmaceutical formulation to a fasting subject.
29. A pharmaceutical formulation according to claim 1, wherein a maximum serum concentration is reached within about 60 minutes after administration of the pharmaceutical formulation to a fasting subject.
30. A pharmaceutical formulation according to claim 1, wherein the drug loading of the proton pump inhibitor in the dry coated granules is greater than about 10 wt-% of proton pump inhibitor.
31. A pharmaceutical formulation according to claim 1, wherein the drug loading of the proton pump inhibitor in the dry coated granules is between about 5 wt-% to about 40 wt-% of proton pump inhibitor.
32. A method of treating an acid related gastrointestinal disorder in a subject in need thereof by administering the pharmaceutical formulation of claim 1.
33. A chewable tablet comprising:
(a) between about 10-80 mgs of a proton pump inhibitor which is microencapsulated with a material comprising a cellulose hydroxypropyl ether;
(b) between about 10 mEq and about 100 mEq of antacid selected from sodium bicarbonate, sodium carbonate, calcium carbonate, magnesium oxide, potassium bicarbonate, magnesium hydroxide, magnesium carbonate, and mixtures thereof; and
(c) at least one flavoring agent;
wherein the drug loading of the proton pump inhibitor into microspheres is between about 20 wt-% to about 50 wt-% of proton pump inhibitor to microencapsulated proton pump inhibitor.
34. A chewable tablet according to claim 33, wherein the flavoring agent comprises peach, menthol, aspartame, sucralose, sucrose, xylitol, mint, or a mixture thereof.
35. A chewable tablet according to claim 33, wherein the antacid is a mixture of sodium bicarbonate and magnesium hydroxide.
36. A chewable tablet according to claim 33, wherein the drug loading of the proton pump inhibitor into microspheres is about 37 wt-% of proton pump inhibitor.
37. A chewable tablet according to claim 33, wherein a serum concentration of the proton pump inhibitor is greater than about 0.5 μg/ml within about 1 hour after administration of the pharmaceutical formulation to a subject.
38. A chewable tablet according to claim 33, wherein upon administration of the chewable tablet to a fasted subject, the maximum serum concentration of the proton pump inhibitor is reached within about 45 minutes after administration.
39. A chewable tablet according to claim 33, wherein greater than about 70% of the proton pump inhibitor is released from the composition within about 1.5 hours after exposure to gastrointestinal fluid.
US14/673,670 2003-07-18 2015-03-30 Pharmaceutical formulations useful for inhibiting acid secretion and methods for making and using them Abandoned US20150202226A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US14/673,670 US20150202226A1 (en) 2003-07-18 2015-03-30 Pharmaceutical formulations useful for inhibiting acid secretion and methods for making and using them

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US48832103P 2003-07-18 2003-07-18
US10/893,203 US20050037070A1 (en) 2003-07-18 2004-07-16 Pharmaceutical formulatins useful for inhibiting acid secretion and methods for making and using them
US11/338,608 US8993599B2 (en) 2003-07-18 2006-01-24 Pharmaceutical formulations useful for inhibiting acid secretion and methods for making and using them
US14/673,670 US20150202226A1 (en) 2003-07-18 2015-03-30 Pharmaceutical formulations useful for inhibiting acid secretion and methods for making and using them

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US11/338,608 Continuation US8993599B2 (en) 2003-07-18 2006-01-24 Pharmaceutical formulations useful for inhibiting acid secretion and methods for making and using them

Publications (1)

Publication Number Publication Date
US20150202226A1 true US20150202226A1 (en) 2015-07-23

Family

ID=34138652

Family Applications (2)

Application Number Title Priority Date Filing Date
US11/338,608 Expired - Fee Related US8993599B2 (en) 2003-07-18 2006-01-24 Pharmaceutical formulations useful for inhibiting acid secretion and methods for making and using them
US14/673,670 Abandoned US20150202226A1 (en) 2003-07-18 2015-03-30 Pharmaceutical formulations useful for inhibiting acid secretion and methods for making and using them

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US11/338,608 Expired - Fee Related US8993599B2 (en) 2003-07-18 2006-01-24 Pharmaceutical formulations useful for inhibiting acid secretion and methods for making and using them

Country Status (1)

Country Link
US (2) US8993599B2 (en)

Families Citing this family (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060165793A1 (en) * 2003-08-04 2006-07-27 Koji Ukai Pharmaceutical preparation to be dispersed before administration
US20080103169A1 (en) 2006-10-27 2008-05-01 The Curators Of The University Of Missouri Compositions comprising acid labile proton pump inhibiting agents, at least one other pharmaceutically active agent and methods of using same
US20080166423A1 (en) * 2007-01-06 2008-07-10 Renjit Sundharadas Combination Medication for Treating the Effects of Stomach Acid Reduction Medication on Bone Integrity
US20080194307A1 (en) * 2007-02-13 2008-08-14 Jeff Sanger Sports-based game of chance
CA2716367C (en) 2008-02-20 2015-05-26 The Curators Of The University Of Missouri Composition comprising a combination of omeprazole and lansoprazole, and a buffering agent, and methods of using same
EP2296632A4 (en) * 2008-07-14 2014-11-12 Otonomy Inc Controlled-release apoptosis modulating compositions and methods for the treatment of otic disorders
WO2011083402A2 (en) * 2010-01-11 2011-07-14 Muneera Mohamed Shafee Immediate release compositions of acid labile drugs
US10632113B2 (en) * 2014-02-05 2020-04-28 Kashiv Biosciences, Llc Abuse-resistant drug formulations with built-in overdose protection
WO2015155281A1 (en) * 2014-04-11 2015-10-15 Sanovel Ilac Sanayi Ve Ticaret A.S. Pharmaceutical combinations of dabigatran and proton pump inhibitors
EP3288556A4 (en) 2015-04-29 2018-09-19 Dexcel Pharma Technologies Ltd. Orally disintegrating compositions
US10674746B2 (en) 2015-10-27 2020-06-09 Cytozyme Animal Nutrition, Inc. Animal nutrition compositions and related methods
BR112018007434A2 (en) 2015-10-27 2018-10-23 Cytozyme Animal Nutrition Inc animal nutrition compositions, uses and related methods
WO2017145146A1 (en) 2016-02-25 2017-08-31 Dexcel Pharma Technologies Ltd. Compositions comprising proton pump inhibitors
US10076494B2 (en) 2016-06-16 2018-09-18 Dexcel Pharma Technologies Ltd. Stable orally disintegrating pharmaceutical compositions

