US20140370093A1 - Oral antimicrobial pharmaceutical compositions - Google Patents

Oral antimicrobial pharmaceutical compositions Download PDF

Info

Publication number
US20140370093A1
US20140370093A1 US14/471,509 US201414471509A US2014370093A1 US 20140370093 A1 US20140370093 A1 US 20140370093A1 US 201414471509 A US201414471509 A US 201414471509A US 2014370093 A1 US2014370093 A1 US 2014370093A1
Authority
US
United States
Prior art keywords
tablet according
tablet
compound
pathological condition
hydrophilic
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
US14/471,509
Inventor
Mauro Ajani
Roberta Bozzella
Giuseppe Celasco
Roberto Villa
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.)
Cosmo Technologies Ltd
Original Assignee
Cosmo Technologies Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Cosmo Technologies Ltd filed Critical Cosmo Technologies Ltd
Priority to US14/471,509 priority Critical patent/US20140370093A1/en
Publication of US20140370093A1 publication Critical patent/US20140370093A1/en
Assigned to COSMO TECHNOLOGIES LTD. reassignment COSMO TECHNOLOGIES LTD. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: AJANI, MAURO, BOZZELLA, ROBERTA, CELASCO, GIUSEPPE, VILLA, ROBERTO
Assigned to COSMO TECHNOLOGIES LTD. reassignment COSMO TECHNOLOGIES LTD. ASSIGNEE ADDRESS CHANGE Assignors: COSMO TECHNOLOGIES LTD.
Priority to US16/289,748 priority patent/US20190192441A1/en
Priority to US17/114,057 priority patent/US20210093576A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • 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
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2013Organic compounds, e.g. phospholipids, fats
    • 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
    • A61K9/2013Organic compounds, e.g. phospholipids, fats
    • A61K9/2018Sugars, or sugar alcohols, e.g. lactose, mannitol; Derivatives thereof, e.g. polysorbates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/205Polysaccharides, e.g. alginate, gums; Cyclodextrin
    • A61K9/2054Cellulose; Cellulose derivatives, e.g. hydroxypropyl methylcellulose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • 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
    • A61K9/282Organic compounds, e.g. fats
    • 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
    • A61K9/282Organic compounds, e.g. fats
    • A61K9/2826Sugars or sugar alcohols, e.g. sucrose; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/12Antidiarrhoeals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/12Drugs for disorders of the metabolism for electrolyte homeostasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P39/00General protective or antinoxious agents
    • A61P39/02Antidotes
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • Intestinal infections are common diseases caused by the colonization of the intestine by foreign pathogenic agents of various origins, or caused by intestinal microorganisms that are normally present becoming virulent.
  • the intestine is divided into two distinct portions: the proximal portion, called the “small intestine”, which is formed, in the craniocaudal direction, by the duodenum, the jejunum and the ileum, and the distal portion, called the “large intestine”, which is formed by the colon and the recto-anus (Faller A, Scevola G. Anatomia e Fisiologia del Corpo Umano ( Anatomy and Physiology of the Human Body ). Vol I. Edizioni Minerva Medica, Turin, 1973, pp. 235-254).
  • the two portions, the small intestine and the large intestine, are completely separated anatomically by the ileocaecal valve which permits the passage of the intestinal contents from the small intestine to the large intestine but not vice versa.
  • the large intestine is quite different from the small intestine also, and above all, from the functional point of view (Braga P C. Enteric microflora and its regulation. In Drugs in Gastroenterology. Raven Press, New York, 1991, pp. 501-508).
  • the large intestine provides for the absorption of water, for the digestion of vegetable fibres and for the completion of some digestive processes initiated in the small intestine.
  • the large intestine differs from the small intestine by the presence of an extremely rich bacterial flora, the balance of which is of fundamental importance in regulating the ambient pH, motility, the production of gas and ammonia, the formation of faeces, and the production of metabolites essential for maintaining the good functioning of the large intestine.
  • the colon is the portion of the large intestine that is host to the majority of the bacterial strains and that offers conditions of pH, anaerobiosis, humidity and slowness of transit that are particularly suitable for the permanent flora potentially becoming virulent or for the proliferation of and colonization by pathogenic bacteria.
  • the colon is the sector of the intestine most susceptible to infection; in fact, infections located in the colon (infectious colites, bacillary dysentery, diarrhoea, pseudomembranous colitis, diverticulitis, etc.) constitute an important and autonomous chapter in the gastroenterological monograph (Sorice F., Vullo V. Intossicazioni alimentari e infezioni del tubo digerente. ( Food Poisoning and Infections of the Alimentary Canal ). In: Medicina Clinica (Clinical Medicine).Edizioni Medico Scientifiche, Turin, 2002).
  • the oral therapy of intestinal infections, and in particular colon infections uses substances having antibacterial activity which must have specific characteristics such as: broad spectrum of activity on Gram+ and Gram ⁇ bacteria, resistance to strongly acidic environments, such as the gastric environment, anti-infectious activity independent of the presence of the intestinal biomass, residence inside the intestine for an appropriate period of time, good penetrability into the infecting host cell and good tolerability (Braga P C. Interaction of antibiotics on enteric microflora. In: Drugs in Gastroenterology. Raven Press, New York, 1991, pp. 509-517).
  • the antibacterial agents if not suitably protected, may lose their efficacy owing to the enzymatic or degradative inactivation which occurs during their passage through the stomach or through the small intestine.
  • the pharmaceutical forms nowadays used although they permit the administration of the active ingredient in discrete doses, release it too rapidly in relation to the time taken to pass through the digestive tract, so that the active ingredient performs its anti-infectious activity in an indiscriminate manner along the entire gastro-intestinal tract.
  • this bacterial flora is important in fundamental biological processes, such as, for example, the digestion and absorption of alimentary nutritive components, the production and absorption of vitamins (vitamin K and B-complex vitamins), the metabolism of biliary acids and of steroid hormones, the activation and inactivation of various substances, the protection of the organism from xenobiotics, (Braga PC. Ibidem).
  • the antimicrobial agents used for the disinfection of the digestive tract often do not have a high rate of metabolism, in order to maintain unaltered the therapeutic possibility connected with the administration of a traditional form containing antimicrobial agents, no phenomenon of metabolic degradation should occur, in order to avoid any weakening of the therapeutic efficacy associated with the presence of the antimicrobial agent.
  • Rifamycin SV which has been known since the 1960s, is a semi-synthetic active ingredient which is derived from rifamycin S and which has a strong antimicrobial and/or anti-infectious activity both locally and parenterally. Its activity has also been evaluated in vitro at minimum concentrations (mcg/ml) on Gram+ bacteria, such as Staphylococcus aureus or Enterococcus faecalis , as well as at higher concentrations on Gram ⁇ bacteria, such as Escherichia coli, Salmonella, Enterobacter aerogenes, Enterobacter cloacae or Pseudomonas aeruginosa.
  • Gram+ bacteria such as Staphylococcus aureus or Enterococcus faecalis
  • Gram ⁇ bacteria such as Escherichia coli, Salmonella, Enterobacter aerogenes, Enterobacter cloacae or Pseudomonas aeruginosa.
  • Rifamycin SV in the form of its sodium salt, is currently marketed under the name Rifocin® both for external topical use and for injection.
  • the topical use indicated for the local treatment of infectious processes, is limited to external use by means of a solution of the active ingredient which is to be diluted at the time of use.
  • Patent application WO01/11077 which is incorporated herein by reference, describes the use of antimicrobial agents, including the generic rifamycin, for the preparation of pharmaceutical compositions that can be used in the treatment of pathologies caused by anomalous bacterial growth (Small Intestine Overgrowth—SIBO) at the expense of the small intestine.
  • Those compositions are formulated in such a manner as to release the active ingredient rapidly in the proximal portion of the intestine, that is to say, solely in the small intestine (duodenum, jejunum and ileum).
  • Metronidazole is a nitroimidazole chemotherapeutic agent having powerful antimicrobial activity and a broad spectrum of action both on Gram+ bacteria and Gram ⁇ bacteria.
  • metronidazole is known to have a proven antiprotozoan activity ( Tracy J. W. et al., Metronidazole, in: Goodmen & Gilman's, The Pharmacological Bases of Therapeutics, IX Ed., 1996, pp 995-998).
  • Current therapy with metronidazole is supported with tablets (Flagyl®) that contain 250 mg of active ingredient and that are formulated for immediate release.
