WO2000045803A2 - Use of 5-aminosalicylates as antimicrobial agents - Google Patents

Use of 5-aminosalicylates as antimicrobial agents Download PDF

Info

Publication number
WO2000045803A2
WO2000045803A2 PCT/US2000/002802 US0002802W WO0045803A2 WO 2000045803 A2 WO2000045803 A2 WO 2000045803A2 US 0002802 W US0002802 W US 0002802W WO 0045803 A2 WO0045803 A2 WO 0045803A2
Authority
WO
WIPO (PCT)
Prior art keywords
pharmaceutical composition
food
aminosalicylate compound
antimicrobial
bacterial species
Prior art date
Application number
PCT/US2000/002802
Other languages
French (fr)
Other versions
WO2000045803A3 (en
Inventor
Henry C. Lin
Mark Pimentel
Original Assignee
Cedars-Sinai Medical Center
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 Cedars-Sinai Medical Center filed Critical Cedars-Sinai Medical Center
Priority to AU35879/00A priority Critical patent/AU3587900A/en
Publication of WO2000045803A2 publication Critical patent/WO2000045803A2/en
Publication of WO2000045803A3 publication Critical patent/WO2000045803A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/60Salicylic acid; Derivatives thereof
    • A61K31/606Salicylic acid; Derivatives thereof having amino groups
    • 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

Definitions

  • This invention relates to the medical arts. In particular, it relates to a method of inhibiting bacterial growth in the gastrointestinal tract of a human or non-human vertebrate by using an antimicrobial agent. 2. DISCUSSION OF THE RELATED ART
  • Antimicrobial or antibiotic agents are used in the treatment of bacterial infections, especially of the gastrointestinal tract. Gastrointestinal infections affect millions of people world-wide, especially children, and pose an increasing health hazard in hospital settings.
  • Bacteroides fragilis group and Clostridium species are common member-Spf the intestinal microflora of healthy individuals, and non-toxigenic strains can be transmitted without causing disease symptoms.
  • Bacteroides fragilis group and Clostridium species are common member-Spf the intestinal microflora of healthy individuals, and non-toxigenic strains can be transmitted without causing disease symptoms.
  • Duerden Bacteroides species in the normal neonatal faecal flora, J. Hyg. [Lond.] 87(2):299-304 [1981]; B.I. Duerden, The isolation and identification of Bacteroides spp. From the normal human faecal flora, J. Med. Microbiol. 13(l):69-78 [1980]; S.S. Long and R.M. Swenson, Development of anaerobic flora in healthy newborn infants, J. Pediatr. 91(2):298-301 [1977]). Other pathogenic bacterial strains have an adverse effect on their hosts.
  • enterotoxigenic strains of Bacteroides fragilis are associated with diarrhea in humans.
  • S.K. Niyogi et al. Association of enterotoxigenic Bacteroides fragilis with childhood diarrhoea, Indian J. Med. Res. 105: 167-69 [1997]
  • R.B. S&c etal Enterotoxigenic Bacteroides fragilis: epidemiologic studies of its role as a human diarrhoeal pathogen, J. Diarrhoeal Dis. Res. 10(l):4-9 [1992]).
  • patients with Crohn's disease were reported to have higher numbers of B. fragilis group bacteria in their intestines than healthy controls.
  • Clostridium species are gram-positive, spore-forming anaerobes; some strains that colonize the human intestines can, under certain circumstances, release potent protein exotoxins that induce inflammation of the intestinal mucosa.
  • M.L. Job and N.F. Jacobs, Jr. Drug-induced Clostridium difficile-associated disease, DmgSaf. 17(1):37-46 [1997]).
  • antibiotics and other chemotherapeutic agents can induce the expression of Toxins A and B from Clostridium difficile. (B.A. Cunha [1998]).
  • C. difficile-associated disease agents known to have a high potential to induce C. difficile- associated disease are aminopenicillins, cephalosporins and clindamycin.
  • M.L. Job and N.F. Jacobs, Jr. Drug-induced Clostridium difficile-associated disease, Drug. Saf. 17(l):37-46 [1997]
  • Y. Hutin et al. Prevalence of and risk factors for Clostridium difficile colonization at admission to an infectious diseases ward, Clin. Infect. Dis. 24(5):920-24 [1997]
  • CD. Settle and M.H. Wilcox [1996] are agents known to have a high potential to induce C. difficile- associated disease.
  • C. difficile An infection of C. difficile can add an average of three weeks to a patient' s hospital stay.
  • Symptoms may include, diarrhea, self-limited colitis, toxic megacolon or potentially lethal fulminant pseudomembranous colitis.
  • Intestinal infection with C. difficile has also been linked to reactive arthritis.
  • I.H. Kocar et al. Clostridium infection in patients with reactive arthritis of undetermined etiology, Scand. J. Rheumatol. 27(5):357-62 [1998]; R.K. Cleary, Clostridium difficile-associated diarrhea and colitis: clinical manifestations, diagnosis, and treatment, Dis. Colon.
  • Bacteraemia and subsequent sepsis is another possible complication of intestinal infection by C. difficile.
  • P. Naaber et al. Bacterial translocation, intestinal microflora and morphological changes of intestinal mucosa in experimental models of Clostridium difficile infection, J. Med. Microbiol. 47(7):591-98 [1998]; R.J. Feldman et al, Bacteremia due to Clostridium difficile: case report and review of extraintestinal C. Difficile infections, Clin. Infect. Dis. 20(6): 1560-62 [1995]).
  • In at least one nosocomial outbreak 17 patients died from C. difficile infection.
  • C. difficile is also thought to be a causal agent of wide-spread acute diarrheal disease.
  • S.K. Niyogi et al Prevalence of Clostridium difficile in hospitalized patients with acute diarrhoea in Calcutta, J. Diarrhoeal Dis. Res. 9(1): 16-19 [1991]; S.Q. Akhtar, Isolation of
  • Enterotoxigenic strains of C. perfringens are linked with a significant number of cases of antibiotic-associated diarrhea, especially among elderly hospitalized patients, children, and infants.
  • A. Wada et al Nosocomial diarrhoea in the elderly due to enterotoxigenic Clostridium perfringens, Microbiol. Immunol.40(10):767-71 [1996]; M.M. Brett et al, Detection of Clostridium perfringens and its enterotoxin in cases of sporadic diarrhoea, J. Clin. Pathol. 45(7):609-l 1 [1992]; S.C.
  • Clostridium perfringens has been implicated as a possible contributor to sudden infant death syndrome (SLDS) in susceptible infants.
  • SLDS sudden infant death syndrome
  • Clostridium perfringens is well known as a causative agent of non-gastrointestinal gangrene, a special problem for many elderly and diabetic patients with poor blood circulation. But also in more extreme cases of gastrointestinal infection, C. perfringens can cause enteritis necroticans, a gangrene of the bowel resulting in necrosis, sepsis, and hemolysis, in humans and domesticated animals.
  • C. perfringens can cause enteritis necroticans, a gangrene of the bowel resulting in necrosis, sepsis, and hemolysis, in humans and domesticated animals.
  • enteritis necroticans Although rare in developed countries, clostridial enteritis necroticans in humans is more common in some developing countries. (D.A. Watson et al, Pig-bel but no pig: enteritis necroticans acquired in Australia, Med. J. Aust. 155(l):47-50 [1991]). In New Guinea, enteritis necroticans, known locally as pigbel, has been a major cause of illness and death among children. (G.W. Lawrence et al, Impact of active immunisation against enteritis necroticans in Papua New Guinea, Lancet 336(8724): 1165-67 [1990]).
  • Clostridium perfringens type C the etiologic agent of enteritis necroticans, was also isolated from Bangladeshis with bloody or watery diarrheal illness. (F.P. van Loon et al, Clostridium perfringens type C in bloody and watery diarrhea in Bangladesh, Trop. Geogr. Med. 42(2): 123-27 [1990]).
  • Entertoxigenic strains of C. perfringens have also been linked to nosocomial and non-nosocomial outbreaks of food poisoning, due to heat-resistant spores and a rapid growth rate in warm food.
  • A.M. Pollack and P.M. Whitty Outbreak of Clostridium perfringens food poisoning, J. Hosp. Infect. 17(3): 179-86 [1991]
  • M. Van Damme-Jongsten et al Synthetic DNA probes for detection of enterotoxigenic Clostridium perfringens strains isolated from outbreaks of food poisoning, J. Clin. Microbiol. 28(l): 131-33 [1990]).
  • Botulism Spores of Clostridium botulinum germinating in warm food can cause another form of food poisoning called botulism.
  • vegetative cells of C. botulinum release an exotoxin that when consumed with the food is activated by trypsin in the stomach, and is absorbed intact by the blood stream.
  • the exotoxin binds to nerve cells, preventing the release of the neurotransmitter acetylcholine.
  • Resulting symptoms of botulism include blurred vision, difficulty in swallowing and speaking, and increasing muscular weakness, and usually nausea and vomiting. Death often results from paralysis of the muscles required for breathing.
  • Clostridium botulinum sometimes colonizes the intestines of infants and can cause infantile botulism, which can lead to respiratory paralysis and sudden infant death. Botulism is a problem for the food packaging industry. Spores of C. botulinum may not be killed if canning is done at too low a temperature. High temperature autoclave treatment may be unsuitable for some foods Mayonnaise and other non-acidic foods are particularly prone to foster the growth of C. botulinum. Now with increasing health concerns about the use of nitrite as a food preservative, alternative antimicrobial agents are needed against the growth of C. botulinum and other food poisoning bacterial pathogens.