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040248942A1 (en) * 2003-02-20 2004-12-09 Bonnie Hepburn Novel formulation, omeprazole antacid complex-immediate release for rapid and sustained suppression of gastric acid

Family Cites Families (275)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS469580Y1 (en) 1968-01-27 1971-04-05
JPS469581Y1 (en) 1968-07-12 1971-04-05
JPS529674B2 (en) 1972-04-13 1977-03-17
JPS495967A (en) 1972-05-23 1974-01-19
JPS5529992B2 (en) 1972-06-03 1980-08-07
JPS4920174A (en) 1972-06-22 1974-02-22
JPS4920173A (en) 1972-06-22 1974-02-22
JPS4941198A (en) 1972-09-11 1974-04-17
JPS4993537A (en) 1973-01-06 1974-09-05
JPS5544755B2 (en) 1973-01-24 1980-11-13
JPS5736278B2 (en) 1973-09-12 1982-08-03
US4045564A (en) 1974-02-18 1977-08-30 Ab Hassle Benzimidazole derivatives useful as gastric acid secretion inhibitors
JPS50112373A (en) 1974-02-27 1975-09-03
JPS5840546B2 (en) 1974-05-14 1983-09-06 ウェルファイド株式会社 2- Benzimidazole carbamic acid alkyl ester
JPS5116669A (en) 1974-07-29 1976-02-10 Yoshitomi Pharmaceutical 22 aminobenzuimidazoorujudotaino seizoho
JPS5117268A (en) 1974-08-01 1976-02-12 Ikeda Bussan Co Jidoshayonaisozaino seizohoho
JPS5214776A (en) 1975-07-21 1977-02-03 Yoshitomi Pharmaceut Ind Ltd Process for preparing cyclohexane derivatives
JPS51131875A (en) 1975-05-12 1976-11-16 Yoshitomi Pharmaceut Ind Ltd A process for preparing novel pyrrolidine derivatives
JPS525769A (en) 1975-07-02 1977-01-17 Yoshitomi Pharmaceut Ind Ltd Preparation of 3-substituted pyrolidine derivatives
JPS52102416A (en) 1976-02-19 1977-08-27 Okawara Mfg Granule making method of herb medicine and like
JPS5297978A (en) 1976-06-02 1977-08-17 Yoshitomi Pharmaceut Ind Ltd Preparation of alicyclic derivatives
JPS5359675A (en) 1976-11-08 1978-05-29 Yoshitomi Pharmaceut Ind Ltd Aminoalcohol derivatives
IN148930B (en) * 1977-09-19 1981-07-25 Hoffmann La Roche
SE7804231L (en) * 1978-04-14 1979-10-15 Haessle Ab Gastric acid secretion
JPS5519211A (en) 1978-07-27 1980-02-09 Mitsui Toatsu Chem Inc Purification of organic amine
JPS5661311A (en) 1979-10-25 1981-05-26 Fujisawa Pharmaceut Co Ltd Thial amide or prolonged pharmaceutical preparation comprising its salt
JPS6043064B2 (en) 1979-11-28 1985-09-26 ウェルファイド株式会社 New isoxazole derivative
US4359465A (en) 1980-07-28 1982-11-16 The Upjohn Company Methods for treating gastrointestinal inflammation
JPS57212180A (en) 1981-06-19 1982-12-27 Yoshitomi Pharmaceut Ind Ltd Tetrahydrofuran(thiophene) compound
US4472409A (en) 1981-11-05 1984-09-18 Byk Gulden Lomberg Chemische Fabrik Gesellschaft Mit Beschrankter Haftung 2-Pyridylmethyl thio(sulfinyl)benzimidazoles with gastric acid secretion inhibiting effects
SE8204879D0 (en) 1982-08-26 1982-08-26 Haessle Ab NOVEL CHEMICAL INTERMEDIATES
JPS5995997A (en) 1982-11-22 1984-06-02 Hitachi Plant Eng & Constr Co Ltd Biologically dephosphorizing method of waste water
SE8300736D0 (en) 1983-02-11 1983-02-11 Haessle Ab NOVEL PHARMACOLOGICALLY ACTIVE COMPOUNDS
SE8301182D0 (en) * 1983-03-04 1983-03-04 Haessle Ab NOVEL COMPOUNDS
RO88351A3 (en) 1984-04-26 1986-04-30 Intreprinderea De Medicamente"Terapia",Ro COMPOSITION FOR THE TREATMENT OF GASTRODUODENAL DISEASES
SE8403179D0 (en) * 1984-06-13 1984-06-13 Haessle Ab NEW COMPOUNDS
JPS6150978A (en) 1984-08-16 1986-03-13 Takeda Chem Ind Ltd Pyridine derivative and preparation thereof
JPS61221117A (en) 1985-03-26 1986-10-01 Fujisawa Pharmaceut Co Ltd Enteric hard capsule
US5246714A (en) 1985-10-11 1993-09-21 Aktiebolaget Hassle Drug preparation
JPS62145083A (en) 1985-12-18 1987-06-29 Yoshitomi Pharmaceut Ind Ltd Condensed imidazole derivative
JPS62277322A (en) 1986-02-13 1987-12-02 Takeda Chem Ind Ltd Stabilized pharmaceutical composition and production thereof
CA1327010C (en) * 1986-02-13 1994-02-15 Tadashi Makino Stabilized solid pharmaceutical composition containing antiulcer benzimidazole compound and its production
US5433959A (en) 1986-02-13 1995-07-18 Takeda Chemical Industries, Ltd. Stabilized pharmaceutical composition
GB2189699A (en) 1986-04-30 1987-11-04 Haessle Ab Coated acid-labile medicaments
GB2189698A (en) 1986-04-30 1987-11-04 Haessle Ab Coated omeprazole tablets
JPH0791273B2 (en) 1986-06-02 1995-10-04 日本ケミフア株式会社 Benzimidazole derivative pharmaceutical composition
AU619444B2 (en) 1986-06-02 1992-01-30 Nippon Chemiphar Co. Ltd. 2-(2-aminobenzylsulfinyl)- benzimidazole derivatives
JPH0643426B2 (en) * 1986-07-25 1994-06-08 東京田辺製薬株式会社 Imidazo [4,5-b] pyridine derivative, method for producing the same, and antiulcer agent containing the same
SE8604566D0 (en) * 1986-10-27 1986-10-27 Haessle Ab NOVEL COMPUNDS
FI90544C (en) 1986-11-13 1994-02-25 Eisai Co Ltd Process for Preparation as Drug Useful 2-Pyridin-2-yl-methylthio- and sulfinyl-1H-benzimidazole derivatives
US5215974A (en) * 1986-11-21 1993-06-01 Aktiebolaget Hassle Certain pyridyl[(methylthio- or methyl sulfinyl)-2 benzimidazol-2-yl]N-methyl phosphonates useful for treating gastric-acid secretion related diseases
US5026560A (en) * 1987-01-29 1991-06-25 Takeda Chemical Industries, Ltd. Spherical granules having core and their production
DE3887417T2 (en) 1987-03-25 1994-05-26 Yoshitomi Pharmaceutical THIENOCINNOLINE COMPOUNDS AND THEIR USE AS A MEDICINE.