  • rifamycin SV was verified by means of an evaluation of the MIC (Minimum Inhibiting Concentration) on specific pathogenic bacterial strains, such as, for example, Escherichia coli, Enterobacter faecalis, Proteus vulgaris, Pseudomonas aeruginosa, Salmonella typhi and Enterobacter cloacae as shown in the following Table A.
  • MIC Minimum Inhibiting Concentration
  • the present invention therefore relates to oral pharmaceutical compositions containing an active ingredient having antimicrobial/anti-infectious activity, such as rifamycin SV and/or metronidazole, characterized in that they are formulated in such a manner as to release the active substances substantially in the portion of the large intestine where their specific sterilizing action is required, but leaving unaltered the non-pathogenic bacterial flora present in the portions of the small intestine which are not affected by the infection.
  • an active ingredient having antimicrobial/anti-infectious activity such as rifamycin SV and/or metronidazole
  • the formulations according to the present invention are capable of releasing the active ingredient solely in the colon, thus ensuring localized and restricted anti-infectious efficacy.
  • the advantage of the formulations of the invention is the particular site-specificity in the large intestine, and in particular in the colon, which permits a greater concentration of the active substance in the infected distal intestinal region with complete preservation of the healthy proximal regions.
  • a further advantageous application of the formulations of the invention is their use during preparation for surgical operations on the large intestine, in ileocolic anastomoses, and in the sterilization of the ammonia-producing colonic flora in order to prevent and/or treat hyperammonaemias.
  • the site-specificity of treatment and the consequent concentration of the activity of the active ingredient may lead to a significant resolution of cases which would otherwise involve substantial complications.
  • the substances having antimicrobial/anti-infectious activity are contained in an amount of from 10 to 90% by weight; in particular rifamycin SV is contained in an amount of from 20% to 60% by weight, while metronidazole is contained in an amount of from 25% to 70% by weight.
  • the oral formulations of the invention are selected from tablets, capsules, granules and/or microgranules.
  • a preferred embodiment of the present invention comprises a system for controlled release which is characterized by the presence of a first, amphiphilic matrix in which the active ingredient is incorporated and which is in turn dispersed in a second lipophilic, matrix. The form so obtained is again in turn dispersed in a third, hydrophilic matrix before producing the final oral pharmaceutical form.
  • the lipophilic matrix of the present invention is represented by substances having a melting point lower than 90° C., such as, for example, beeswax, carnauba wax, stearic acid, stearin and the like;
  • the amphiphilic matrix is represented by substances selected, for example, from phospholipids, ceramides, sphingomyelins, lecithins, alkyl block copolymers, salts of sulphated alkyl acids, polyoxyethylenated alkyl, derivatives of sorbitan and the like
  • the hydrophilic matrix is represented by generally cross-linked or linear polymeric or copolymeric substances, which are known as hydrogels, that is to say, substances capable of increasing their mass and their weight, owing to the polar groups present in the main or side polymer chains, when they come into contact with molecules of water.
  • the hydrophilic matrix corresponds to substances selected, for example, from cellulose derivatives, such as hydroxyalkylcelluloses, alkylcelluloses, carboxyalkylcelluloses and their salts or derivatives, polyvinyl alcohols, carboxyvinyl derivatives, polysaccharide derivatives of anionic or cationic nature, such as, for example, hyduronic acid, glucuronic acid, or glucosamines, pectins and/or their derivatives.
  • cellulose derivatives such as hydroxyalkylcelluloses, alkylcelluloses, carboxyalkylcelluloses and their salts or derivatives
  • polyvinyl alcohols such as, for example, hyduronic acid, glucuronic acid, or glucosamines, pectins and/or their derivatives.
  • the matrices are dispersed in one another in succession together with the active ingredient, thus bringing about the formation of a homogeneous structure responsible for the site-specificity of release.
  • the tablets obtained are finally subjected to a coating process using gastroresistant substances, such as, for example, polymers of acrylic and methacrylic acids (Eudragit) and/or derivatives of cellulose phthalate.
  • gastroresistant substances such as, for example, polymers of acrylic and methacrylic acids (Eudragit) and/or derivatives of cellulose phthalate.
  • 200 g of rifamycin SV are mixed with 5 g of stearic acid, 7 g of carnauba wax, 8 g of sodium dioctyl sulphosuccinate, 100 g of lactose and 10 g of sodium edetate and granulated with a solution containing 25 g of low-viscosity polyvinylpyrrolidone in 0.2 litre of purified water.
  • the granulate When the granulate has been dried, it is mixed with 100 g of sodium carboxymethylcellulose, 25 g of silica, 5 g of glycerol palmitostearate and 10 mg of talcum before being subjected to compression to the unit weight of 495 mg/tablet.
  • the cores so obtained are then film-coated with a hydroalcoholic dispersion of acrylic and methacrylic acid esters, titanium dioxide, talcum and triethyl citrate, which confers on the product resistance to disintegration in a strongly acidic environment, simulating the environment of the stomach and the small intestine.
  • the dissolution of the tablets is practically zero in pH conditions of less than 7 and is progressive in an enteric buffer at pH 7.2 with the following percentage quotas:
  • 500 g of rifamycin SV are mixed with 10 g of stearic acid, 10 g of beeswax, 10 g of sodium lauryl sulphate, 200 g of mannitol and 10 g of sodium edetate and granulated with a solution containing 50 g of hydroxypropylcellulose in 0.5 litre of water.
  • the granulate When the granulate has been dried, it is mixed with 150 g of sodium hydroxypropylmethylcellulose, 25 g of silica, 5 g of glycerol palmitostearate and 10 mg of talcum before being subjected to compression to the unit weight of 490 mg/tablet.
  • the cores so obtained are then film-coated with an aqueous dispersion of acrylic and methacrylic acid esters, iron oxide, talcum and triethyl citrate, with confers on the product resistance to disintegration in an acidic environment, simulating the environment of the stomach and the small intestine.
  • the dissolution of the tablets is practically zero in pH conditions of less than 7 and is progressive in an enteric buffer at pH 7.2 with the following percentage quotas:
  • metronidazole 2.5 kg are mixed with 70 g of stearic acid, 70 g of beeswax, 400 g of saccharose, 140 g of hydroxypropylmethylcellulose and 20 g of polysorbate and wet-granulated by the addition of purified water to a suitable consistency.
  • the granulate is then dried and standardized in terms of dimensions before the addition of a further 200 g of hydroxymethylpropylcellulose, 600 g of microcrystalline cellulose, 30 g of glycerol palmitostearate and 70 g of silicon dioxide. After mixing, the powder is sent for compression to the unit weight of 450 mg/tablet.
  • the cores so obtained are then subjected to film-coating with a hydroalcoholic dispersion of acrylic and methacrylic acid esters, iron oxide, talcum and triethyl citrate, which confers on the product resistance to disintegration in an acidic environment.
  • the dissolution of the tablets is practically zero in pH conditions of less than 7 and is progressive in an enteric buffer at pH 7.2 with the following percentage quotas:
  • 500 g of metronidazole are mixed with the components of the lipophilic/amphiphilic matrix, 5 g of stearic acid and 5 g of soya lecithin, some of the hydrophilic polymer, 100 g of hydroxypropylcellulose, and diluents, 150 g of mannitol.
  • the mixture is then made into a paste with a solution of low-viscosity hydroxypropylcellulose in purified water until a consistent granulate is obtained.
  • the granulate obtained is mixed with a further 100 g of hydroxypropylcellulose, to which are added flow agents and lubricants, 5 g of silica, 5 g of talcum and 5 g of magnesium stearate, then compressed to a final weight of 925 mg/tablet.
  • the tablets are finally coated with an alcohol-based suspension of acrylic and methacrylic copolymers capable of imparting to the tablets efficacious gastroresistance.
  • the rate of dissolution of those tablets is progressive and controlled, with approximately 20% of the active ingredient being released after the first hour of residence in enteric juice at pH 7.2, 50% after 2 hours and more than 80% after 4 hours, these figures being understood as quotas that are clearly subsequent to 2 hours' exposure at pH 1 and 1 hour's exposure at pH 6.4, reflecting the environment of the stomach and of the small intestine, respectively.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical & Material Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Epidemiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Biophysics (AREA)
  • Molecular Biology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Diabetes (AREA)
  • Toxicology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicinal Preparation (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)