  • Antimicrobial agents with selective toxicity for a specific spectrum or range of pathogenic microorganisms are well known in the art.
  • One class of antimicrobial agents is the antibiotics, which are compounds, synthesized and excreted by various microorganisms, that are selectively toxic to other microorganisms, specifically bacteria.
  • some antibiotics can be artificially modified to produce antimicrobial agents that are more effective and/or more able to overcome antibiotic resistance.
  • Clostridium has been a 10-day course of metronidazole or vancomycin, which may be administered orally, intravenously, or rectally.
  • metronidazole farnesoid senor
  • anaerobe e.g., P. Muir et al. , Breath hydrogen excretion by healthy cats after oral administration of oxytetracycline and metronidazole, Vet. Rec. 138:635-39 [1996]; B. Lembcke et al. Different actions of neomycin and metronidazole on breath hydrogen (H 2 ) exhalation, Z. Gastroenterol. 18(3): 155-60 [1980]), it yields an unpleasant after-taste to many patients, even when delivered intravenously. Other common side effects of metronidazole are neuropathy and gastrointestinal distress.
  • metronidazole is a known reproductive toxicant affecting mammalian sperm cells. (R.E. Linder et al, Endpoints of spermatotoxicity in the rat after short duration exposures to fourteen reproductive toxicants, Reprod. Toxicol. 6(6):491-505 [1992]).
  • vancomycin On the other hand, a course of vancomycin is prohibitively expensive (10-50 times more expensive than metronidazole), and there are concerns about the rapid development of vancomycin- resistance among pathogenic Clostridium, Enterococcus, Pediococcus, Citrobacter, Klebsiella, Enterobacter, and Staphylococcus species, because the plasmid-borne vancomycin resistance gene (VanR) is readily transmissible.
  • vancomycin resistance gene VanR
  • Teicoplanin is another antibiotic found to be effective against gram positive anaerobes such as Propionibacterium acnes, Clostridium perfringens, C. difficile, and other Clostridium spp., Peptococcus spp., Peptostreptococcus spp. (H. Hassan et al, In vitro activity of teicoplanin, vancomycin, A16686, clindamycin, erythromycin andfusidic acid against anaerobic bacteria, Singapore Med. J.
  • teicoplanin is not widely available.
  • the peptide antibiotic bacitracin is also reported to be effective in treating C. difficile-induced diarrhea, but it is not widely available in an enteric formulation. (M.N. Dudley et al. , Oral bacitracin vs. vancomycin therapy for Clostridium difficile- induced diarrhea, A randomized double-blind trial, Arch. Intern. Med. 146(6): 1101-04 [1986]).
  • adjunct treatments are reportedly effective for refractory C. difficile-related disease, including whole-bowel irrigation and enteric administration of the non-pathogenic yeast S ⁇ cch ⁇ romyces boul ⁇ rdii.
  • CA. Liacouras and D.F.A. Piccoli Whole-bowel irrigation as an adjunct to the treatment of chronic, relapsing Clostridium difficile colitis, J. Clin. Gastroenterol. 22(3): 186-89 [1996]
  • CM. Surawicz Clostridium difficile disease: diagnosis and treatment, Gastroenterologist 6(1 ):60-65 [1998]).
  • But such treatments are uncomfortable or distasteful for many patients and are less suitable than easily administered antibiotics as a first line treatment regimen.
  • 4-(p)-aminosalicylic acid i.e., 4-ASA or para-aminosalicylic acid
  • 4-(p)-aminosalicylate sodium salt e.g., Nemasol-Sodium ® or Tubasal ®
  • the 5-aminosalicylates are known as anti-inflammatory chemotherapeutic agents and have not been used as antimicrobial agents. These compounds include 5-aminosalicylic acid (i.e., 5-ASA, mesalamine, or mesalazine) and conjugated derivatives thereof, known for their anti- inflammatory properties. These anti-inflammatory agents are commercially available in various pharmaceutical preparations such as Asacol ® , Rowasa ® , Claversal ® , Pentasa ® , Salofalk ® , Dipentum ® , Azulfidine ® (SAZ) and others.
  • Asacol ® i.e., 5-ASA, mesalamine, or mesalazine
  • conjugated derivatives thereof known for their anti- inflammatory properties.
  • anti-inflammatory agents are commercially available in various pharmaceutical preparations such as Asacol ® , Rowasa ® , Claversal ® , Pentasa ® , Salofalk ® ,
  • 5-Aminosalicylates have been used widely to reduce mucosal inflammation in inflammatory bowel disease, ulcerative colitis and Crohn' s disease.
  • S.B. Hanauer and FB. Baert Medical Therapy of Inflammatory Bowel Disease, Inflamm. Bowel Dis. 78(6): 1413-25 [1994]
  • C.J. Mulder et al Drug therapy: dose-response relationship of oral mesalazine in inflammatory bowel disease, Mediators Inflamm. 7(3): 135-36 [1998]
  • W. Kruis et al Olsalazine versus mesalazine in the treatment of mild to moderate ulcerative colitis, Aliment.
  • 5-aminosalicylates The mechanism underlying the anti-inflammatory properties of the 5-aminosalicylates is unknown, but it may result from their ability to inhibit oxidation at the surface of endothelial membranes, perhaps through radical scavenging, and to prevent lipid peroxidation.
  • 5- Aminosalicylates may be able to act synergistically with endogenous antioxidants such as alpha- tocopherol to prevent the oxidative damage implicated in the pathogenesis of inflammatory bowel diseases.
  • the method of the present invention employs the antimicrobial properties of 5-aminosalicylates in a method of inhibiting bacterial growth in a human or non-human vertebrate.
  • a pharmaceutical composition comprising a 5-aminosalicylate
  • the growth of bacterial species can be arrested, including the growth of anaerobic pathogens of the genus Clostridium, for example, C. perfringens, C. difficile, C. botulinum and C. tetani.
  • the present method can be used to treat the gastrointestinal tract or any other non-gastrointestinal body site or tissue to inhibit bacterial growth.
  • the present method also has veterinary applications and can be used to treat a wide variety of non-human vertebrates, including wild, domestic and farm animals.
  • the present invention is also related to an antimicrobial method for inhibiting the growth of a bacterial species in a foodstuff and to foodstuffs containing a 5-aminosalicylate compound, useful for preventing food poisoning or botulism.
  • the present invention is also related to food containers and food- handling implements with bacteriostatic properties, intended for holding a foodstuff, and to cleansers, polishes, paints, sprays, soaps, and detergents for inhibiting the growth of a bacterial species on surfaces.
  • the 5-aminosalicylates are generally well-tolerated and competitively priced compounds that can fill a definite need for an antimicrobial agent, which is selectively effective against gastrointestinal and non-gastrointestinal clostridial diseases.
  • the present invention is directed to the use of 5-aminosalicylates as antimicrobial agents in a method of inhibiting bacterial growth in a human or non-human vertebrate, in the gastrointestinal tract or any other body site or tissue.
  • the method involves administering to a human or non-human vertebrate subject a pharmaceutically acceptable composition of the present invention.
  • compositions in accordance with the present invention are formulated for acceptable delivery to a human or non-human vertebrate gastrointestinal tract or other body site or tissue. Topical delivery to gastrointestinal mucosa and/or systemic delivery thereto are intended. Also included are pharmaceutically acceptable formulations for systemic or topical delivery to a non-gastrointestinal body site or tissue subject to bacterial growth, and particularly subject to clostridial growth.
  • the present invention is also related to pharmaceutically acceptable compositions containing a 5-aminosalicylate compound.
  • Five-aminosalicylate compounds include 5-aminosalicylic acid or any compound containing a 5-aminosalicylate moiety, or any conjugated derivative thereof, that is effective in inhibiting the growth of a bacterial species in a human or non-human vertebrate gastrointestinal tract or any other body site, limb, organ, such as heart, lung or kidney, or any other tissue of a vertebrate body.
  • a most preferred 5-aminosalicylate compound is 5-aminosalicylic acid, also known as mesalamine or mesalazine, and commercially available, for example, in pharmaceutical preparations known as Asacol ® (Procter and Gamble), Pentasa ® (Ferring A/S Vanlose), Claversal ® , Mesasal ® , Rowasa ® , and Salofalk ® (Dr. Falk GmbH).
  • Asacol ® Procter and Gamble
  • Pentasa ® Fering A/S Vanlose
  • Claversal ® Mesasal ®
  • Mesasal ® Mesasal ®
  • Rowasa ® Rowasa ®
  • Salofalk ® Dr. Falk GmbH
  • 5-aminosalicylate salts including the sodium or potassium salts of conjugated 5-aminosalicylates.
  • 5-aminosalicylate compounds for purposes of the present invention, are compounds containing a 5-aminosalicylic acid moiety, or a 5-aminosalicylate moiety, conjugated to another 5-aminosalicylic acid moiety or 5-aminosalicylate moiety, or conjugated to a pharmaceutically acceptable carrier molecule.
  • Preferred conjugated 5-aminosalicylate compounds include, but are not limited to, olsalazine or Poly-ASA.
  • Olsalazine is known variously as disodium-diazosalicylate, azodisalicylate, or Dipentum ® (Pharmacia).