NZ224252A (en) * 1987-04-21 1991-09-25 Erba Carlo Spa An anthracycline glycoside and its preparation
WO1989000566A1 (en) 1987-07-21 1989-01-26 Yoshitomi Pharmaceutical Industries, Ltd. Pyridine compounds and medicinal use thereof
ATE84213T1 (en) 1987-11-11 1993-01-15 Pharmascience Lab PHARMACEUTICAL PREPARATION CONTAINING EXIFON AND A WATER SOLUBLE POLYMER.
GB8809421D0 (en) * 1988-04-21 1988-05-25 Fordonal Sa Antacid compositions with prolonged gastric residence time
AU3683889A (en) * 1988-06-30 1990-01-23 Upjohn Company, The Transdermal antisecretory agents for gastrointestinal disease
JPH0737383B2 (en) 1988-07-11 1995-04-26 エーザイ株式会社 Solid formulation containing gastric acid secretion inhibitor
US5075323A (en) 1988-08-24 1991-12-24 Aktiebolaget Hassle Compounds including omeprazole in the treatment of glaucoma
WO1990003380A1 (en) 1988-09-21 1990-04-05 Yoshitomi Pharmaceutical Industries, Ltd. Thienocycloheptapyridazine compounds and medicinal uses thereof
NZ230747A (en) 1988-09-30 1992-05-26 Bror Morein Immunomodulating matrix comprising a complex of at least one lipid and at least one saponin; certain glycosylated triterpenoid saponins derived from quillaja saponaria molina
SE8804629D0 (en) * 1988-12-22 1988-12-22 Ab Haessle NEW THERAPEUTICALLY ACTIVE COMPOUNDS
IE64199B1 (en) * 1988-12-22 1995-07-12 Haessle Ab Compound with gastric acid inhibitory effect and process for its preparation
SE8804628D0 (en) * 1988-12-22 1988-12-22 Ab Haessle NEW COMPOUNDS
JP2694361B2 (en) 1989-02-09 1997-12-24 アストラ アクチエボラグ Antibacterial agent
DE69014141T2 (en) * 1989-02-10 1995-05-24 Takeda Chemical Industries Ltd Use of benzimidazole derivatives as antibacterial agents.
GB8909793D0 (en) 1989-04-28 1989-06-14 Beecham Group Plc Pharmaceutical formulation
JPH0334967A (en) 1989-06-29 1991-02-14 Yoshitomi Pharmaceut Ind Ltd Butane compound and salt thereof and pharmaceutical use thereof
JPH0348680A (en) 1989-07-18 1991-03-01 Yoshitomi Pharmaceut Ind Ltd Antimicrobial agent
JPH0352887A (en) 1989-07-20 1991-03-07 Yoshitomi Pharmaceut Ind Ltd Pyridine compound
SE8903563D0 (en) * 1989-10-26 1989-10-26 Haessle Ab A NOVEL DISSOLUTION SYSTEM
US5204118A (en) * 1989-11-02 1993-04-20 Mcneil-Ppc, Inc. Pharmaceutical compositions and methods for treating the symptoms of overindulgence
KR930000861B1 (en) * 1990-02-27 1993-02-08 한미약품공업 주식회사 Omeprazole rectal composition
CA2038716A1 (en) 1990-03-28 1991-09-29 Yoshio Ueda Stabilizer for 4-ethyl-2-hydroxyimino-5-nitro-3-hexenamide-containing preparation, stabilizing method thereof and drug stabilized thereby
SE9002043D0 (en) * 1990-06-07 1990-06-07 Astra Ab IMPROVED METHOD FOR SYNTHESIS
WO1991019712A1 (en) * 1990-06-20 1991-12-26 Aktiebolaget Astra Dialkoxy-pyridinyl-benzimidazole derivatives, process for their preparation and their pharmaceutical use
ATE145208T1 (en) 1990-07-06 1996-11-15 Yoshitomi Pharmaceutical CONDENSED THIOPHENE COMPOUNDS AND THEIR USE
EP0548103B1 (en) 1990-09-14 2001-12-05 Byk Gulden Lomberg Chemische Fabrik Gmbh Use of pyridylmethylsulphinyl-1h-benzimidazole derivates in the treatment of illnesses caused by helicobacter bacteria
DE4035455A1 (en) 1990-11-08 1992-05-14 Byk Gulden Lomberg Chem Fab ENANTIOMER SEPARATION
JPH0825905B2 (en) 1990-11-20 1996-03-13 武田薬品工業株式会社 Stabilizing agent for pharmaceutical solid composition and stabilizing method
US5232706A (en) 1990-12-31 1993-08-03 Esteve Quimica, S.A. Oral pharmaceutical preparation containing omeprazol
ES2024993A6 (en) 1990-12-31 1992-03-01 Genesis Para La Investigacion Procedure for obtaining an oral pharmaceutical preparation containing omeprazole
WO1992012976A1 (en) 1991-01-16 1992-08-06 Yoshitomi Pharmaceutical Industries, Ltd. Use of pyridine compound as selective drug and novel pyridine compound
US5244670A (en) 1991-04-04 1993-09-14 The Procter & Gamble Company Ingestible pharmaceutical compositions for treating upper gastrointestinal tract distress
SE502569C2 (en) 1991-05-31 1995-11-13 British Tech Group Use of an immunologically inert matrix of a sterol and saponins which can form spherical nanoparticles of narrow size distribution such as drug carriers, particles, composition and kits
YU48263B (en) 1991-06-17 1997-09-30 Byk Gulden Lomberg Chemische Fabrik Gmbh. PROCEDURE FOR OBTAINING PANTOPRAZOLE PHARMACEUTICAL PRODUCT
US5464632C1 (en) 1991-07-22 2001-02-20 Prographarm Lab Rapidly disintegratable multiparticular tablet
WO1993005770A1 (en) 1991-09-20 1993-04-01 Fujisawa Pharmaceutical Co., Ltd. Long-acting preparation
NZ244301A (en) * 1991-09-20 1994-08-26 Merck & Co Inc Preparation of 2-pyridylmethylsulphinylbenzimidazole and pyridoimidazole derivatives from the corresponding sulphenyl compounds
JP2975189B2 (en) 1991-10-11 1999-11-10 株式会社レオテック Die casting method of aluminum alloy
JPH05117268A (en) 1991-10-22 1993-05-14 Yoshitomi Pharmaceut Ind Ltd Pyridine compound
JPH05255088A (en) 1991-11-05 1993-10-05 Yoshitomi Pharmaceut Ind Ltd Pharmaceutical preparation containing antiulcer agent
JPH05194225A (en) 1991-11-07 1993-08-03 Yoshitomi Pharmaceut Ind Ltd Stabilized antiulcer agent-containing preparation
TW224049B (en) * 1991-12-31 1994-05-21 Sunkyong Ind Ltd