Abstract

The present invention relates to oral pharmaceutical compositions with controlled and/or programmed release containing at least one active ingredient having antimicrobial and/or anti-infectious activity for the treatment of infections of the large intestine, in particular the colon.

Description

    CROSS REFERENCE TO RELATED APPLICATIONS
  • This application is a continuation of U.S. patent application Ser. No. 13/592,097, filed on Aug. 22, 2012, which is a continuation of U.S. patent application Ser. No. 13/451,111, filed Apr. 19, 2012, now U.S. Pat. No. 8,263,120, which is a continuation of U.S. patent application Ser. No. 11/571,044, filed Feb. 5, 2007, now U.S. Pat. No. 8,486,446, which is a U.S. National Phase under 35 U.S.C. §371 of International Application No. PCT/EP2005/052025, filed May 3, 2005, which claims the benefit of Italian Patent Application MI2004 A 001295 filed on Jun. 25, 2004, all of which are incorporated by reference herein in their entirety.
  • DESCRIPTION
  • Intestinal infections are common diseases caused by the colonization of the intestine by foreign pathogenic agents of various origins, or caused by intestinal microorganisms that are normally present becoming virulent.
  • It is known that the intestine is divided into two distinct portions: the proximal portion, called the “small intestine”, which is formed, in the craniocaudal direction, by the duodenum, the jejunum and the ileum, and the distal portion, called the “large intestine”, which is formed by the colon and the recto-anus (Faller A, Scevola G. Anatomia e Fisiologia del Corpo Umano (Anatomy and Physiology of the Human Body). Vol I. Edizioni Minerva Medica, Turin, 1973, pp. 235-254).
  • The two portions, the small intestine and the large intestine, are completely separated anatomically by the ileocaecal valve which permits the passage of the intestinal contents from the small intestine to the large intestine but not vice versa. Besides from the anatomical-structural point of view, the large intestine is quite different from the small intestine also, and above all, from the functional point of view (Braga P C. Enteric microflora and its regulation. In Drugs in Gastroenterology. Raven Press, New York, 1991, pp. 501-508).
  • While the small intestine is assigned to the digestion of the majority of the food, to the absorption thereof, to the production of B-complex vitamins and vitamin K, to the metabolism of biliary acids and various other organic substances and to the rapid transfer of the alimentary bolus to the sections further downstream, the large intestine provides for the absorption of water, for the digestion of vegetable fibres and for the completion of some digestive processes initiated in the small intestine. In addition, the large intestine differs from the small intestine by the presence of an extremely rich bacterial flora, the balance of which is of fundamental importance in regulating the ambient pH, motility, the production of gas and ammonia, the formation of faeces, and the production of metabolites essential for maintaining the good functioning of the large intestine.
  • These many differences between the small intestine and the large intestine explain the distinctive nature of some pathologies which occur at the expense of the large intestine and in particular the colon.
  • The colon is the portion of the large intestine that is host to the majority of the bacterial strains and that offers conditions of pH, anaerobiosis, humidity and slowness of transit that are particularly suitable for the permanent flora potentially becoming virulent or for the proliferation of and colonization by pathogenic bacteria. For those reasons, the colon is the sector of the intestine most susceptible to infection; in fact, infections located in the colon (infectious colites, bacillary dysentery, diarrhoea, pseudomembranous colitis, diverticulitis, etc.) constitute an important and autonomous chapter in the gastroenterological monograph (Sorice F., Vullo V. Intossicazioni alimentari e infezioni del tubo digerente. (Food Poisoning and Infections of the Alimentary Canal). In: Medicina Clinica (Clinical Medicine).Edizioni Medico Scientifiche, Turin, 2002).
  • In addition, the increased endoluminal pressure, linked with the production of gas and associated with predisposing local factors, can promote the occurrence of diverticula which are susceptible to infection and inflammation and which are located exclusively in the colon (Jackson B T. Diverticular disease. In: Inflammatory Bowel Diseases Churchill Livingston, N.Y., 1997, pp. 443-447).
  • Currently, the oral therapy of intestinal infections, and in particular colon infections, uses substances having antibacterial activity which must have specific characteristics such as: broad spectrum of activity on Gram+ and Gram− bacteria, resistance to strongly acidic environments, such as the gastric environment, anti-infectious activity independent of the presence of the intestinal biomass, residence inside the intestine for an appropriate period of time, good penetrability into the infecting host cell and good tolerability (Braga P C. Interaction of antibiotics on enteric microflora. In: Drugs in Gastroenterology. Raven Press, New York, 1991, pp. 509-517).
  • Therapy with antibacterial agents administered in the oral preparations employed today has at least two limitations. In the first place, the antibacterial agents, if not suitably protected, may lose their efficacy owing to the enzymatic or degradative inactivation which occurs during their passage through the stomach or through the small intestine.
  • In addition, the pharmaceutical forms nowadays used, although they permit the administration of the active ingredient in discrete doses, release it too rapidly in relation to the time taken to pass through the digestive tract, so that the active ingredient performs its anti-infectious activity in an indiscriminate manner along the entire gastro-intestinal tract.
  • This leads to the disappearance of the non-pathogenic bacterial flora living in the small intestine (duodenum, jejunum and the ileum), which flora, since it is not normally the seat of infection, should be protected and not subjected to the sterilizing action characteristic of the formulations used today.
  • For it is known that this bacterial flora is important in fundamental biological processes, such as, for example, the digestion and absorption of alimentary nutritive components, the production and absorption of vitamins (vitamin K and B-complex vitamins), the metabolism of biliary acids and of steroid hormones, the activation and inactivation of various substances, the protection of the organism from xenobiotics, (Braga PC. Ibidem).
  • In particular, the usual oral antibacterial therapies for the treatment of pathologies located in the colon have often given a contradictory result, probably owing to the excessive dilution of the active ingredients in the intestinal lumen; this dilution is caused by the premature release of the antimicrobial agent from the pharmaceutical form containing it, which takes place as early as in the stomach and in the immediate vicinity of the patient's pyloric valve.