  • Olsalazine comprises two 5-aminosalicylate moieties linked through a diazobond.
  • Poly- ASA comprises a water-soluble polymer that contains 5-aminosalicylate moieties linked through diazobonds to an inert sulfanilamide ethylene polymer.
  • Balsalazide (balsalazine) is another preferred conjugated 5-aminosalicylate compound.
  • Balsalazide is more effective and better tolerated than mesalamine in the treatment of acute ulcerative colitis.
  • Abacus Investigator Group Aliment. Pharmacol. Ther. 12(12): 1207-16 [1998b]).
  • Another preferred conjugated 5-aminosalicylate compound is sulphasalazine, also known as sulfasalazine, salazosulfapyridine, or Azulfidine ® , in which a 5-aminosalicylic acid moiety is linked through a diazobond to sulfapyridine, the carrier molecule.
  • sulphasalazine also known as sulfasalazine, salazosulfapyridine, or Azulfidine ®
  • a 5-aminosalicylic acid moiety is linked through a diazobond to sulfapyridine, the carrier molecule.
  • sulfapyridine moiety and may include headache, nausea, vomiting, fever, rash, epidermolysis, hemolytic anemia, pancreatitis, pulmonary fibrosis, agranulocytosis, or liver toxicity; the use of sulphasalazine should be discontinued immediately when these symptoms occur.
  • S.B. Hanauer and G. Stathopoulos Risk-benefit assessment of drugs used in the treatment of inflammatory bowel disease, Drug Saf. 6(3): 192-219 [1992]; H. Allgayer [1992]). Impairment of male fertility is another possible side effect of sulphasalazine related to sulfapyridine.
  • V.A. Botoman and G.F. Bonner Management of inflammatory bowel disease, Am. Fam. Physician 57(l):57-68 [1998]).
  • conjugated 5-aminosalicylate compounds are ipsalazine (ipsalazide) and salicylazobenzoic acid; these are also well tolerated molecules comprising a 5-aminosalicylic acid moiety or 5-aminosalicylate moiety linked by a diazobond to another pharmaceutically acceptable chemical unit.
  • conjugated 5-aminosalicylate compounds for gastrointestinal use are bile acids conjugated with a 5-aminosalicylic acid moiety or 5-aminosalicylate moiety.
  • ursodeoxycholic acid-5-aminosalicylic acid ursodeoxycholic acid is linked through an amide bond to 5-aminosalicylic acid.
  • the conjugated 5-aminosalicylate compound is synthesized by adding a basic solution of 5-aminosalicylic acid into the mixed anhydride of ursodeoxycholic acid and ethyl chloroformate. (A.K.
  • certain hydrolases e.g., cholylglycine hydrolase
  • Other particular varieties of 5-aminosalicylate conjugate can be subject to deconjugation by other enzyme systems. Whether or not a 5-aminosalicylic acid or 5-aminosalicylate moiety is actually deconjugated from a carrier molecule or from another 5-aminosalicylate moiety, in a human or non-human vertebrate gastrointestinal tract or other body site, does not limit the embodiments of a 5- aminosalicylate compound contemplated by the present invention.
  • the present invention is not committed to any particular mechanism by which a particular 5-aminosalicylate compound operates to inhibit bacterial growth.
  • the 5-aminosalicylate compound is administered by any suitable method.
  • Representative methods include giving, providing, feeding or force-feeding, dispensing, inserting, prescribing, furnishing, treating with, taking, swallowing, eating or applying a pharmaceutical composition of the present invention.
  • the pharmaceutical compositions of the present invention can optionally contain pharmaceutically acceptable solvent(s), adjuvant(s) or non-medicinal carrier(s), binder(s), thickener(s), or filler substance(s) that are known to the skilled artisan.
  • pharmaceutically acceptable solvent(s), adjuvant(s) or non-medicinal carrier(s), binder(s), thickener(s), or filler substance(s) that are known to the skilled artisan.
  • Common fillers include, but are not limited to, sucrose or lactose, or polymeric substances like starch.
  • additional medicinal or nutritive additives in combination with a 5-aminosalicylate compound as may be desired to suit the more particular needs of the practitioner.
  • a dose effective to inhibit the growth of a bacterial species is such dose as sufficient to prevent cellular proliferation of a bacterial species, by either killing bacterial cells or by preventing or slowing cellular growth or reproduction of a bacterial species, compared to the rate of growth or reproduction in the absence of a 5-aminosalicylate compound.
  • a 5-aminosalicylate compound By way of example, 12.5 mg or more of 5-aminosalicylic acid have been found sufficient to inhibit the growth of Clostridium perfringens on 10-mL blood agar plates.
  • the effective dose for each human or non-human vertebrate subject will depend upon the size and physiologic reactions of the subject to whom or to which the pharmaceutical preparations of the present invention are administered. And these reactions and the antimicrobial activity of the administered dose can be monitored by the prescribing physician or veterinarian.
  • the pharmaceutical compositions of the present invention can be formulated and manufactured at more than one concentration, such that modular incremental amounts of 5-aminosalicylate compound are easily administered.
  • 5-aminosalicylate compounds can be obtained in variable dosage units, including, for example: 250 mg (e.g., Salofalk ® , Pentasa ® , Azulfadine ® , Dipentum ® , or olsalazine); 400 mg (e.g., Asacol ® ); 0.5, 1.5 g or 3.0 g (e.g., Pentasa ® , Rowasa ® , Azulfadine ® , balsalazide).
  • 250 mg e.g., Salofalk ® , Pentasa ® , Azulfadine ® , Dipentum ® , or olsalazine
  • 400 mg e.g., Asacol ®
  • 0.5, 1.5 g or 3.0 g e.g., Pentasa ® , Rowasa ® , Azulfadine ® , bals
  • a minimum effective dose is as little as between 6.25 and 12.5 mg of a 5-aminosalicylate compound per day.
  • Effective doses of a 5-aminosalicylate compound at 12.5 to 150 mg per day and 150 to 250 mg per day are sufficient for smaller human adults, children, and smaller non-human vertebrates, such as rodents, canines, chickens and turkeys.
  • a higher effective dose for an adult human is 250-6400 mg of a 5-aminosalicylate per day.
  • Pediatric doses are typically 10-20% of effective doses for adult humans. Higher effective doses, from 6,450 mg/day, up to 1,000 mg/kg body mass per day can be used for large non-human vertebrates, for example, for sheep and larger animals such as cattle, horses, and elephants.
  • Antimicrobial activity by 5-aminosalicylate compounds against a specific bacterial species of interest is determined by routine means well known to the skilled practitioner. For example, a "lawn" of a bacterial species can be plated on an appropriate solid medium, and zones of growth inhibition around sterile cellulose disks impregnated with a 5-aminosalicylate compound of interest can be measured. Similarly, inhibition assays in liquid media are also routinely accomplished.
  • Inhibition of bacterial growth in a gastrointestinal tract is measured by conventional means well known to the skilled artisan. Many fermentative bacterial species found in the gastrointestinal tract produce detectable quantities of hydrogen or methane gas in the presence of certain sugars, which gases enter the blood stream of the host and are exhaled. This is the basis for intestinal bacterial growth detection means, such as, but not limited to, the lactulose, glucose, or lactose breath hydrogen tests.
  • intestinal bacterial growth detection means such as, but not limited to, the lactulose, glucose, or lactose breath hydrogen tests.
  • P. Kerlin and L. Wong Breath hydrogen testing in bacterial overgrowth of the small intestine, Gastroenterol. 95(4):982-88 [1988]
  • A. Strocchi et al Detection of malabsorption of low doses of carbohydrate: accuracy of various breath H 2 criteria, Gastroenterol. 105(5): 1404-1410 [1993]).
  • bacterial growth in a gastrointestinal tract is measured by detection of 13 CO 2 or 14 C0 2 breath emissions after administering an isotope-labeled sugar that is metabolizable by gastrointestinal bacteria but non-digestible by the host, such as, but not limited to, xylose or lactulose in humans.
  • an isotope-labeled sugar that is metabolizable by gastrointestinal bacteria but non-digestible by the host, such as, but not limited to, xylose or lactulose in humans.
  • Direct gastrointestinal sampling or biopsy from any body site or tissue can also be used to measure the inhibition of bacterial growth in a gastrointestinal tract or other body site or tissue.
  • direct sampling at time intervals provides information about the growth inhibition of specific bacterial species of interest, to which breath testing is not well-suited.
  • Samples are diluted and bacterial numbers can be assessed by conventional microbiological means such as, but not limited to colony plating or Most Probable Number (MPN) techniques, or direct counting of bacterial cells.
  • MPN Most Probable Number
  • FACS fluorescence activated cell sorting
  • evidence of inhibition of bacterial growth can be inferred by the practitioner treating a bacterial infection or intestinal bacterial overgrowth in a human or non-human vertebrate subject with observation of an improvement in various infection-or overgrowth-related symptoms in response to the administration of an antimicrobial composition of the present invention.
  • 5-aminosalicylic acid is an antimicrobial agent that does not affect many beneficial or commensal gastrointestinal bacteria but selectively inhibits potentially pathogenic clostridial species, such as, but not limited to, C. perfringens, C. difficile, C. tetani and C. botulinum.
  • the method of the present invention is also useful for veterinary purposes.
  • An antimicrobial composition comprising a 5-aminosalicylate compound can be administered to a non-human vertebrate including, but not limited to, a wild, domestic or farm animal.