IS3990A (en) * 1992-04-24 1993-10-25 Ab Astra Method of mixing substances that inhibit gastric acid and bacterial degrading agent in an acidic environment
US5504082A (en) * 1992-06-01 1996-04-02 Yoshitomi Pharmaceutical Industries, Ltd. Pyridine compound and pharmaceutical compostions
FR2692146B1 (en) * 1992-06-16 1995-06-02 Ethypharm Sa Stable compositions of gastro-protected omeprazole microgranules and process for obtaining them.
SE9201930D0 (en) 1992-06-24 1992-06-24 Astra Ab GASTRIC ANTIBACTERIAL TREATMENT
AU4513393A (en) 1992-07-17 1994-02-14 Astra Aktiebolag Pharmaceutical composition containing antiulcer agent
JPH0733659A (en) 1992-07-17 1995-02-03 Yoshitomi Pharmaceut Ind Ltd Antiulcer agent-containing pharmaceutical preparation
US5447918A (en) 1992-07-27 1995-09-05 Mccullough; Ricky W. Gastrointestinal anti-irritant composition comprising sucralfate and methods of use
JP3954115B2 (en) 1992-07-28 2007-08-08 アストラゼネカ・アクチエボラーグ Injection and injection kit
ATE144421T1 (en) 1992-07-28 1996-11-15 Astra Ab INJECTABLE DRUG AND PACKAGE CONTAINING OMOPRAZOLE OR RELATED COMPOUNDS
JPH05194224A (en) 1992-09-17 1993-08-03 Yoshitomi Pharmaceut Ind Ltd Stabilized antiulcer agent-containing preparation
JPH06100449A (en) 1992-09-18 1994-04-12 Yoshitomi Pharmaceut Ind Ltd Antibacterial agent
SE9301489D0 (en) * 1993-04-30 1993-04-30 Ab Astra VETERINARY COMPOSITION
SE9301830D0 (en) 1993-05-28 1993-05-28 Ab Astra NEW COMPOUNDS
US5877192A (en) * 1993-05-28 1999-03-02 Astra Aktiebolag Method for the treatment of gastric acid-related diseases and production of medication using (-) enantiomer of omeprazole
SE9302396D0 (en) * 1993-07-09 1993-07-09 Ab Astra A NOVEL COMPOUND FORM
SE9302395D0 (en) 1993-07-09 1993-07-09 Ab Astra NEW PHARMACEUTICAL FORMULATION
CA2128820A1 (en) 1993-07-27 1995-01-28 Walter G. Gowan, Jr. Rapidly disintegrating pharmaceutical dosage form and process for preparation thereof
TW359614B (en) * 1993-08-31 1999-06-01 Takeda Chemical Industries Ltd Composition containing benzimidazole compounds for rectal administration
ES2145102T3 (en) 1993-09-09 2000-07-01 Takeda Chemical Industries Ltd FORMULATION COMPRISING AN ANTIBACTERIAL SUBSTANCE AND AN ANTI-ULCER SUBSTANCE.
US5935600A (en) 1993-09-10 1999-08-10 Fuisz Technologies Ltd. Process for forming chewable quickly dispersing comestible unit and product therefrom
US5965162A (en) 1993-09-10 1999-10-12 Fuisz Technologies Ltd. Process for forming chewable quickly dispersing multi-vitamin preparation and product therefrom
WO1995007913A1 (en) 1993-09-17 1995-03-23 Meiji Milk Products Co., Ltd. Antibacterial and antifungal agent
AU7833194A (en) 1993-09-20 1995-04-10 Procter & Gamble Company, The Use of triclosan phosphates for the treatment of gastrointestinal disorders due to heliobacter infection
PT723777E (en) 1993-10-12 2002-11-29 Mitsubishi Pharma Corp COMPRESSED CONTAINING ENTERIC GRANULOS
TW280770B (en) 1993-10-15 1996-07-11 Takeda Pharm Industry Co Ltd
US5374730A (en) 1993-11-04 1994-12-20 Torcan Chemical Ltd. Preparation of omeprazole and lansoprazole
IS4232A (en) 1993-12-06 1995-06-07 Astra Aktiebolag Substituted benzimidazole, its methods of manufacture and pharmacological use
US5714505A (en) * 1994-01-05 1998-02-03 Astra Aktiebolag Method for treatment of psoriasis, by omeprazole or related compounds
DE670160T1 (en) 1994-03-01 1996-03-14 Gerhard Gergely An effervescent system and a granular product or tablet containing an active pharmaceutical ingredient, and a method for their production.
TNSN95062A1 (en) 1994-05-27 1996-02-06 Astra Ab NEW DIALKOXY-PYRIDINYLE-BENZIMIDAZOLE DERIVATIVES
WO1995032957A1 (en) 1994-05-27 1995-12-07 Astra Aktiebolag Novel ethoxycarbonyloxymethyl derivatives of substituted benzimidazoles
JPH0821446A (en) * 1994-07-06 1996-01-23 Matsushita Electric Ind Co Ltd Smooth surface part
DK0723436T3 (en) 1994-07-08 2001-11-26 Astrazeneca Ab Tabulated multi-unit dosage form
SE9402431D0 (en) 1994-07-08 1994-07-08 Astra Ab New tablet formulation
BR9508261A (en) 1994-07-08 1997-12-23 Astra Ab Enterically coated oral formulation using the same process to prepare the formulation to inhibit gastric acid secretion in mammals and humans and to treat gastric acid-related diseases in humans and humans and cross-pressure blister pack
GB2290965A (en) 1994-07-11 1996-01-17 Therapicon Srl Multiple layer capsules for drugs
SE504459C2 (en) 1994-07-15 1997-02-17 Astra Ab Process for the preparation of substituted sulfoxides
AUPM695294A0 (en) 1994-07-20 1994-08-11 Borody, Thomas Julius Improved combination therapy dose unit
SE9404192D0 (en) 1994-12-02 1994-12-02 Astra Ab New process
US5628981A (en) 1994-12-30 1997-05-13 Nano Systems L.L.C. Formulations of oral gastrointestinal diagnostic x-ray contrast agents and oral gastrointestinal therapeutic agents
SE9500422D0 (en) 1995-02-06 1995-02-06 Astra Ab New oral pharmaceutical dosage forms
SE9500478D0 (en) 1995-02-09 1995-02-09 Astra Ab New pharmaceutical formulation and process
AU5780696A (en) 1995-06-02 1996-12-18 Takeda Chemical Industries Ltd. Stabilized composition comprising an antiulcerative benzimid azole
HRP960232A2 (en) 1995-07-03 1998-02-28 Astra Ab A process for the optical purification of compounds