  • In addition, although the antimicrobial agents used for the disinfection of the digestive tract often do not have a high rate of metabolism, in order to maintain unaltered the therapeutic possibility connected with the administration of a traditional form containing antimicrobial agents, no phenomenon of metabolic degradation should occur, in order to avoid any weakening of the therapeutic efficacy associated with the presence of the antimicrobial agent.
  • Therefore, in such cases, in order to ensure the real efficacy of the anti-infectious therapy, it is felt that there is a need for the possibility of a controlled and site-specific form of administration.
  • For the release of the antimicrobial/anti-infectious active ingredient in the immediate vicinity of the region where a diverticulum or a generic infection becomes established, leads to the formation of a much higher concentration gradient than in the case of a conventional form of oral administration, with the consequent greater possibility that the antimicrobial agent will succeed in penetrating to the inside of the diverticulum.
  • In that situation, particular importance is attached to the possibility of the remission also of infectious pathologies which are not widespread but which are of considerable socio-epidemiological importance, such as bacillary dysentery and pseudomembranous colitis, and also of infectious complications in surgical operations at the expense of the large intestine and in particular the colon.
  • Rifamycin SV, which has been known since the 1960s, is a semi-synthetic active ingredient which is derived from rifamycin S and which has a strong antimicrobial and/or anti-infectious activity both locally and parenterally. Its activity has also been evaluated in vitro at minimum concentrations (mcg/ml) on Gram+ bacteria, such as Staphylococcus aureus or Enterococcus faecalis, as well as at higher concentrations on Gram− bacteria, such as Escherichia coli, Salmonella, Enterobacter aerogenes, Enterobacter cloacae or Pseudomonas aeruginosa.
  • Rifamycin SV, in the form of its sodium salt, is currently marketed under the name Rifocin® both for external topical use and for injection. In particular, the topical use, indicated for the local treatment of infectious processes, is limited to external use by means of a solution of the active ingredient which is to be diluted at the time of use.
  • Patent application WO01/11077, which is incorporated herein by reference, describes the use of antimicrobial agents, including the generic rifamycin, for the preparation of pharmaceutical compositions that can be used in the treatment of pathologies caused by anomalous bacterial growth (Small Intestine Overgrowth—SIBO) at the expense of the small intestine. Those compositions are formulated in such a manner as to release the active ingredient rapidly in the proximal portion of the intestine, that is to say, solely in the small intestine (duodenum, jejunum and ileum).
  • Metronidazole is a nitroimidazole chemotherapeutic agent having powerful antimicrobial activity and a broad spectrum of action both on Gram+ bacteria and Gram− bacteria. In addition, metronidazole is known to have a proven antiprotozoan activity (Tracy J. W. et al., Metronidazole, in: Goodmen & Gilman's, The Pharmacological Bases of Therapeutics, IX Ed., 1996, pp 995-998). Current therapy with metronidazole is supported with tablets (Flagyl®) that contain 250 mg of active ingredient and that are formulated for immediate release.
  • It has now surprisingly been found that the efficacy of antimicrobial/anti-infectious active ingredients, such as rifamycin SV and/or metronidazole, in the treatment of infections of the large intestine, and in particular of the colon, can be substantially potentiated thanks to the elimination of the undesired effects described above (avitaminosis, destruction of non-pathogenic bacterial flora, etc.) which are caused by the premature release of the active ingredients in the first portions of the digestive canal, such as the stomach, the duodenum and the jejunum, and thanks to the protection from the metabolic-enzymatic inactivation of the active ingredients which is brought about before the ingredients can reach the site of infection.
  • In particular, the efficacy of rifamycin SV was verified by means of an evaluation of the MIC (Minimum Inhibiting Concentration) on specific pathogenic bacterial strains, such as, for example, Escherichia coli, Enterobacter faecalis, Proteus vulgaris, Pseudomonas aeruginosa, Salmonella typhi and Enterobacter cloacae as shown in the following Table A.
  • TABLE A
    Bacterial species MIC (mcg/ml)
    Escherichia coli (ATCC 30218) 400
    Enterobacter faecalis (ATCC 29212) 25
    Proteus vulgaris (ATCC 13315) 400
    Pseudomonas aeruginosa (ATCC 27853) >400
    Salmonella typhi (ATCC 13331) >400
    Enterobacter cloacae (ATCC 17446431) >400
    Staphylococcus aureus (ATCC 25213) <0.4
  • The present invention therefore relates to oral pharmaceutical compositions containing an active ingredient having antimicrobial/anti-infectious activity, such as rifamycin SV and/or metronidazole, characterized in that they are formulated in such a manner as to release the active substances substantially in the portion of the large intestine where their specific sterilizing action is required, but leaving unaltered the non-pathogenic bacterial flora present in the portions of the small intestine which are not affected by the infection.
  • In particular, the formulations according to the present invention are capable of releasing the active ingredient solely in the colon, thus ensuring localized and restricted anti-infectious efficacy.
  • Consequently, the advantage of the formulations of the invention is the particular site-specificity in the large intestine, and in particular in the colon, which permits a greater concentration of the active substance in the infected distal intestinal region with complete preservation of the healthy proximal regions.
  • This advantage is displayed mainly during the treatment of specific pathological situations in the colon region, such as infectious colites, bacillary dysentery, diverticular disease and diverticulitis where the site-specificity and the tolerability of the formulations play a key role in the resolution of the pathology.
  • A further advantageous application of the formulations of the invention is their use during preparation for surgical operations on the large intestine, in ileocolic anastomoses, and in the sterilization of the ammonia-producing colonic flora in order to prevent and/or treat hyperammonaemias. In these last-mentioned cases, the site-specificity of treatment and the consequent concentration of the activity of the active ingredient may lead to a significant resolution of cases which would otherwise involve substantial complications.
  • In the formulations of the invention, the substances having antimicrobial/anti-infectious activity are contained in an amount of from 10 to 90% by weight; in particular rifamycin SV is contained in an amount of from 20% to 60% by weight, while metronidazole is contained in an amount of from 25% to 70% by weight. The oral formulations of the invention are selected from tablets, capsules, granules and/or microgranules.