  • the present method is useful for treating a mammal such as a non-human primate, mouse, rat, rabbit, gerbil, hamster, canine, feline, ovine, bovine, swine, pachyderm, equine, or marine mammal.
  • the method is useful to inhibit the growth of bacteria in a bird (avian) or poultry, such as a duck, chicken, goose, turkey, ostrich, emu, dove, pigeon, quail, pheasant, peafowl, or guinea fowl.
  • avian avian
  • poultry such as a duck, chicken, goose, turkey, ostrich, emu, dove, pigeon, quail, pheasant, peafowl, or guinea fowl.
  • the pharmaceutically acceptable composition is administered by a non- systemic delivery route to the site of bacterial infection or overgrowth that is not primarily by way of the blood stream of a human or non-human vertebrate.
  • suitable non-systemic delivery routes include, but are not limited to, an ingestive delivery route or a colonic delivery route.
  • a most preferred delivery route is an ingestive delivery route, whereby the antimicrobial agent enters the gastrointestinal or digestive tract by way of voluntary or forced ingestion through the mouth.
  • the organs of a gastrointestinal tract include the esophagus, stomach, large intestine, small intestine, or rectum.
  • the skilled artisan will be aware that in a non-human vertebrate the digestive tract may include a rumen, crop, gullet, cecum, or other specialized organ as pertains to a particular vertebrate species.
  • Another non-systemic delivery route is useful for non-gastrointestinal bacterial infections, particularly infections of the skin or externally accessible wounds; this delivery route is topical application to the affected area of an antimicrobial cream, gel, or ointment.
  • compositions of the present invention employing a non-systemic delivery route
  • a suppository or foam for delivery of a composition comprising a 5-aminosalicylate compound via anus or rectum to the colon.
  • a 5- aminosalicylate compound of the present invention will act topically at the intestinal mucosa.
  • Such suppository or foam delivery systems are known in the art; a commercially available example is Rowasa ® mesalamine suppositories for anti-inflammatory purposes.
  • the pharmaceutical preparation of the present invention such as a suppository, can employ a variety of conventional thickeners, such as alginate, xanthan gum, or petrolatum.
  • suppositories or foams comprising hydrophilic polymers, such as hydroxypropyl cellulose, hydroxypropyl methylcellulose, hydroxyethylcellulose, dextran, pectin, poly vinyl pyrrolidone, starch, gelatin, or any of a number of other polymers known to be useful for this purpose.
  • hydrophilic polymers such as hydroxypropyl cellulose, hydroxypropyl methylcellulose, hydroxyethylcellulose, dextran, pectin, poly vinyl pyrrolidone, starch, gelatin, or any of a number of other polymers known to be useful for this purpose.
  • compositions of the present invention is a gel for non- systemic delivery of a composition comprising a 5-aminosalicylate compound via the colon, similar to gels commonly used for the delivery of other chemotherapeutic agents.
  • Hydrogel matrices are known for this purpose. (Feijen, Biodegradable hydrogel matrices for the controlled release of pharmacologically active agents, U.S. Pat. No. 4,925,677).
  • Such biodegradable gel matrices may be formed, for example, by cross-linking a proteinaceous component and a polysaccharide or mucopolysaccharide component, then loading with a 5-aminosalicylate compound, for deli verability over an extended period.
  • compositions comprising a 5-aminosalicylate compound in an osmotically suitable enema solution.
  • Commercially available preparations include Rowasa ® mesalamine enemas for anti-inflammatory purposes. (See, R.N. Brogden and E.M. Sorkin, Mesalazine. A review of its pharmacodynamic andpharmacokinetic properties, and therapeutic potential in chronic inflammatory bowel disease, Drugs 38(4):500-24 [1989]).
  • a most preferred embodiment of the pharmaceutical composition of the present invention is formulated for a non-systemic ingestive delivery system, such as, but not limited to a tablet, capsule, or caplet.
  • the non-systemic ingestive delivery system comprises an enteric coating to prevent esophageal or gastric release of 5-aminosalicylate compound.
  • enteric-coated pharmaceuticals disintegrate after leaving the stomach, resulting in drug dispersion in the small intestine or colon where 5-aminosalicylates will act topically at the intestinal mucosa.
  • enteric-coated drug delivery systems are typically pH-sensitive, polymer-coated tablets, capsules, or caplets.
  • a polymer coating can be selected that will direct release of a 5-aminosalicylate compound to a particular region of the gut.
  • Such polymers include, but are not limited acrylic polymers such as Eudragit-L or Eudragit-S, and cellulosic polymers, such as ethylcellulose.
  • enteric-coated 5-aminosalicylates are mesalamine preparations known as Asacol ® and Claversal ® , used as anti-inflammatory agents.
  • the acrylic polymer coating of Claversal ® starts to dissolve at pH 6.0 after passing through the far more acidic milieu of the stomach, and as a result, mesalamine is reliably released in the distal small intestine (ileum) and colon of a human patient.
  • Salofalk ® is another example; it is coated first with a semipermeable layer of ethylcellulose and second with an acrylic polymer (i.e., Eudragit-L), which is degraded at above pH 5.6 in the distal small intestine and colon to release mesalamine there.
  • Pentasa ® contains 5-aminosalicylic acid microgranules coated with a semipermeable membrane of amphionic ethylcellulose, which can dissolve at either acidic or basic pH to release the microgranules. (F. Martin [1987]).
  • Pentasa ® is an example of a commercially available ingestible slow release form of a 5-aminosalicylate compound.
  • Pentasa See P. Fockens et al, Comparison of the efficacy and safety of 1.5 compared with 3.0 g oral slow release mesalazine [Pentasa] in the maintenance treatment of ulcerative colitis. Dutch Pentasa Study Group, Eur. J. Gastroenterol. Hepatol. 7(11): 1025-30 [1995]).
  • Another preferred ingestive delivery system comprises a lavage system, whereby a patient will ingest a large volume of an osmotically balanced flushing solution, containing 5-aminosalicylic acid, or a conventional flushing solution in conjunction with another ingestible form of a 5-aminosalicylate antimicrobial agent.
  • a lavage system can virtually eliminate bacterial populations from the intestine. This may be especially desirable in refractory cases of bacterial overgrowth or in preparing a patient for abdominal surgery.
  • a pharmaceutical preparation of the present invention is formulated and prepared to be ingested by an animal along with its food, as part of a pharmaceutically acceptable feed mixture.
  • a pharmaceutically acceptable food additive for humans is also contemplated.
  • a preferred embodiment of the antimicrobial method of the present invention involves a systemic delivery route, i.e., a route whereby delivery of a 5-aminosalicylate compound to the site of infection or bacterial growth is primarily via the blood stream. This embodiment can be used to inhibit bacterial growth in any body site or tissue, including the gastrointestinal tract.
  • a systemic delivery route is also particularly, but not exclusively useful for gastrointestinally infected patients who are unable to effectively ingest a non-systemic formulation of the composition of the present invention due to persistent nausea, difficulty in swallowing, or other ingestion-preventing conditions, or who, due to resection or other condition of the bowel cannot accept a colonic delivery system.
  • a systemic delivery route can be employed to deliver a 5-aminosalicylate compound to body sites or tissues other than those of the gastrointestinal tract, including, but not limited to, skin, heart, lung, blood, kidney, liver, brain, arms, legs, digits, sexual organs, trunk, head, neck, or tail.
  • Applications can include but are not limited to treating or preventing clostridial infections at any body site or tissue of a vertebrate.
  • Such clostridial infections include, but are not limited to, gangrene or tetanus, caused, respectively, by C. perfringens and C. tetani, when these species grow in wounds and damaged tissues with low oxygen tension.
  • a systemic delivery route can also include delivery through the skin, mucosa or epithelium of the mouth including the sublingual epithelium, the rectum, or the vaginal epithelium.
  • Systemic delivery systems that are contemplated by the present invention include, but are not limited to, ingestion, injection, or intravenous drip, most conventionally.
  • Other useful systemic delivery systems are known and include, but are not limited to, implant; adhesive transdermal patches; topical creams, gels or ointments for transdermal delivery; transmucosal delivery matrices or suppositories or gels.
  • the compositions of the present invention are formulated to deliver an effective dose of a 5-aminosalicylate compound by these or any other pharmaceutically acceptable systemic delivery system.
  • a preferred embodiment of the compositions of the present invention employing a systemic delivery route is a topical cream, ointment or gel to be applied to the skin.
  • a composition of the present invention comprises a 5-aminosalicylate compound in a pharmaceutically acceptable delivery system comprising a permeation or penetration enhancer, such as polyethylene glycol monolaurate, dimethyl sulfoxide, N-vinyl-2-pyrrolidone, N-(2-hydroxyethyl)-pyrrolidone, or 3-hydroxy- N-methyl-2-pyrrolidone.
  • a permeation or penetration enhancer such as polyethylene glycol monolaurate, dimethyl sulfoxide, N-vinyl-2-pyrrolidone, N-(2-hydroxyethyl)-pyrrolidone, or 3-hydroxy- N-methyl-2-pyrrolidone.
  • a permeation or penetration enhancer such as polyethylene glycol monolaurate, dimethyl sulfoxide, N-vinyl-2-pyrrolidone, N-(2-hydroxyethyl)-pyrrolidone, or 3-hydroxy- N-methyl-2-pyrrolidone.