US6132768A (en) 1995-07-05 2000-10-17 Byk Gulden Lomberg Chemische Fabrik Gmbh Oral pharmaceutical composition with delayed release of active ingredient for reversible proton pump inhibitors
US5945124A (en) 1995-07-05 1999-08-31 Byk Gulden Chemische Fabrik Gmbh Oral pharmaceutical composition with delayed release of active ingredient for pantoprazole
SE9503102D0 (en) 1995-09-08 1995-09-08 Astra Ab Aseptic transfer
US5824339A (en) 1995-09-08 1998-10-20 Takeda Chemical Industries, Ltd Effervescent composition and its production
SE9503143D0 (en) 1995-09-12 1995-09-12 Astra Ab New preparation
JPH0987110A (en) 1995-09-20 1997-03-31 Yoshitomi Pharmaceut Ind Ltd Composition containing 2-thiocyanopyridine-1-oxide
HUP9900028A3 (en) * 1995-10-17 1999-11-29 Astrazeneca Ab Pharmaceutically active quinazoline compounds, their production, use thereof and medicament containing them
SE521100C2 (en) 1995-12-15 2003-09-30 Astra Ab Process for the preparation of a benzimidazole compound
US6645988B2 (en) 1996-01-04 2003-11-11 Curators Of The University Of Missouri Substituted benzimidazole dosage forms and method of using same
US6489346B1 (en) 1996-01-04 2002-12-03 The Curators Of The University Of Missouri Substituted benzimidazole dosage forms and method of using same
US5840737A (en) 1996-01-04 1998-11-24 The Curators Of The University Of Missouri Omeprazole solution and method for using same
US20050054682A1 (en) * 1996-01-04 2005-03-10 Phillips Jeffrey O. Pharmaceutical compositions comprising substituted benzimidazoles and methods of using same
US6699885B2 (en) * 1996-01-04 2004-03-02 The Curators Of The University Of Missouri Substituted benzimidazole dosage forms and methods of using same
SE512835C2 (en) 1996-01-08 2000-05-22 Astrazeneca Ab Dosage form containing a plurality of units all containing acid labile H + K + ATPase inhibitors
SE9600070D0 (en) 1996-01-08 1996-01-08 Astra Ab New oral pharmaceutical dosage forms
SE9600071D0 (en) * 1996-01-08 1996-01-08 Astra Ab New oral formulation of two active ingredients I
SE9600072D0 (en) 1996-01-08 1996-01-08 Astra Ab New oral formulation of two active ingredients II
EP0799616A1 (en) 1996-04-01 1997-10-08 Takeda Chemical Industries, Ltd. Oral composition comprising a fumagillol derivative
ID18663A (en) * 1996-04-12 1998-04-30 Novartis Ag COMPOSITION OF PHARMACEUTICAL PLATED PHARMACEUTICALS
DE69713948D1 (en) * 1996-04-23 2002-08-22 Janssen Pharmaceutica Nv Rapidly releasing pH-independent solid dosage forms containing cisapride
SE508669C2 (en) 1996-04-26 1998-10-26 Astra Ab New procedure
SE9602442D0 (en) 1996-06-20 1996-06-20 Astra Ab Administration of pharmaceuticals
US6169102B1 (en) * 1996-06-25 2001-01-02 Takeda Chemical Industries, Ltd. Oxazolone derivatives and their use as anti-Helicobacter pylori agent
DE19626045C2 (en) 1996-06-28 1998-12-03 Klinge Co Chem Pharm Fab A stable dosage form for oral administration containing omeprazole as the active ingredient and methods of making the same
JPH1017470A (en) 1996-07-03 1998-01-20 Yoshitomi Pharmaceut Ind Ltd Medicine combination containing pyridine
JPH1017471A (en) 1996-07-04 1998-01-20 Yoshitomi Pharmaceut Ind Ltd Medicine combination containing pyridine compound
DE19628643A1 (en) 1996-07-16 1998-01-22 Merck Patent Gmbh Materials for the manufacture of transport containers
WO1998006385A1 (en) * 1996-08-15 1998-02-19 Losan Pharma Gmbh Easy to swallow oral medicament composition
US5972389A (en) 1996-09-19 1999-10-26 Depomed, Inc. Gastric-retentive, oral drug dosage forms for the controlled-release of sparingly soluble drugs and insoluble matter
SE9603725D0 (en) 1996-10-11 1996-10-11 Astra Ab New teatment
SE510666C2 (en) 1996-12-20 1999-06-14 Astra Ab New Crystal Modifications
IT1289522B1 (en) 1996-12-24 1998-10-15 Zambon Spa PROCESS FOR THE PREPARATION OF ETEROARIL-FENILALANINE
SE9700885D0 (en) 1997-03-12 1997-03-12 Astra Ab New pharmaceutical formulation
US5885594A (en) * 1997-03-27 1999-03-23 The Procter & Gamble Company Oral compositions having enhanced mouth-feel
JP2001524131A (en) 1997-05-09 2001-11-27 セイジ、ファーマスーティカルズ、インク Stable oral pharmaceutical dosage form
US5939091A (en) 1997-05-20 1999-08-17 Warner Lambert Company Method for making fast-melt tablets
TW580397B (en) 1997-05-27 2004-03-21 Takeda Chemical Industries Ltd Solid preparation
SE9702000D0 (en) 1997-05-28 1997-05-28 Astra Ab New pharmaceutical formulation
SE510650C2 (en) 1997-05-30 1999-06-14 Astra Ab New association
SE510643C2 (en) 1997-06-27 1999-06-14 Astra Ab Thermodynamically stable omeprazole sodium form B
SE9702533D0 (en) 1997-07-01 1997-07-01 Astra Ab New oral formulation
HN1998000115A (en) 1997-08-21 1999-06-02 Warner Lambert Co SOLID PHARMACEUTICAL DOSAGE FORMS
US6096340A (en) 1997-11-14 2000-08-01 Andrx Pharmaceuticals, Inc. Omeprazole formulation
SE9704183D0 (en) 1997-11-14 1997-11-14 Astra Ab New process
AU1671799A (en) 1997-11-28 1999-06-16 Byk Gulden Lomberg Chemische Fabrik Gmbh Medicament preparation in the form of a tablet or pellet for acid-labile active substances
DE19752843C2 (en) 1997-11-28 2003-01-09 Byk Gulden Lomberg Chem Fab Pharmaceutical preparation in tablet or pellet form for pantoprazole and omeprazole
SI1037634T1 (en) 1997-12-08 2006-02-28 Altana Pharma Ag Oral administration form comprising a proton pump inhibitor (e.g.pantoprazole)