  • A preferred embodiment of the present invention comprises a system for controlled release which is characterized by the presence of a first, amphiphilic matrix in which the active ingredient is incorporated and which is in turn dispersed in a second lipophilic, matrix. The form so obtained is again in turn dispersed in a third, hydrophilic matrix before producing the final oral pharmaceutical form.
  • The lipophilic matrix of the present invention is represented by substances having a melting point lower than 90° C., such as, for example, beeswax, carnauba wax, stearic acid, stearin and the like; the amphiphilic matrix is represented by substances selected, for example, from phospholipids, ceramides, sphingomyelins, lecithins, alkyl block copolymers, salts of sulphated alkyl acids, polyoxyethylenated alkyl, derivatives of sorbitan and the like, while the hydrophilic matrix is represented by generally cross-linked or linear polymeric or copolymeric substances, which are known as hydrogels, that is to say, substances capable of increasing their mass and their weight, owing to the polar groups present in the main or side polymer chains, when they come into contact with molecules of water.
  • In particular, the hydrophilic matrix corresponds to substances selected, for example, from cellulose derivatives, such as hydroxyalkylcelluloses, alkylcelluloses, carboxyalkylcelluloses and their salts or derivatives, polyvinyl alcohols, carboxyvinyl derivatives, polysaccharide derivatives of anionic or cationic nature, such as, for example, hyduronic acid, glucuronic acid, or glucosamines, pectins and/or their derivatives.
  • In this preferred embodiment, the matrices are dispersed in one another in succession together with the active ingredient, thus bringing about the formation of a homogeneous structure responsible for the site-specificity of release.
  • In a further embodiment of the present invention, the tablets obtained are finally subjected to a coating process using gastroresistant substances, such as, for example, polymers of acrylic and methacrylic acids (Eudragit) and/or derivatives of cellulose phthalate.
  • Systems of controlled and/or programmed release suitable for the present invention are described in EP 1183014, GB 2245492 and EP572942, which are also incorporated herein by reference.
  • The following Examples describe the invention in detail without limiting the content thereof in any way.
  • EXAMPLE 1
  • 200 g of rifamycin SV are mixed with 5 g of stearic acid, 7 g of carnauba wax, 8 g of sodium dioctyl sulphosuccinate, 100 g of lactose and 10 g of sodium edetate and granulated with a solution containing 25 g of low-viscosity polyvinylpyrrolidone in 0.2 litre of purified water. When the granulate has been dried, it is mixed with 100 g of sodium carboxymethylcellulose, 25 g of silica, 5 g of glycerol palmitostearate and 10 mg of talcum before being subjected to compression to the unit weight of 495 mg/tablet. The cores so obtained are then film-coated with a hydroalcoholic dispersion of acrylic and methacrylic acid esters, titanium dioxide, talcum and triethyl citrate, which confers on the product resistance to disintegration in a strongly acidic environment, simulating the environment of the stomach and the small intestine. The dissolution of the tablets is practically zero in pH conditions of less than 7 and is progressive in an enteric buffer at pH 7.2 with the following percentage quotas:
      • less than 20% after 1 hour's residence,
      • less than 50% after 3 hours' residence,
      • more than 70% after 8 hours' residence.
    EXAMPLE 2
  • 500 g of rifamycin SV are mixed with 10 g of stearic acid, 10 g of beeswax, 10 g of sodium lauryl sulphate, 200 g of mannitol and 10 g of sodium edetate and granulated with a solution containing 50 g of hydroxypropylcellulose in 0.5 litre of water. When the granulate has been dried, it is mixed with 150 g of sodium hydroxypropylmethylcellulose, 25 g of silica, 5 g of glycerol palmitostearate and 10 mg of talcum before being subjected to compression to the unit weight of 490 mg/tablet. The cores so obtained are then film-coated with an aqueous dispersion of acrylic and methacrylic acid esters, iron oxide, talcum and triethyl citrate, with confers on the product resistance to disintegration in an acidic environment, simulating the environment of the stomach and the small intestine. The dissolution of the tablets is practically zero in pH conditions of less than 7 and is progressive in an enteric buffer at pH 7.2 with the following percentage quotas:
      • less than 30% after 1 hour's residence,
      • less than 60% after 3 hours' residence,
      • more than 80% after 8 hours' residence.
    EXAMPLE 3
  • 2.5 kg of metronidazole are mixed with 70 g of stearic acid, 70 g of beeswax, 400 g of saccharose, 140 g of hydroxypropylmethylcellulose and 20 g of polysorbate and wet-granulated by the addition of purified water to a suitable consistency. The granulate is then dried and standardized in terms of dimensions before the addition of a further 200 g of hydroxymethylpropylcellulose, 600 g of microcrystalline cellulose, 30 g of glycerol palmitostearate and 70 g of silicon dioxide. After mixing, the powder is sent for compression to the unit weight of 450 mg/tablet.
  • The cores so obtained are then subjected to film-coating with a hydroalcoholic dispersion of acrylic and methacrylic acid esters, iron oxide, talcum and triethyl citrate, which confers on the product resistance to disintegration in an acidic environment. The dissolution of the tablets is practically zero in pH conditions of less than 7 and is progressive in an enteric buffer at pH 7.2 with the following percentage quotas:
      • less than 25% within the first hour of residence,
      • more than 25% and less than 70% within the third hour of residence,
      • more than 80% after 8 hours' residence.
    EXAMPLE 4
  • 500 g of metronidazole are mixed with the components of the lipophilic/amphiphilic matrix, 5 g of stearic acid and 5 g of soya lecithin, some of the hydrophilic polymer, 100 g of hydroxypropylcellulose, and diluents, 150 g of mannitol.
  • The mixture is then made into a paste with a solution of low-viscosity hydroxypropylcellulose in purified water until a consistent granulate is obtained. After drying, the granulate obtained is mixed with a further 100 g of hydroxypropylcellulose, to which are added flow agents and lubricants, 5 g of silica, 5 g of talcum and 5 g of magnesium stearate, then compressed to a final weight of 925 mg/tablet. The tablets are finally coated with an alcohol-based suspension of acrylic and methacrylic copolymers capable of imparting to the tablets efficacious gastroresistance.
  • The rate of dissolution of those tablets is progressive and controlled, with approximately 20% of the active ingredient being released after the first hour of residence in enteric juice at pH 7.2, 50% after 2 hours and more than 80% after 4 hours, these figures being understood as quotas that are clearly subsequent to 2 hours' exposure at pH 1 and 1 hour's exposure at pH 6.4, reflecting the environment of the stomach and of the small intestine, respectively.