  • compositions of the present invention is a formulation for systemic transmucosal delivery of a 5-aminosalicylate compound.
  • a variety of pharmaceutically acceptable systems for transmucosal delivery of therapeutic agents are known in the art and are compatible with the practice of the present invention. (Heiber et al, Transmucosal delivery of macromolecular drugs, U.S. Pat. Nos. 5,346,701 and 5,516,523; Longenecker et al, Transmembrane formulations for drug administration, U.S. Pat. No. 4,994,439).
  • Transmucosal delivery devices may be in free form, such as a cream, gel, or ointment, or may comprise a determinate form such as a tablet, patch, or troche.
  • delivery of a 5-aminosalicylate compound may be via a transmucosal delivery system comprising a laminated composite of, for example, an adhesive layer, a backing layer, a permeable membrane defining a reservoir containing a 5-aminosalicylate compound a peel seal disc underlying the membrane, one or more heat seals, and a removable release liner.
  • a tablet or patch for delivery through the oral mucosa can comprise an inner layer containing the therapeutic agent of choice, a permeation enhancer, such as a bile salt or fusidate, and a hydrophilic polymer, such as hydroxypropyl cellulose, hydroxypropyl methylcellulose, hydroxyethylcellulose, dextran, pectin, poly vinyl pyrrolidone, starch, gelatin, or any of a number of other polymers known to be useful for this purpose.
  • This inner layer can have one surface adapted to contact and adhere to the moist mucosal tissue of the oral cavity and may have an opposing surface adhering to an overlying non-adhesive inert layer.
  • such a transmucosal delivery system can be in the form of a bilayer tablet, in which the inner layer also contains additional binding agents, flavoring agents, or fillers.
  • Some useful systems employ a non-ionic detergent along with a permeation enhancer. These examples are merely illustrative of available transmucosal delivery technology and are not limiting of the present invention.
  • compositions of the present invention is a gel for systemic delivery of a 5-aminosalicylate via the rectal or vaginal mucosa, similar to gels commonly used for the delivery of various other therapeutic agents.
  • Hydrogel matrices are known for this purpose. (Feijen, Biodegradable hydrogel matrices for the controlled release of pharmacologically active agents, U.S. Pat. No. 4,925,677).
  • biodegradable gel matrices can be formed, for example, by cross-linking a proteinaceous component and a polysaccharide or mucopolysaccharide component, then loading with a 5-aminosalicylate compound to be delivered.
  • compositions of the present invention is the systemic delivery of a 5-aminosalicylate compound via a biodegradable matrix implanted within the body or under the skin of a human or non-human vertebrate.
  • the implant matrix may be a hydrogel similar to those described above. Alternatively, it may be formed from a poly-alpha-amino acid component. (Sidman, Biodegradable, implantable drug delivery device, and process for preparing and using same, U.S. Pat. No. 4,351,337).
  • a preferred embodiment of the composition of the present invention employing a systemic delivery route is a transdermal delivery system of a kind known in the art for delivering various drugs.
  • Transdermal delivery systems can be of any number of varieties known in the art.
  • delivery of a 5-aminosalicylate compound can be via a transdermal delivery system comprising a laminated composite of an adhesive layer, a backing layer, a permeable membrane defining a reservoir containing a 5-aminosalicylate compound, a peel seal disc underlying the membrane, one or more heat seals, and a removable release liner.
  • a transdermal delivery system with adhesive overlay and peel seal disc U.S. Pat No. 5,662,925; Chang et al. Device for administering an active agent to the skin or mucosa, U.S. Pat. Nos. 4,849,224 and 4,983,395).
  • a transdermal delivery device can be a matrix type transdermal patch.
  • a matrix type transdermal patch Chien et al, Transdermal estrogen/progestin dosage unit, system and process, U.S. Pat. Nos. 4,906,169 and 5,023,084; Cleary et al, Diffusion matrix for transdermal drug administration and transdermal drug delivery devices including same, U.S. Pat. No.4,911,916;
  • Sectionlaud et al EVA-based transdermal matrix system for the administration of an estrogen and/or a progestogen, U.S. Pat. No. 5.605,702; Venkateshwaran et al, Transdermal drug delivery matrix for coadministering estradiol and another steroid, U.S.
  • the matrix of the patch can comprise a basal support layer, such as an acrylic or ethylene/vinyl acetate copolymer or a polyurethane foam or cellulosic material, and an adhesive, such as, but not limited to, polysiloxane.
  • a basal support layer such as an acrylic or ethylene/vinyl acetate copolymer or a polyurethane foam or cellulosic material
  • an adhesive such as, but not limited to, polysiloxane.
  • the polymer matrix also contains a 5-aminoslaicylate compound, as described above, and optionally, a penetration-enhancing vehicle or carrier, such as N-vinyl-2-pyrrolidone, N-(2- hydroxyethyl)-pyrrolidone, or 3-hydroxy-N-methyl-2-pyrrolidone.
  • the adhesive patch may be pressure- sensitive, to release the 5-aminosalicylate compound across the skin of the patient when the patch matrix has been applied to the skin.
  • the patch may optionally comprise an inert backing layer in addition to a matrix layer, or can comprise multiple dosage units or layers.
  • the present invention is also related to a method of inhibiting the growth of a bacterial species on a foodstuff.
  • a foodstuff for purposes of the present invention is any food or beverage that can be ingested.
  • This method is intended for bacteriostatic food packaging or handling and relies on the antimicrobial properties of 5-aminosalicylates.
  • the present method involves treating a food-contacting surface of a material that is pharmaceutically acceptable for food packaging or food handling purposes, with a 5-aminosalicylate compound in an amount effective to inhibit the growth of a bacterial species on a food contacting the surface.
  • Acceptable materials for food packaging or handling include, but are not limited to, paper, wood, cardboard, or other cellulosic polymers, including transparent and non-transparent cellulosic polymers; plastic polymers; waxes; glass or silica; pottery, earthenware, or other ceramic; or metallic materials.
  • a food-contacting surface of a suitable polymeric material is treated with a 5-aminosalicylate compound by enmeshing, implanting, or impregnating the compound within the polymeric material, by means known to the artisan skilled in food packaging and handling materials.
  • a suitable polymeric material is treated with a 5-aminosalicylate compound by covalent linkage, such as by conjugation of the 5-aminosalicylate compound to the polymeric material through, for example, diazo bonds or amide bonds.
  • a polymeric or non-polymeric material is treated with a 5-aminosalicylate compound by coating the material with a coating formulation suitable for a desired coating means, such as, but not limited to, dipping, spraying or brushing onto a surface intended for food contact.
  • a suitable coating formulation contains, in addition to a 5-aminosalicylate compound, an appropriate carrier(s), which carriers are known in the art.
  • an appropriate carrier(s) which carriers are known in the art.
  • the skilled practitioner can employ as a carrier a non- toxic polymeric resin, additionally containing an effective amount of a 5-aminosalicylate compound, which resin can be used to coat the food-contacting surface of the material, hardening in place upon it.
  • An effective amount of a 5-aminosalicylate compound for treating the food-contacting surface of the material is between 0.1 and 10 mg per cm 2 of the surface. Most preferably, the amount of 5- aminosalicylate compound is 0.4 to 2 mg per cm 2 of the surface. This is sufficient concentration to inhibit the growth of a food-poisoning or botulism-causing bacterial species as demonstrated in the detailed examples described herein. While the amount of the 5-aminosalicylate compound on the food- contacting surface of the material is relatively low, it is preferable not to use sulphasalazine as the 5- aminosalicylate compound, due to the possibility of sulfapyridine-related reactions in a minority of food consumers, as described above. Also, it is preferable not to use a 5-aminosalicylate-conjugated bile acid, as this may adversely affect the stability or taste of the food contacting the surface
  • the present invention is also related to food containers and food-handling implements for holding a foodstuff, which includes containing, packaging, covering, storing, displaying, processing, cutting, chopping, impaling, kneading, manipulating or otherwise handling the foodstuff, such that a surface of the food container or implement comes in contact with the foodstuff.
  • the present food containers and food-handling implements comprise a material suitable for contact with food and have a food-contacting surface treated with a 5-aminosalicylate compound, as described above, in an amount effective to inhibit the growth of a bacterial species.
  • the containers and implements are in any suitable disposable (i.e., single-use) or non-disposable (i.e., multi-use) configuration capable of holding a foodstuff. These configurations include, but are not limited to, foils, shear wraps, sheets, papers, waxed papers, bags, cartons, trays, plates, bowls, covered and uncovered storage vessels, serving dishes, cups, cans, jars, bottles, or any other suitable container configuration for a particular foodstuff.
  • Additional configurations especially useful for food handling purposes include, but are not limited to, gloves or mitts; utensils such as forks, spoons, knives, slicers, processors, juicers, grinders, chippers, hooks, presses, screws, openers, cutters, peelers, tongs, ladles, scoops, cups, chutes or spatulas; and cutting boards, kneading boards, mixing bowls, drying or cooling racks, or shelves.
  • utensils such as forks, spoons, knives, slicers, processors, juicers, grinders, chippers, hooks, presses, screws, openers, cutters, peelers, tongs, ladles, scoops, cups, chutes or spatulas
  • cutting boards kneading boards, mixing bowls, drying or cooling racks, or shelves.