US5955107A (en) 1997-12-12 1999-09-21 Fmc Corporation Pharmaceutical suspension tablet compositions
SE9704869D0 (en) 1997-12-22 1997-12-22 Astra Ab New pharmaceutical formulaton II
SE9704870D0 (en) 1997-12-22 1997-12-22 Astra Ab New pharmaceutical formulation I
US6365180B1 (en) * 1998-01-20 2002-04-02 Glenn A. Meyer Oral liquid compositions
FR2774288B1 (en) * 1998-01-30 2001-09-07 Ethypharm Sa GASTROPROTEGED OMEPRAZOLE MICROGRANULES, PROCESS FOR OBTAINING AND PHARMACEUTICAL PREPARATIONS
IT1299198B1 (en) 1998-03-05 2000-02-29 Nicox Sa NITRATED SALTS OF ANTI-ULCER DRUGS
US6235311B1 (en) * 1998-03-18 2001-05-22 Bristol-Myers Squibb Company Pharmaceutical composition containing a combination of a statin and aspirin and method
CA2298823C (en) * 1998-04-20 2011-06-07 Eisai Co., Ltd. Stabilized composition comprising a benzimidazole type compound
JP4127740B2 (en) 1998-04-20 2008-07-30 エーザイ・アール・アンド・ディー・マネジメント株式会社 Stabilized benzimidazole compound-containing composition
SE9801526D0 (en) 1998-04-29 1998-04-29 Astra Ab New compounds
EP2263660B1 (en) 1998-05-18 2017-09-27 Takeda Pharmaceutical Company Limited Orally disintegrable tablets
GB2338896B (en) 1998-07-02 2003-05-21 Reckitt & Colmann Prod Ltd Chewable Capsules
EP1561458B1 (en) 1998-07-28 2010-09-15 Takeda Pharmaceutical Company Limited Rapidly disintegrable solid preparation
US6166213A (en) 1998-08-11 2000-12-26 Merck & Co., Inc. Omeprazole process and compositions thereof
WO2000009092A1 (en) 1998-08-12 2000-02-24 Byk Gulden Lomberg Chemische Fabrik Gmbh Oral administration form for pyridin-2-ylmethylsulfinyl-1h-benzimidazoles
SE9802793D0 (en) 1998-08-21 1998-08-21 Astra Ab New compounds
US6319513B1 (en) 1998-08-24 2001-11-20 The Procter & Gamble Company Oral liquid mucoadhesive compounds
AU5504599A (en) 1998-09-10 2000-04-03 Nycomed Danmark A/S Quick release pharmaceutical compositions of drug substances
US6047829A (en) * 1998-09-18 2000-04-11 Westvaco Corporation Unit dose packaging system (UDPS) having a child resistant locking feature
AU1907100A (en) 1998-10-30 2000-05-22 Curators Of The University Of Missouri, The Omeprazole solution and method of using same
SE9803772D0 (en) 1998-11-05 1998-11-05 Astra Ab Pharmaceutical formulation
UA72748C2 (en) 1998-11-10 2005-04-15 Astrazeneca Ab A novel crystalline form of omeprazole
US6984404B1 (en) * 1998-11-18 2006-01-10 University Of Florida Research Foundation, Inc. Methods for preparing coated drug particles and pharmaceutical formulations thereof
WO2000028975A2 (en) 1998-11-18 2000-05-25 Astrazeneca Ab Improved chemical process and pharmaceutical formulation
SE9804003D0 (en) 1998-11-23 1998-11-23 Astra Ab A method of producing drug particles
SE9804314D0 (en) 1998-12-14 1998-12-14 Astra Ab New pharmaceutical formulation
JP2000212180A (en) 1999-01-21 2000-08-02 Welfide Corp Quinoline compound
SE9900274D0 (en) 1999-01-28 1999-01-28 Astra Ab New compound
GB0000710D0 (en) 1999-02-06 2000-03-08 Zeneca Ltd Drug combination
BR0008403A (en) 1999-02-23 2002-01-29 Merck & Co Inc Pharmaceutical formulation for oral administration
US6294192B1 (en) * 1999-02-26 2001-09-25 Lipocine, Inc. Triglyceride-free compositions and methods for improved delivery of hydrophobic therapeutic agents
US6248363B1 (en) * 1999-11-23 2001-06-19 Lipocine, Inc. Solid carriers for improved delivery of active ingredients in pharmaceutical compositions
GB9911017D0 (en) 1999-05-13 1999-07-14 Zeneca Ltd Pharmaceutical compositions
TWI275587B (en) * 1999-06-17 2007-03-11 Takeda Chemical Industries Ltd A crystal of (R)-2-[[[3-methyl-4-(2,2,2-trifluoroethoxy)-2-pyridyl]methyl]sulfinyl]-1H-benzimidazole
JP3437124B2 (en) 1999-06-18 2003-08-18 積水化成品工業株式会社 Conductive polymer gel, gel pad and bioelectrode using the same
CA2374760A1 (en) * 1999-06-18 2000-12-28 Takeda Chemical Industries, Ltd. Quickly disintegrating solid preparations
SE9902386D0 (en) 1999-06-22 1999-06-22 Astra Ab New formulation
US6555139B2 (en) * 1999-06-28 2003-04-29 Wockhardt Europe Limited Preparation of micron-size pharmaceutical particles by microfluidization
EP1191025B1 (en) 1999-06-30 2005-06-22 Takeda Pharmaceutical Company Limited Crystals of lansoprazole
EP1210091A4 (en) 1999-07-12 2009-07-29 Smithkline Beecham Corp Heartburn treatment
US6500459B1 (en) 1999-07-21 2002-12-31 Harinderpal Chhabra Controlled onset and sustained release dosage forms and the preparation thereof
JP2001040006A (en) 1999-07-29 2001-02-13 Shin Etsu Chem Co Ltd Method for producing polymer of monomer having ethylenic double bond
US6262085B1 (en) 1999-08-26 2001-07-17 Robert R. Whittle Alkoxy substituted Benzimidazole compounds, pharmaceutical preparations containing the same, and methods of using the same
US6268385B1 (en) 1999-08-26 2001-07-31 Robert R. Whittle Dry blend pharmaceutical formulations
US6262086B1 (en) 1999-08-26 2001-07-17 Robert R. Whittle Pharmaceutical unit dosage form
US6369087B1 (en) 1999-08-26 2002-04-09 Robert R. Whittle Alkoxy substituted benzimidazole compounds, pharmaceutical preparations containing the same, and methods of using the same
SE0100823D0 (en) * 2001-03-09 2001-03-09 Astrazeneca Ab Method I to obtain microparticles