Claims (49)

1. A tablet comprising granules and at least one hydrophilic compound, wherein the granules comprise:
(a) rifamycin SV;
(b) at least one amphiphilic compound; and
(c) at least one lipophilic compound.
2. The tablet according to claim 1, wherein the at least one hydrophilic compound is selected from the group consisting of a hydroxyalkyl cellulose, an alkyl cellulose, a carboxyalkyl cellulose, a polyvinyl alcohol, a carboxyvinyl polymer, a carboxyvinyl copolymer, a polysaccharide derivative, a hyaluronic acid, a glucuronic acid, a glucosamine and a pectin.
3. The tablet according to claim 1, wherein the at least one hydrophilic compound is a hydroxyalkyl cellulose or a carboxyalkyl cellulose.
4. The tablet according to claim 1, wherein the at least one hydrophilic compound is a hydrophilic copolymeric substance.
5. The tablet according to claim 1, wherein the at least one hydrophilic compound is a hydrogel.
6. The tablet according to claim 1, wherein the at least one amphiphilic compound is selected from the group consisting of lecithin, a sphingomyelin, a phospholipid, a ceramide, an alkyl block copolymer, a salt of a sulphated alkyl acid, a polyoxyethylenated alkyl and a sorbitan derivative.
7. The tablet according to claim 1, wherein the at least one amphiphilic compound is lecithin.
8. The tablet according to claim 1, wherein the at least one lipophilic compound is selected from the group consisting a wax, stearic acid and stearin.
9. The tablet according to claim 1, wherein the at least one lipophilic compound is stearic acid.
10. The tablet according to claim 1, further comprising a gastro-resistant film coating.
11. The tablet according to claim 10, wherein the gastro-resistant film coating comprises acrylic and/or methacrylic acid polymers, or copolymers, or cellulose phthalate derivatives.
12. The tablet according to claim 10, wherein the gastro-resistant film coating comprises acrylic and methacrylic acid copolymers.
13. A tablet coated with a gastro-resistant film, wherein the gastro-resistant film comprises methacrylic acid polymers and/or copolymers, and the tablet comprises:
(i) a hydrophilic copolymeric substance; and
(ii) granules in the hydrophilic copolymeric substance, wherein the granules comprise:
(a) rifamycin SV;
(b) lecithin; and
(c) stearic acid.
14. A tablet comprising rifamycin SV, at least one amphiphilic compound, at least one lipophilic compound and at least one hydrophilic compound prepared by a process comprising the steps:
(a) mixing rifamycin SV, at least one amphiphilic compound and at least one lipophilic compound to form a first mixture;
(b) granulating the first mixture to form granules;
(c) mixing the granules with at least one hydrophilic compound to form a second mixture; and
(d) tableting the second mixture to form a tablet.
15. The tablet according to claim 14, wherein the at least one hydrophilic compound is selected from the group consisting of a hydroxyalkyl cellulose, an alkyl cellulose, a carboxyalkyl cellulose, a polyvinyl alcohol, a carboxyvinyl polymer, a carboxyvinyl copolymer, a polysaccharide derivative, a hyaluronic acid, a glucuronic acid, a glucosamine and a pectin.
16. The tablet according to claim 14, wherein the at least one hydrophilic compound is a hydroxyalkyl cellulose or a carboxyalkyl cellulose.
17. The tablet according to claim 14 wherein the at least one hydrophilic compound is a hydrophilic copolymeric substance.
18. The tablet according to claim 14, wherein the at least one hydrophilic compound is a hydrogel.
19. The tablet according to claim 14, wherein the at least one amphiphilic compound is selected from the group consisting of lecithin, a sphingomyelin, a phospholipid, a ceramide, an alkyl block copolymer, a salt of a sulphated alkyl acid, a polyoxyethylenated alkyl and a sorbitan derivative.
20. The tablet according to claim 14, wherein the at least one amphiphilic compound is lecithin.
21. The tablet according to claim 14, wherein the at least one lipophilic compound is selected from the group consisting a wax, stearic acid and stearin.
22. The tablet according to claim 14, wherein the at least one lipophilic compound is stearic acid.
23. The tablet according to claim 14, wherein the tablet further comprises a gastro-resistant film coating prepared by (e) coating the tablet of step (d).
24. The tablet according to claim 23, wherein the gastro-resistant film coating comprises acrylic and/or methacrylic acid polymers, or copolymers, or cellulose phthalate derivatives.
25. The tablet according to claim 23, wherein the gastro-resistant film coating comprises acrylic and methacrylic acid copolymers.
26. A tablet comprising rifamycin SV, at least one amphiphilic compound, at least one lipophilic compound and at least one hydrophilic compound prepared by a process comprising the steps:
(a) mixing rifamycin SV and at least one amphiphilic compound to form a first mixture;
(b) mixing the first mixture with the at least one lipophilic compound to form a second mixture;
(c) mixing the second mixture with the at least one hydrophilic compound to form a third mixture; and
(d) tableting the third mixture to form a tablet.
27. The tablet according to claim 26, wherein the at least one hydrophilic compound is selected from the group consisting of a hydroxyalkyl cellulose, an alkyl cellulose, a carboxyalkyl cellulose, a polyvinyl alcohol, a carboxyvinyl polymer, a carboxyvinyl copolymer, a polysaccharide derivative, a hyaluronic acid, a glucuronic acid, a glucosamine and a pectin.
28. The tablet according to claim 26, wherein the at least one hydrophilic compound is a hydroxyalkyl cellulose or a carboxyalkyl cellulose.
29. The tablet according to claim 26 wherein the at least one hydrophilic compound is a hydrophilic copolymeric substance.
30. The tablet according to claim 26, wherein the at least one hydrophilic compound is a hydrogel.
31. The tablet according to claim 26, wherein the at least one amphiphilic compound is selected from the group consisting of lecithin, a sphingomyelin, a phospholipid, a ceramide, an alkyl block copolymer, a salt of a sulphated alkyl acid, a polyoxyethylenated alkyl and a sorbitan derivative.
32. The tablet according to claim 26, wherein the at least one amphiphilic compound is lecithin.
33. The tablet according to claim 26, wherein the at least one lipophilic compound is selected from the group consisting a wax, stearic acid and stearin.
34. The tablet according to claim 26, wherein the at least one lipophilic compound is stearic acid.
35. The tablet according to claim 26, wherein the tablet further comprises a gastro-resistant film coating prepared by (e) coating the tablet of step (d).
36. The tablet according to claim 35, wherein the gastro-resistant film coating comprises acrylic and/or methacrylic acid polymers, or copolymers, or cellulose phthalate derivatives.
37. The tablet according to claim 35, wherein the gastro-resistant film coating comprises acrylic and methacrylic acid copolymers.
38. A method of treating a pathological condition of the colon comprising administering to a patient in need thereof the controlled release oral pharmaceutical composition according to claim 1, wherein the pathological condition is infectious colitis, bacillary dysentery, pseudomembranous colitis, travellers' diarrhoea, diverticular disease and/or diverticulitis.
39. A method of treating a pathological condition of the colon comprising administering to a patient in need thereof the controlled release oral pharmaceutical composition according to claim 1, wherein the pathological condition is travellers' diarrhoea.
40. A method of treating a pathological condition of the colon comprising administering to a patient in need thereof the controlled release oral pharmaceutical composition according to claim 1, wherein the pathological condition is diverticular disease and/or diverticulitis.
41. A method of treating a pathological condition of the colon comprising administering to a patient in need thereof the controlled release oral pharmaceutical composition according to claim 13, wherein the pathological condition is infectious colitis, bacillary dysentery, pseudomembranous colitis, travellers' diarrhoea, diverticular disease and/or diverticulitis.
42. A method of treating a pathological condition of the colon comprising administering to a patient in need thereof the controlled release oral pharmaceutical composition according to claim 13, wherein the pathological condition is travellers' diarrhoea.
43. A method of treating a pathological condition of the colon comprising administering to a patient in need thereof the controlled release oral pharmaceutical composition according to claim 13, wherein the pathological condition is diverticular disease and/or diverticulitis.
44. A method of treating a pathological condition of the colon comprising administering to a patient in need thereof the controlled release oral pharmaceutical composition according to claim 14, wherein the pathological condition is infectious colitis, bacillary dysentery, pseudomembranous colitis, travellers' diarrhoea, diverticular disease and/or diverticulitis.
45. A method of treating a pathological condition of the colon comprising administering to a patient in need thereof the controlled release oral pharmaceutical composition according to claim 14, wherein the pathological condition is travellers' diarrhoea.
46. A method of treating a pathological condition of the colon comprising administering to a patient in need thereof the controlled release oral pharmaceutical composition according to claim 14, wherein the pathological condition is diverticular disease and/or diverticulitis.
47. A method of treating a pathological condition of the colon comprising administering to a patient in need thereof the controlled release oral pharmaceutical composition according to claim 26, wherein the pathological condition is infectious colitis, bacillary dysentery, pseudomembranous colitis, travellers' diarrhoea, diverticular disease and/or diverticulitis.
48. A method of treating a pathological condition of the colon comprising administering to a patient in need thereof the controlled release oral pharmaceutical composition according to claim 26, wherein the pathological condition is travellers' diarrhoea.
49. A method of treating a pathological condition of the colon comprising administering to a patient in need thereof the controlled release oral pharmaceutical composition according to claim 26, wherein the pathological condition is diverticular disease and/or diverticulitis.
US14/471,509 2004-06-25 2014-08-28 Oral antimicrobial pharmaceutical compositions Abandoned US20140370093A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US14/471,509 US20140370093A1 (en) 2004-06-25 2014-08-28 Oral antimicrobial pharmaceutical compositions
US16/289,748 US20190192441A1 (en) 2004-06-25 2019-03-01 Oral antimicrobial pharmaceutical compositions
US17/114,057 US20210093576A1 (en) 2004-06-25 2020-12-07 Oral antimicrobial pharmaceutical compositions