  • the present method, containers, and implements are especially useful in inhibiting the growth of bacterial species that can release pathogenic exotoxins, such as, but not limited to, Clostridium perfringens, or Clostridium botulinum, the exotoxins of which cause botulism. They are especially, but not exclusively, useful in commercial and institutional food preparation contexts where food is handled and packaged in bulk, such as, but not limited to, food processing factories, canning plants, slaughterhouses, restaurants, cafeterias, salad bars, grocery outlets, and hospitals. The present method, containers and implements are useful for any kinds of foodstuff.
  • the present invention is also related to antimicrobial cleansers, polishes, paints, sprays, soaps, or detergents formulated for application to surfaces to inhibit the growth of a bacterial species thereon. These surfaces include, but are not limited to surfaces, such as, countertops, desks, chairs, laboratory benches, tables, floors, bed stands, tools or equipment, doorknobs, and windows.
  • the present cleansers, polishes, paints, sprays, soaps, or detergents contain a 5-aminosalicylate compound that provides a bacteriostatic property to them. They can optionally contain suitable solvent(s), carrier(s), thickeners, pigments, fragrances, deodorizers, emulsifiers, surfactants, wetting agents, waxes, or oils.
  • a preferred embodiment is a formulation for external use as a pharmaceutically acceptable skin cleanser, particularly for the surfaces of human hands.
  • the concentration of the 5-aminosalicylate compound is between 0.625 and 200 mg per mL. Most preferably the concentration of 5-aminosalicylate compound is 1.25 to 50 mg per mL.
  • the present cleansers, polishes, paints, sprays, soaps, and detergents are useful in homes and institutions, particularly but not exclusively in hospital settings for the prevention of nosocomial infections.
  • the present invention is also related to an antimicrobial method for inhibiting the growth of a bacterial species in a foodstuff and to foodstuffs containing a 5-aminosalicylate compound.
  • Bacterial growth in foodstuff if uninhibited, can result in the release of bacterial exotoxins that can cause illness or death in a human or non-human vertebrate that consumes the foodstuff.
  • the present method and foodstuffs are particularly useful in preventing clostridial food poisoning by, for example, Clostridium perfringens, or by Clostridium botulinum, the exotoxins of which cause botulism.
  • the present method employs a 5-aminosalicylate compound added to the foodstuff.
  • Any foodstuff can be treated using the present method, but foods for which the present method is especially useful include non-acidic foods, such as mayonnaise or other egg products, potato products, and other vegetable or meat products.
  • Five-aminosalicylates are autoclavable and thus may be used effectively in canned foods.
  • the 5-aminosalicylate compound for adding to the foodstuff can be part of any comestible formulation that can also include a suitable medium or carrier for convenient mixing or dissolving into a particular foodstuff.
  • the medium or carrier is preferably one that will not interfere with the familiar flavor of the food of interest, such as are known by the artisan skilled in food processing techniques.
  • An effective amount of a 5-aminosalicylate compound to be added to a foodstuff is less than that required for administration to a vertebrate subject.
  • the present foodstuffs contain a concentration of a 5-aminosalicylate compound between 0.625 and 10 mg per gram of the foodstuff. Most preferably the concentration of 5-aminosalicylate compound is 1.25 to 5 mg per gram of foodstuff. This is sufficient concentration to inhibit the growth of a food-poisoning or botulism-causing bacterial species as demonstrated in the detailed examples described herein.
  • the concentration of the 5-aminosalicylate compound in the foodstuff is relatively low, it is again, preferable not to use sulphasalazine as the 5-aminosalicylate compound, due to the possibility of sulfapyridine-related reactions in a minority of food consumers, as described above. Also, it is preferable not to use a 5-aminosalicylate-conjugated bile acid, as this may adversely affect the stability or taste of the foodstuff before consumption.
  • the present antimicrobial method for inhibiting bacterial growth in a foodstuff and foodstuffs employing the antimicrobial properties of 5-aminosalicylates are useful alternative means of preventing food poisoning or botulism, instead of nitrite preservatives or gamma irradiation of food, which are disfavored by a sizeable number of consumers.
  • the methods and pharmaceutical compositions of the present invention provide a much needed addition to the antimicrobial armamentarium against bacterial overgrowths and infections, especially clostridial infections that affect great numbers of humans and animals worldwide.
  • Five-aminosalicylate compounds are modestly priced relative to other antibiotics and are conveniently administered.
  • the administration of 5-aminosalicylates is well tolerated by the vast majority of patients with only rare side effects, which, of course, the practitioner should monitor in each individual patient.
  • 5-aminosalicylic acid is known to pose no substantial teratogenic risk in humans and can be administered safely to pregnant women.
  • Bacteroides fragilis (Gram negative anaerobe), Clostridium perfringens (Gram positive obligate anaerobe), Escherichia coli (Gram negative facultative anaerobe), a Lactobacillus isolate (Gram positive aerotolerant anaerobe), and Enterococcus faecalis (Gram negative facultative anaerobe) were examined, because each produces a detectable gaseous fermentation product under certain anaerobic growth conditions.
  • Mesalamine was obtained commercially in the form of Asacol (Procter and Gamble).
  • the enteric coating on 400 mg Asacol tablets was removed with acetone. After the coating was dissolved, intact tablets were air dried and sterilized by autoclaving at 121 °C, 15 psi, for 15 minutes.
  • the sterile tablets were aseptically dissolved in sterile distilled water.
  • coli or Enterococcus faecalis were incubated aerobically for 24 hours, and plates inoculated with C. perfringens, B. fragilis, or Lactobacillus were allowed to incubate anaerobically in GasPak chambers for 48 hours. Colony counts were assessed from each of the four plates of each concentration of mesalamine for each species. Paired t-test was used to compare groups of readings. Four replicate control plates at each concentration of mesalamine were incubated as described above after plating with 1 mL of sterile saline. (All controls were negative for bacterial colonies.)
  • Example 3 Results Resulting colony counts for E. coli and a C. perfringens isolate are tabulated in Table 1. In the concentration range between 12.5 and 100 mg/mL, mesalamine inhibited the growth of the C. perfringens isolate but had no effect on E. coli colony counts. Similar growth inhibition by mesalamine was detected for Clostridium perfringens ATCC 13124 and Clostridium difficile ATCC 9689. (Data not shown.) But mesalamine did not affect the colony numbers of Lactobacillus, Enterococcus, or Bacteroides. O 00/45803

Abstract

A method of inhibiting the growth of a bacterial species in a human or non-human vertebrate employs the antimicrobial (i.e., antibiotic) properties of 5-aminosalicylates. These antimicrobial properties are also employed in an antimicrobial method of inhibiting the growth of a bacterial species in a foodstuff and in foodstuffs containing a 5-aminosalicylate compound. Pharmaceutical compositions, foodstuffs, food containers, food-handling implements, cleansers, polishes, paints, sprays, soaps, or detergents comprise 5-aminosalicylate compounds, such as mesalamine, sulphasalazine, olsalazine, ipsalazine, salicylazobenzoic acid, balsalazide, or conjugated bile acids, including ursodeoxycholic acid-5-aminosalicylic acid. The present pharmaceutical compositions can be formulated for ingestive, colonic, or topical non-systemic delivery systems or for any systemic delivery systems. Formulation can be for human or veterinary administration. Using the method and pharmaceutical preparations the growth of bacterial species, such as Clostridium perfringens, Clostridium difficile, Clostridium botulinum, and Clostridium tetani can be inhibited.

Description

USE OF 5-AMINOSALICYLATES AS ANTIMICROBIAL AGENTS
BACKGROUND OF THE INVENTION
Throughout this application various publications are referenced within parentheses. The disclosures of these publications in their entireties are hereby incorporated by reference in this application in order to more fully describe the state of the art to which this invention pertains. 1. THE FIELD OF THE INVENTION
This invention relates to the medical arts. In particular, it relates to a method of inhibiting bacterial growth in the gastrointestinal tract of a human or non-human vertebrate by using an antimicrobial agent. 2. DISCUSSION OF THE RELATED ART
Antimicrobial or antibiotic agents are used in the treatment of bacterial infections, especially of the gastrointestinal tract. Gastrointestinal infections affect millions of people world-wide, especially children, and pose an increasing health hazard in hospital settings.
Of course, bacteria inhabit healthy intestines to the benefit of their human and animal hosts. Anaerobic bacteria, including the Bacteroides fragilis group and Clostridium species are common member-Spf the intestinal microflora of healthy individuals, and non-toxigenic strains can be transmitted without causing disease symptoms. (B.A. Cunha, Nosocomial diarrhea, Crit. Care Clin. 14(2):329-38 [1998]; H. Kato et al, Application of typing by pulsed-field gel electrophoresis to the study of Clostridium difficile in a neonatal intensive care unit, J. Clin. Microbiol. 32(9):2067-70 [1997]; V.O. Rotimi and B.I. Duerden, Bacteroides species in the normal neonatal faecal flora, J. Hyg. [Lond.] 87(2):299-304 [1981]; B.I. Duerden, The isolation and identification of Bacteroides spp. From the normal human faecal flora, J. Med. Microbiol. 13(l):69-78 [1980]; S.S. Long and R.M. Swenson, Development of anaerobic flora in healthy newborn infants, J. Pediatr. 91(2):298-301 [1977]). Other pathogenic bacterial strains have an adverse effect on their hosts. For example, enterotoxigenic strains of Bacteroides fragilis are associated with diarrhea in humans. (S.K. Niyogi et al., Association of enterotoxigenic Bacteroides fragilis with childhood diarrhoea, Indian J. Med. Res. 105: 167-69 [1997]; R.B. S&c etal, Enterotoxigenic Bacteroides fragilis: epidemiologic studies of its role as a human diarrhoeal pathogen, J. Diarrhoeal Dis. Res. 10(l):4-9 [1992]). And patients with Crohn's disease were reported to have higher numbers of B. fragilis group bacteria in their intestines than healthy controls. (J.G. Ruseler-van Embden and H.C. Both-patoir, Anaerobic gram-negative faecal flora in patients with Crohn 's disease and healthy subjects, Antonie van Leeuwenhoek 49(2): 125-32 [1983]).