CA2386277A1 (en) 1999-10-01 2001-04-12 Natco Pharma Limited An improved pharmaceutical composition and a process for its preparation
CN1197574C (en) 1999-10-20 2005-04-20 卫材株式会社 Method for stabilizing benzimidazole compounds
SE9904044D0 (en) 1999-11-09 1999-11-09 Astra Ab Compounds
US20020044962A1 (en) * 2000-06-06 2002-04-18 Cherukuri S. Rao Encapsulation products for controlled or extended release
US7678387B2 (en) * 2000-06-06 2010-03-16 Capricorn Pharma, Inc. Drug delivery systems
US6572900B1 (en) * 2000-06-09 2003-06-03 Wm. Wrigley, Jr. Company Method for making coated chewing gum products including a high-intensity sweetener
US20020146451A1 (en) 2000-07-15 2002-10-10 Sharma Virender K. Method for the administration of acid-labile drugs
ATE342263T1 (en) 2000-08-04 2006-11-15 Takeda Pharmaceutical SALTS OF BENZIMIDAZOLE DERIVATIVES AND THEIR USE
WO2002030920A1 (en) * 2000-10-12 2002-04-18 Takeda Chemical Industries, Ltd. Benzimidazole compounds, process for producing the same and use thereof
WO2002040054A1 (en) * 2000-11-17 2002-05-23 Takeda Chemical Industries, Ltd. Pharmaceutical preparation containing copolyvidone
US6749867B2 (en) 2000-11-29 2004-06-15 Joseph R. Robinson Delivery system for omeprazole and its salts
CA2436825C (en) * 2000-12-01 2011-01-18 Takeda Chemical Industries, Ltd. Process for the crystallization of (r)- or (s)-lansoprazole
DE10061136C1 (en) 2000-12-07 2002-10-24 Byk Gulden Lomberg Chem Fab Stable, rapidly disintegrating tablets containing proton pump inhibitors, useful for treating elevated gastric secretion, comprising multiple drug units and auxiliaries including basic filler
WO2002051381A1 (en) * 2000-12-26 2002-07-04 Takeda Chemical Industries, Ltd. Porous substance and process for producing the same
US20040097555A1 (en) * 2000-12-26 2004-05-20 Shinegori Ohkawa Concomitant drugs
EP1353624B1 (en) 2000-12-28 2009-03-18 TAP Pharmaceutical Products, Inc. Pharmaceutical compositions of a non-enteric coated proton pump inhibitor with a carbonate salt and bicarbonate salt combination
US20020192299A1 (en) 2000-12-28 2002-12-19 Rajneesh Taneja Pharmaceutical compositions of a non-enteric coated proton pump inhibitor with a carbonate salt and bicarbonate salt combination
WO2002078705A1 (en) * 2001-03-28 2002-10-10 Takeda Chemical Industries, Ltd. Hsp inductor
US6673936B2 (en) * 2001-04-20 2004-01-06 Linda B. Whittall Process for purifying 6-methoxy omeprazole
SI20875A (en) 2001-04-25 2002-10-31 LEK, tovarna farmacevtskih in kemi�nih izdelkov, d.d. Crystal form of omeprazole
PT1405621E (en) 2001-06-20 2011-05-31 Takeda Pharmaceutical Method of manufacturing tablet
EP1404300B1 (en) * 2001-06-22 2009-09-30 Pfizer Products Inc. Pharmaceutical compositions of dispersions of drugs and neutral polymers
AR036354A1 (en) 2001-08-31 2004-09-01 Takeda Chemical Industries Ltd SOLID PREPARATION
US20030050620A1 (en) * 2001-09-07 2003-03-13 Isa Odidi Combinatorial type controlled release drug delivery device
US20030235628A1 (en) 2001-09-19 2003-12-25 Rajneesh Taneja Methods and pharmaceutical formulations for protecting pharmaceutical compounds from acidic environments
BR0206062A (en) 2001-09-28 2004-01-13 Mcneil Ppc Inc Dosage forms having an inner core and outer shell
US20040146559A1 (en) 2002-09-28 2004-07-29 Sowden Harry S. Dosage forms having an inner core and outer shell with different shapes
US20030091630A1 (en) * 2001-10-25 2003-05-15 Jenny Louie-Helm Formulation of an erodible, gastric retentive oral dosage form using in vitro disintegration test data
FR2832311B1 (en) * 2001-11-21 2004-04-16 Besins Int Belgique FILM-FORMING POWDER, COMPOSITIONS COMPRISING SAME, PREPARATION METHODS AND USES THEREOF
US20040082618A1 (en) * 2002-07-03 2004-04-29 Rajneesh Taneja Liquid dosage forms of acid labile drugs
US20040081671A1 (en) * 2002-07-03 2004-04-29 Rajneesh Taneja Liquid dosage forms of non-enterically coated acid-labile drugs
AU2003247792B2 (en) * 2002-07-03 2009-09-24 Nicox S.A. Nitrosated nonsteroidal antiinflammatory compounds, compositions and methods of use
US20040005362A1 (en) * 2002-07-03 2004-01-08 Rajneesh Taneja Liquid dosage forms of acid labile drugs
US20040081700A1 (en) * 2002-07-03 2004-04-29 Rajneesh Taneja Dose titratable liquid dosage forms of acid labile drugs
US20040006109A1 (en) * 2002-07-03 2004-01-08 Rajneesh Taneja Liquid dosage forms of non-enterically coated acid-labile drugs
JP4388331B2 (en) * 2002-10-25 2009-12-24 オリンパス株式会社 Fever treatment device
US20040121004A1 (en) * 2002-12-20 2004-06-24 Rajneesh Taneja Dosage forms containing a PPI, NSAID, and buffer
US20040131676A1 (en) 2002-12-20 2004-07-08 Rajneesh Taneja Dosage forms containing a PPI, NSAID, and buffer
TWI398273B (en) * 2003-07-18 2013-06-11 Santarus Inc Pharmaceutical formulations useful for inhibiting acid secretion and methods for making and using them
US8062664B2 (en) * 2003-11-12 2011-11-22 Abbott Laboratories Process for preparing formulations of lipid-regulating drugs
US8906940B2 (en) 2004-05-25 2014-12-09 Santarus, Inc. Pharmaceutical formulations useful for inhibiting acid secretion and methods for making and using them
US7268165B2 (en) * 2004-08-20 2007-09-11 Steris Inc. Enhanced activity alcohol-based antimicrobial compositions