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
IT001295A ITMI20041295A1 (en) 2004-06-25 2004-06-25 ORAL ANTI-MICROBIAL PHARMACEUTICAL COMPOSITIONS
ITMI2004A001295 2004-06-25
PCT/EP2005/052025 WO2006003043A1 (en) 2004-06-25 2005-05-03 Oral antimicrobial pharmceutical compositions
US57104407A 2007-02-05 2007-02-05
US13/451,111 US8263120B2 (en) 2004-06-25 2012-04-19 Oral antimicrobial pharmaceutical compositions
US13/592,097 US20120316229A1 (en) 2004-06-25 2012-08-22 Oral antimicrobial pharmaceutical compositions
US14/471,509 US20140370093A1 (en) 2004-06-25 2014-08-28 Oral antimicrobial pharmaceutical compositions

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US13/592,097 Continuation US20120316229A1 (en) 2004-06-25 2012-08-22 Oral antimicrobial pharmaceutical compositions

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/289,748 Continuation US20190192441A1 (en) 2004-06-25 2019-03-01 Oral antimicrobial pharmaceutical compositions

Publications (1)

Publication Number Publication Date
US20140370093A1 true US20140370093A1 (en) 2014-12-18

Family

ID=34968899

Family Applications (9)

Application Number Title Priority Date Filing Date
US11/571,044 Active 2027-04-02 US8486446B2 (en) 2004-06-25 2005-05-03 Oral antimicrobial pharmaceutical compositions
US12/133,568 Abandoned US20080233193A1 (en) 2004-06-25 2008-06-05 Oral antimicrobial pharmaceutical compositions
US13/451,111 Active US8263120B2 (en) 2004-06-25 2012-04-19 Oral antimicrobial pharmaceutical compositions
US13/592,088 Active US8741948B2 (en) 2004-06-25 2012-08-22 Oral antimicrobial pharmaceutical compositions
US13/592,097 Abandoned US20120316229A1 (en) 2004-06-25 2012-08-22 Oral antimicrobial pharmaceutical compositions
US13/616,582 Active US8529945B2 (en) 2004-06-25 2012-09-14 Oral antimicrobial pharmaceutical compositions
US14/471,509 Abandoned US20140370093A1 (en) 2004-06-25 2014-08-28 Oral antimicrobial pharmaceutical compositions
US16/289,748 Abandoned US20190192441A1 (en) 2004-06-25 2019-03-01 Oral antimicrobial pharmaceutical compositions
US17/114,057 Abandoned US20210093576A1 (en) 2004-06-25 2020-12-07 Oral antimicrobial pharmaceutical compositions

Family Applications Before (6)

Application Number Title Priority Date Filing Date
US11/571,044 Active 2027-04-02 US8486446B2 (en) 2004-06-25 2005-05-03 Oral antimicrobial pharmaceutical compositions
US12/133,568 Abandoned US20080233193A1 (en) 2004-06-25 2008-06-05 Oral antimicrobial pharmaceutical compositions
US13/451,111 Active US8263120B2 (en) 2004-06-25 2012-04-19 Oral antimicrobial pharmaceutical compositions
US13/592,088 Active US8741948B2 (en) 2004-06-25 2012-08-22 Oral antimicrobial pharmaceutical compositions
US13/592,097 Abandoned US20120316229A1 (en) 2004-06-25 2012-08-22 Oral antimicrobial pharmaceutical compositions
US13/616,582 Active US8529945B2 (en) 2004-06-25 2012-09-14 Oral antimicrobial pharmaceutical compositions

Family Applications After (2)

Application Number Title Priority Date Filing Date
US16/289,748 Abandoned US20190192441A1 (en) 2004-06-25 2019-03-01 Oral antimicrobial pharmaceutical compositions
US17/114,057 Abandoned US20210093576A1 (en) 2004-06-25 2020-12-07 Oral antimicrobial pharmaceutical compositions

Country Status (22)

Country Link
US (9) US8486446B2 (en)
EP (2) EP1894560A1 (en)
JP (2) JP5026262B2 (en)
KR (2) KR20080059337A (en)
CN (2) CN1976685B (en)
AT (1) ATE392889T1 (en)
CA (2) CA2569683C (en)
CY (1) CY1108196T1 (en)
DE (1) DE602005006278T2 (en)
DK (1) DK1763339T3 (en)
ES (1) ES2303255T3 (en)
FI (1) FIC20190031I1 (en)
HR (1) HRP20080252T3 (en)
HU (1) HUS1900037I1 (en)
IT (1) ITMI20041295A1 (en)
MX (1) MXPA06014698A (en)
PL (1) PL1763339T3 (en)
PT (1) PT1763339E (en)
RS (1) RS50581B (en)
SI (1) SI1763339T1 (en)
WO (1) WO2006003043A1 (en)
ZA (1) ZA200610577B (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11564883B2 (en) 2017-11-10 2023-01-31 Cosmo Technologies Ltd. Oral rifamycin SV compositions

Families Citing this family (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8497258B2 (en) 2005-11-12 2013-07-30 The Regents Of The University Of California Viscous budesonide for the treatment of inflammatory diseases of the gastrointestinal tract
EP3714878B1 (en) 2007-07-06 2022-01-19 Lupin Limited Pharmaceutical compositions of rifaximin
US20100239682A1 (en) * 2007-09-17 2010-09-23 Antoine Andremont Colonic delivery of antimicrobial agents
US9314444B2 (en) 2009-01-12 2016-04-19 Biokier, Inc. Composition and method for treatment of NASH
US9006288B2 (en) 2009-01-12 2015-04-14 Biokier, Inc. Composition and method for treatment of diabetes
KR20110120866A (en) 2009-01-12 2011-11-04 바이오키어 인코포레이티드 Composition and method for treatment of diabetes
WO2010144865A2 (en) 2009-06-12 2010-12-16 Meritage Pharma, Inc. Methods for treating gastrointestinal disorders
US9301938B2 (en) 2009-09-23 2016-04-05 Biokier, Inc. Composition and method for treatment of diabetes
WO2012016707A2 (en) * 2010-08-06 2012-02-09 Ratiopharm Gmbh Oral dosage form for the modified release of dimebolin
US20130259947A1 (en) * 2010-11-29 2013-10-03 Dr. Reddy's Laboratories Ltd. Oral metronidazole pharmaceutical compositions
IT202000005782A1 (en) 2020-03-18 2021-09-18 Dpl Pharma S P A CONTROLLED RELEASE PHARMACEUTICAL FORMULATIONS FOR THE TREATMENT OF INTESTINAL INFECTIONS

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4312866A (en) * 1979-07-10 1982-01-26 Dr. L. Zambeletti S.P.A. Use of rifamycin SV and its related salts in the treatment of rheumatoid arthritis, and related formulations suited for the purpose
US4409209A (en) * 1979-10-12 1983-10-11 Ciba-Geigy Corporation Novel phosphorylmuramyl peptides and processes for the manufacture thereof