More prominent agents of gastrointestinal disease than Bacteroides, are the Clostridium species, especially C. difficile and C. perfringens. Clostridium species are gram-positive, spore-forming anaerobes; some strains that colonize the human intestines can, under certain circumstances, release potent protein exotoxins that induce inflammation of the intestinal mucosa. (M.L. Job and N.F. Jacobs, Jr., Drug-induced Clostridium difficile-associated disease, DmgSaf. 17(1):37-46 [1997]). For example, antibiotics and other chemotherapeutic agents can induce the expression of Toxins A and B from Clostridium difficile. (B.A. Cunha [1998]). Agents known to have a high potential to induce C. difficile- associated disease are aminopenicillins, cephalosporins and clindamycin. (M.L. Job and N.F. Jacobs, Jr., Drug-induced Clostridium difficile-associated disease, Drug. Saf. 17(l):37-46 [1997]; Y. Hutin et al., Prevalence of and risk factors for Clostridium difficile colonization at admission to an infectious diseases ward, Clin. Infect. Dis. 24(5):920-24 [1997]; CD. Settle and M.H. Wilcox [1996]).
In developed countries, the great majority of cases of C. difficile infection are hospital-acquired, and the number of nosocomial clostridial infections is reported to be rising. (CD. Settle and M.H. Wilcox, Review article: antibiotic-induced Clostridium difficile infection, Aliment. Pharmacol. Ther. 10(6):835-41[1996]; J.S. Brazier, The epidemiology and typing of Clostridium difficile, J. Antimicrob. Chemother. 41 Suppl. C47-57 [1998]; S. Tabaqchali and M. Wilks, Epidemiological aspects of infections caused by Bacteroides fragilis and Clostridium difficile, Eur. J. Clin. Microbiol. Infect. Dis. 11 ( 11 ): 1049-57 [1992] ; C.R. Clabots et al. , Acquisition of Clostridium difficile by hospitalized patients: evidence for colonized new admissions as a source of infection, J. Infect. Dis. 166(3):561-67 [1992]).
A nosocomial pleural infection with C. difficile, following surgical insertion of a chest drain has also been reported (A.J. Simpson et al., Nosocomial empyema caused by Clostridium difficile, J. Clin. Pathol. 49(2): 172-73 [1996]), but intestinal infections are the greatest problem.
Nosocomial diarrhea due to gastrointestinal infection with C. difficile has become a major health care problem, causing 20-30% of all nosocomial diarrheas and affecting up to 8% of hospitalized patients. (L.R. Peterson and P.J. Kelly, The role of the clinical microbiology laboratory in the management of Clostridia difficile-associated diarrhea, Infect. Dis. Clin. North Am.7(2):277-93 [1993]). Clostridium difficile is considered to be the premier cause of diarrhea among hospitalized patients. (M. Delmee et al., Treatment of Clostridium difficile colitis. Summary of a round table held in Brussels on March 3, 1994, Acta Clin. Belg. 50(2): 114-116 [1995]).
An infection of C. difficile can add an average of three weeks to a patient' s hospital stay. (CD. Settle and M.H. Wilcox [1996]). Symptoms may include, diarrhea, self-limited colitis, toxic megacolon or potentially lethal fulminant pseudomembranous colitis. Intestinal infection with C. difficile has also been linked to reactive arthritis. (I.H. Kocar et al., Clostridium infection in patients with reactive arthritis of undetermined etiology, Scand. J. Rheumatol. 27(5):357-62 [1998]; R.K. Cleary, Clostridium difficile-associated diarrhea and colitis: clinical manifestations, diagnosis, and treatment, Dis. Colon. Rectum 41(11): 1435-49 [1998]). Bacteraemia and subsequent sepsis is another possible complication of intestinal infection by C. difficile. (P. Naaber et al., Bacterial translocation, intestinal microflora and morphological changes of intestinal mucosa in experimental models of Clostridium difficile infection, J. Med. Microbiol. 47(7):591-98 [1998]; R.J. Feldman et al, Bacteremia due to Clostridium difficile: case report and review of extraintestinal C. Difficile infections, Clin. Infect. Dis. 20(6): 1560-62 [1995]). In at least one nosocomial outbreak, 17 patients died from C. difficile infection. (CD. Settle and M.H. Wilcox [1996]). Clostridium difficile intestinal infections in children, unassociated with antibiotic use or hospital stays, can cause chronic diarrhea and failure to grow. (T.E. Liston, Clostridium difficile toxin associated with chronic diarrhea and failure to gain weight, Clin. Pediatr. (Phila.) 22(6):458-60 [1983]).
In developing countries, C. difficile is also thought to be a causal agent of wide-spread acute diarrheal disease. (S.K. Niyogi et al, Prevalence of Clostridium difficile in hospitalized patients with acute diarrhoea in Calcutta, J. Diarrhoeal Dis. Res. 9(1): 16-19 [1991]; S.Q. Akhtar, Isolation of
Clostridium difficile from diarrhoea patients in Bangladesh, J. Trop. Med. Hyg. 90(4): 189-92 [1987]).
Enterotoxigenic strains of C. perfringens are linked with a significant number of cases of antibiotic-associated diarrhea, especially among elderly hospitalized patients, children, and infants. (A. Wada et al, Nosocomial diarrhoea in the elderly due to enterotoxigenic Clostridium perfringens, Microbiol. Immunol.40(10):767-71 [1996]; M.M. Brett et al, Detection of Clostridium perfringens and its enterotoxin in cases of sporadic diarrhoea, J. Clin. Pathol. 45(7):609-l 1 [1992]; S.C. Samuel et al, An investigation into Clostridium perfringens enterotoxin-associated diarrhoea, J. Hosp. Infect. 18(3):219-30 [1991]; S.P. Boriello et al, Epidemiology of diarrhoea caused by enterotoxigenic Clostridium perfringens, J. Med. Microbiol. 20(3):363-72 [1985]; R. Willliams et al, Diarrhoea due to entertoxigenic Clostridium perfringens: clinical features and management of a cluster of 10 cases, Age Ageing 14(5):296-302 [1985]). Clostridium perfringens has been implicated as a possible contributor to sudden infant death syndrome (SLDS) in susceptible infants. (R.R. Meer et al, Human disease associatedwith Clostridium perfringens enterotoxin, Rev. Environ. Contam. Toxicol. 150:75-94 [1997]).
Clostridium perfringens is well known as a causative agent of non-gastrointestinal gangrene, a special problem for many elderly and diabetic patients with poor blood circulation. But also in more extreme cases of gastrointestinal infection, C. perfringens can cause enteritis necroticans, a gangrene of the bowel resulting in necrosis, sepsis, and hemolysis, in humans and domesticated animals. (L.E. Clarke et al. , Enteritis necroticans with midgut necrosis caused by Clostridium perfringens, Arch. Surg. 129(5):557-60 [1994]; D. Bueschel et al, Enterotoxigenic Clostridium perfringens type A necrotic enteritis in a foal, J. Am. Vet. Med. Assoc. 213(9): 1305-07 [1998]; E.G. Pearson et al, Hemorrhagic enteritis caused by Clostridium perfringens type C in a foal, J. Am. Vet. Med. 188(11): 1309-10 [1986]; F. Al-Sheikhy and R.B. Truscott, The interaction of Clostridium perfringens and its toxins in the production of necrotic enteritis of chickens, Avian Dis. 21(2):256-63 [1977]).
Although rare in developed countries, clostridial enteritis necroticans in humans is more common in some developing countries. (D.A. Watson et al, Pig-bel but no pig: enteritis necroticans acquired in Australia, Med. J. Aust. 155(l):47-50 [1991]). In New Guinea, enteritis necroticans, known locally as pigbel, has been a major cause of illness and death among children. (G.W. Lawrence et al, Impact of active immunisation against enteritis necroticans in Papua New Guinea, Lancet 336(8724): 1165-67 [1990]). Clostridium perfringens type C, the etiologic agent of enteritis necroticans, was also isolated from Bangladeshis with bloody or watery diarrheal illness. (F.P. van Loon et al, Clostridium perfringens type C in bloody and watery diarrhea in Bangladesh, Trop. Geogr. Med. 42(2): 123-27 [1990]).