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040248942A1 (en) * 2003-02-20 2004-12-09 Bonnie Hepburn Novel formulation, omeprazole antacid complex-immediate release for rapid and sustained suppression of gastric acid

Also Published As

Publication number Publication date
US20060204585A1 (en) 2006-09-14
US8993599B2 (en) 2015-03-31

Similar Documents

Publication Publication Date Title
US8993599B2 (en) Pharmaceutical formulations useful for inhibiting acid secretion and methods for making and using them
CA2531564C (en) Pharmaceutical composition for inhibiting acid secretion
US20220133778A1 (en) Pharmaceutical formulations useful for inhibiting acid secretion and methods for making and using them
US20050239845A1 (en) Combination of proton pump inhibitor, buffering agent, and prokinetic agent
AU2011200642B2 (en) Pharmaceutical formulation and method for treating acid-caused gastrointestinal disorders
US8906940B2 (en) Pharmaceutical formulations useful for inhibiting acid secretion and methods for making and using them
EP2068841B1 (en) Novel formulations of proton pump inhibitors and methods of using these formulations
JP2007510733A (en) Combination of proton pump inhibitor and hypnotic
US20070292498A1 (en) Combinations of proton pump inhibitors, sleep aids, buffers and pain relievers
WO2007086846A1 (en) Pharmaceutical formulations useful for inhibiting acid secretion and methods for making and using them
AU2004257779B2 (en) Pharmaceutical composition for inhibiting acid secretion
AU2014233597A1 (en) Pharmaceutical formulation and method for treating acid-caused gastrointestinal disorders

Legal Events

Date Code Title Description
AS Assignment

Owner name: BARCLAYS BANK PLC, NEW YORK

Free format text: SECURITY INTEREST;ASSIGNORS:ATON PHARMA, INC.;BAUSCH & LOMB INCORPORATED;BAUSH & LOMB PHARMA HOLDINGS CORP.;AND OTHERS;REEL/FRAME:039380/0385

Effective date: 20160718

AS Assignment

Owner name: THE BANK OF NEW YORK MELLON, NEW YORK

Free format text: SECURITY INTEREST;ASSIGNOR:SANTARUS, INC.;REEL/FRAME:043041/0913

Effective date: 20170717

AS Assignment

Owner name: THE BANK OF NEW YORK MELLON, AS COLLATERAL AGENT, NEW YORK

Free format text: SECURITY INTEREST;ASSIGNORS:ATON PHARMA, INC.;BAUSCH & LOMB INCORPORATED;BAUSCH & LOMB PHARMA HOLDINGS CORP.;AND OTHERS;REEL/FRAME:045444/0634

Effective date: 20180213

Owner name: BARCLAYS BANK PLC, AS COLLATERAL AGENT, NEW YORK

Free format text: SECURITY INTEREST;ASSIGNORS:ATON PHARMA, INC.;BAUSCH & LOMB INCORPORATED;BAUSCH & LOMB PHARMA HOLDINGS CORP.;AND OTHERS;REEL/FRAME:045444/0299

Effective date: 20180213

Owner name: THE BANK OF NEW YORK MELLON, AS COLLATERAL AGENT,

Free format text: SECURITY INTEREST;ASSIGNORS:ATON PHARMA, INC.;BAUSCH & LOMB INCORPORATED;BAUSCH & LOMB PHARMA HOLDINGS CORP.;AND OTHERS;REEL/FRAME:045444/0634

Effective date: 20180213

STCB Information on status: application discontinuation

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