Family Cites Families (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IT1230576B (en) * 1988-10-20 1991-10-28 Angeli Inst Spa ORAL PHARMACEUTICAL FORMULATIONS WITH SELECTIVE LIBERATION IN THE COLON
IE61651B1 (en) 1990-07-04 1994-11-16 Zambon Spa Programmed release oral solid pharmaceutical dosage form
IT1260505B (en) 1992-06-01 1996-04-09 Poli Ind Chimica Spa ORAL PHARMACEUTICAL SYSTEMS WITH DELAYED DELIVERY FOR THE SPECIFIC RELEASE IN THE COLON
CN1097306A (en) * 1993-07-12 1995-01-18 中国医科大学第二临床学院 Compound anti-inflammation suppository
US5849327A (en) * 1994-07-29 1998-12-15 Advanced Polymer Systems, Inc. Delivery of drugs to the lower gastrointestinal tract
US6861053B1 (en) * 1999-08-11 2005-03-01 Cedars-Sinai Medical Center Methods of diagnosing or treating irritable bowel syndrome and other disorders caused by small intestinal bacterial overgrowth
US6245351B1 (en) * 1996-03-07 2001-06-12 Takeda Chemical Industries, Ltd. Controlled-release composition
US5985823A (en) * 1997-06-09 1999-11-16 Ambi Inc. Method for the treatment of diarrheal disease and for eliminating particular bacterial populations from the colon
KR100412327B1 (en) * 1997-07-01 2003-12-31 화이자 인코포레이티드 Sertraline salts and sustained-release dosage forms of sertraline
DE19850445A1 (en) 1998-11-02 2000-05-04 Falk Pharma Gmbh Medicines for the topical treatment of inflammatory bowel disease
US6248363B1 (en) 1999-11-23 2001-06-19 Lipocine, Inc. Solid carriers for improved delivery of active ingredients in pharmaceutical compositions
GB9910773D0 (en) * 1999-05-11 1999-07-07 West Pharm Serv Drug Res Ltd Novel single unit coated dosage forms that can be used to minimise the effect of food on the absorption of drugs from the gastrointestinal tract
US7431943B1 (en) 1999-06-14 2008-10-07 Cosmo Technologies Limited Controlled release and taste masking oral pharmaceutical compositions
JP4393098B2 (en) 2002-06-21 2010-01-06 独立行政法人科学技術振興機構 New uses of ansamycin antibiotics and screening methods for new angiogenesis inhibitors
CN1201736C (en) * 2002-07-19 2005-05-18 安徽省药物研究所 Slow-releasing metronidazole prepn for position of colon
ES2312847T3 (en) 2002-08-26 2009-03-01 S.L.A. Pharma Ag TOPICAL FORMULATION THAT INCLUDES AT LEAST 10% METRONIZADOL IN WHITE VASELINE AND ITS USE IN THE ANAL AND RECTAL REGION.
WO2005030173A1 (en) 2003-09-25 2005-04-07 Ranbaxy Laboratories Limited Colon-specific drug delivery using interpolymer complexations

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4312866A (en) * 1979-07-10 1982-01-26 Dr. L. Zambeletti S.P.A. Use of rifamycin SV and its related salts in the treatment of rheumatoid arthritis, and related formulations suited for the purpose
US4409209A (en) * 1979-10-12 1983-10-11 Ciba-Geigy Corporation Novel phosphorylmuramyl peptides and processes for the manufacture thereof

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
FRATINI et al (Beeswax: A minireview of its antimicrobial activity and its application in medicine.Asian Pac J Trop Med. 2016 Sep;9(9):839-43. *
PEREZ et al (A synthetic alternative to natural lecithins with antimicrobial properties.Colloids Surf B Biointerfaces. 2004 Jun 1;35(3-4):235-42) *
RENDI-WAGNER et al (Drug Prophylaxis for Travelers' Diarrhea. Clin Infect Dis. (2002) 34 (5): 628-633) *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11564883B2 (en) 2017-11-10 2023-01-31 Cosmo Technologies Ltd. Oral rifamycin SV compositions

Also Published As

Publication number Publication date
SI1763339T1 (en) 2008-08-31
KR20080059337A (en) 2008-06-26
US20080233193A1 (en) 2008-09-25
HUS1900037I1 (en) 2019-07-29
US8529945B2 (en) 2013-09-10
US20120316229A1 (en) 2012-12-13
US8486446B2 (en) 2013-07-16
PL1763339T3 (en) 2008-10-31
DK1763339T3 (en) 2008-08-11
US20120316228A1 (en) 2012-12-13
WO2006003043A1 (en) 2006-01-12
KR101157486B1 (en) 2012-07-09
US20070248664A1 (en) 2007-10-25
JP2008503540A (en) 2008-02-07
ZA200610577B (en) 2008-07-30
CA2628005A1 (en) 2006-01-12
CA2569683C (en) 2012-04-03
DE602005006278T2 (en) 2009-10-08
JP5026262B2 (en) 2012-09-12
US20190192441A1 (en) 2019-06-27
HRP20080252T3 (en) 2008-07-31
US20130004569A1 (en) 2013-01-03
EP1894560A1 (en) 2008-03-05
KR20070028441A (en) 2007-03-12
US20210093576A1 (en) 2021-04-01
CN1976685A (en) 2007-06-06
ES2303255T3 (en) 2008-08-01
CY1108196T1 (en) 2014-02-12
CN101317826A (en) 2008-12-10
US8741948B2 (en) 2014-06-03
DE602005006278D1 (en) 2008-06-05
ITMI20041295A1 (en) 2004-09-25
CN1976685B (en) 2011-05-18
CA2569683A1 (en) 2006-01-12
US8263120B2 (en) 2012-09-11
EP1763339B1 (en) 2008-04-23
JP2008163038A (en) 2008-07-17
PT1763339E (en) 2008-06-19
RS50581B (en) 2010-05-07
FIC20190031I1 (en) 2019-05-28
EP1763339A1 (en) 2007-03-21
ATE392889T1 (en) 2008-05-15
MXPA06014698A (en) 2007-03-26
US20120207834A1 (en) 2012-08-16

Similar Documents

Publication Publication Date Title
US8529945B2 (en) Oral antimicrobial pharmaceutical compositions
JP5622773B2 (en) Tetracycline once a day formulation
CN111918646B (en) Delayed release deferiprone tablets and methods of use thereof
US5985823A (en) Method for the treatment of diarrheal disease and for eliminating particular bacterial populations from the colon
US20130259947A1 (en) Oral metronidazole pharmaceutical compositions
US5958873A (en) Oral formulation for treatment of bacteria-induced diseases of the colon
US20100272794A1 (en) Pharmaceutical composition of memantine
AU2008243202B2 (en) Oral antimicrobial pharmaceutical compositions
US20230330076A1 (en) Controlled release formulations of flavoxate and process for preparation thereof
AU2002300676B2 (en) A method for the treatment of diarrheal disease and for eliminating particular bacterial populations from the colon

Legal Events

Date Code Title Description
AS Assignment

Owner name: COSMO TECHNOLOGIES LTD., IRELAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:AJANI, MAURO;BOZZELLA, ROBERTA;CELASCO, GIUSEPPE;AND OTHERS;SIGNING DATES FROM 20070109 TO 20161018;REEL/FRAME:040072/0047

AS Assignment

Owner name: COSMO TECHNOLOGIES LTD., IRELAND

Free format text: ASSIGNEE ADDRESS CHANGE;ASSIGNOR:COSMO TECHNOLOGIES LTD.;REEL/FRAME:040261/0658

Effective date: 20151109

STCV Information on status: appeal procedure

Free format text: NOTICE OF APPEAL FILED

STCB Information on status: application discontinuation

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