Entertoxigenic strains of C. perfringens have also been linked to nosocomial and non-nosocomial outbreaks of food poisoning, due to heat-resistant spores and a rapid growth rate in warm food. (A.M. Pollack and P.M. Whitty, Outbreak of Clostridium perfringens food poisoning, J. Hosp. Infect. 17(3): 179-86 [1991]; M. Van Damme-Jongsten et al, Synthetic DNA probes for detection of enterotoxigenic Clostridium perfringens strains isolated from outbreaks of food poisoning, J. Clin. Microbiol. 28(l): 131-33 [1990]). Spores of Clostridium botulinum germinating in warm food can cause another form of food poisoning called botulism. Growing particularly in non-acidic foods lacking nitrites, and protected from oxygen, the vegetative cells of C. botulinum release an exotoxin that when consumed with the food is activated by trypsin in the stomach, and is absorbed intact by the blood stream. The exotoxin binds to nerve cells, preventing the release of the neurotransmitter acetylcholine. Resulting symptoms of botulism include blurred vision, difficulty in swallowing and speaking, and increasing muscular weakness, and usually nausea and vomiting. Death often results from paralysis of the muscles required for breathing. (R.Y. Stanier et al, The Microbial World, 5th ed., Prentice Hall, Englewood Cliffs, NJ, pp.626-27 [1986]). Clostridium botulinum sometimes colonizes the intestines of infants and can cause infantile botulism, which can lead to respiratory paralysis and sudden infant death. Botulism is a problem for the food packaging industry. Spores of C. botulinum may not be killed if canning is done at too low a temperature. High temperature autoclave treatment may be unsuitable for some foods Mayonnaise and other non-acidic foods are particularly prone to foster the growth of C. botulinum. Now with increasing health concerns about the use of nitrite as a food preservative, alternative antimicrobial agents are needed against the growth of C. botulinum and other food poisoning bacterial pathogens.
Antimicrobial agents with selective toxicity for a specific spectrum or range of pathogenic microorganisms are well known in the art. One class of antimicrobial agents is the antibiotics, which are compounds, synthesized and excreted by various microorganisms, that are selectively toxic to other microorganisms, specifically bacteria. In addition, some antibiotics can be artificially modified to produce antimicrobial agents that are more effective and/or more able to overcome antibiotic resistance.
The first line antibiotic treatment for diseases associated with gastrointestinal infections of
Clostridium has been a 10-day course of metronidazole or vancomycin, which may be administered orally, intravenously, or rectally. (R.K. Cleary [1998]; C.P. Kelly and J.T. LaMont, Clostridium difficile infection, Annu. Rev. Med. 49:375-90 [1998]; CM. Reinke and C.R. Messick, Update on Clostridium difficile-induced colitis, Part 2, Am. J. Hosp. Pharm. 51(15): 1892-1901 [1994]).
Neither of these antibiotics has been completely satisfactory. While metronidazole (flagyl) is effective against obligate anaerobes (e.g., P. Muir et al. , Breath hydrogen excretion by healthy cats after oral administration of oxytetracycline and metronidazole, Vet. Rec. 138:635-39 [1996]; B. Lembcke et al. Different actions of neomycin and metronidazole on breath hydrogen (H2) exhalation, Z. Gastroenterol. 18(3): 155-60 [1980]), it yields an unpleasant after-taste to many patients, even when delivered intravenously. Other common side effects of metronidazole are neuropathy and gastrointestinal distress. Also, metronidazole is a known reproductive toxicant affecting mammalian sperm cells. (R.E. Linder et al, Endpoints of spermatotoxicity in the rat after short duration exposures to fourteen reproductive toxicants, Reprod. Toxicol. 6(6):491-505 [1992]).
On the other hand, a course of vancomycin is prohibitively expensive (10-50 times more expensive than metronidazole), and there are concerns about the rapid development of vancomycin- resistance among pathogenic Clostridium, Enterococcus, Pediococcus, Citrobacter, Klebsiella, Enterobacter, and Staphylococcus species, because the plasmid-borne vancomycin resistance gene (VanR) is readily transmissible. (ASHP therapeutic position statement on the preferential use of metronidazole for the treatment of Clostridium difficile-associated disease, Am. J. Health Syst. Pharm. 55(13): 1407-1 [1998]; S.H. Cohen et al, Isolation of a toxin B-deficient mutant strain of Clostridium difficile in a case of recurrent C. difficile-associated diarrhea, Clin. Infect. Dis. 26(2): 1250 [1998]; C. Edlund et al. , Effect ofvancomycin on intestinal flora of patients who previously received antimicrobial therapy, Clin. Infect. Dis. 25(3):729-32 [1997]; CA. O'Donovan et al, Enteric eradication of vancomycin-resistant Enterococcus faecium with oral bacitracin, Diagn. Microbiol. Infect. Dis. 18(2): 105-09 [1994]; E. Yamaguchi et al, Colonization pattern of vancomycin-resistant Enterococcus faecium, Am. J. Infect. Control 22(4):202-06 [1994]; C.P. Kelly and J.T. LaMont [1998]).
In addition, relapses of clostridial infections occur in about 5-42% of those treated with metronidazole or vancomycin and are believed to be caused by persistent endogenous clostridial spores, which are antibiotic resistant. (S.H. Cohen et al. [1998] ; R. Fekety et al. , Recurrent Clostridium difficile diarrhea: characteristics of and risk factors for patients enrolled in a prospective, randomized, double- blinded trial, Clin. Infect. Dis. 24(3):324-33 [1997]; S. Johnson et al., Treatment of asymptomatic Clostridium difficile carriers [fecal excretors] with vancomycin and metronidazole. A randomized, placebo-controlled trial, Ann. Intern. Med. 117(4):297-302; M.J. Zimmerman et al, Review article: treatment of Clostridium difficile infection, Aliment. Pharmacol. Ther. 11(6): 1003-12 [1997]).
Teicoplanin is another antibiotic found to be effective against gram positive anaerobes such as Propionibacterium acnes, Clostridium perfringens, C. difficile, and other Clostridium spp., Peptococcus spp., Peptostreptococcus spp. (H. Hassan et al, In vitro activity of teicoplanin, vancomycin, A16686, clindamycin, erythromycin andfusidic acid against anaerobic bacteria, Singapore Med. J. 3 l(l):56-58 [1990]; The Swedish CD AD Study Group, Treatment of Clostridium difficile associated diarrhea and colitis with an oral preparation of teicoplanin; a dose finding study, Scand. J. Infect. Dis. 26(3):309-16 [1994]; C Wenisch et al, Comparison ofvancomycin, teicoplanin, metronidazole, andfusidic acid for the treatment of Clostridium difficile-associated diarrhea, Clin. Infect. Dis. 22(5):813-18 [1996]).
However, teicoplanin is not widely available. The peptide antibiotic bacitracin is also reported to be effective in treating C. difficile-induced diarrhea, but it is not widely available in an enteric formulation. (M.N. Dudley et al. , Oral bacitracin vs. vancomycin therapy for Clostridium difficile- induced diarrhea, A randomized double-blind trial, Arch. Intern. Med. 146(6): 1101-04 [1986]).
Other adjunct treatments are reportedly effective for refractory C. difficile-related disease, including whole-bowel irrigation and enteric administration of the non-pathogenic yeast Sαcchαromyces boulαrdii. (CA. Liacouras and D.F.A. Piccoli, Whole-bowel irrigation as an adjunct to the treatment of chronic, relapsing Clostridium difficile colitis, J. Clin. Gastroenterol. 22(3): 186-89 [1996]; CM. Surawicz, Clostridium difficile disease: diagnosis and treatment, Gastroenterologist 6(1 ):60-65 [1998]). But such treatments are uncomfortable or distasteful for many patients and are less suitable than easily administered antibiotics as a first line treatment regimen.
Accordingly, there remains a definite need for a modestly priced antimicrobial agent for treating gastrointestinal infections, without the commonly unpleasant side effects and bacterial resistance associated with metronidazole and vancomycin.
Pharmaceutical preparations of 4-(p)-aminosalicylic acid (i.e., 4-ASA or para-aminosalicylic acid) or 4-(p)-aminosalicylate sodium salt (e.g., Nemasol-Sodium® or Tubasal®) have been used systemically in cases of tuberculosis as antimicrobial chemotherapeutic agents against Mycobacterium tuberculosis.
On the other hand, the 5-aminosalicylates are known as anti-inflammatory chemotherapeutic agents and have not been used as antimicrobial agents. These compounds include 5-aminosalicylic acid (i.e., 5-ASA, mesalamine, or mesalazine) and conjugated derivatives thereof, known for their anti- inflammatory properties. These anti-inflammatory agents are commercially available in various pharmaceutical preparations such as Asacol®, Rowasa®, Claversal®, Pentasa®, Salofalk®, Dipentum®, Azulfidine® (SAZ) and others.
5-Aminosalicylates have been used widely to reduce mucosal inflammation in inflammatory bowel disease, ulcerative colitis and Crohn' s disease. (S.B. Hanauer and FB. Baert, Medical Therapy of Inflammatory Bowel Disease, Inflamm. Bowel Dis. 78(6): 1413-25 [1994]; C.J. Mulder et al, Drug therapy: dose-response relationship of oral mesalazine in inflammatory bowel disease, Mediators Inflamm. 7(3): 135-36 [1998]; W. Kruis et al, Olsalazine versus mesalazine in the treatment of mild to moderate ulcerative colitis, Aliment. Pharmacol.12(8):707-15 [1998]; J.N. Healey, Gastrointestinal transit and release of mesalazine tablets in patients with inflammatory bowel disease, Scand. J. Gastroenterol. 172:47-51 [1990]).
The mechanism underlying the anti-inflammatory properties of the 5-aminosalicylates is unknown, but it may result from their ability to inhibit oxidation at the surface of endothelial membranes, perhaps through radical scavenging, and to prevent lipid peroxidation. (D.C Pearson et al, The anti- oxidant properties of 5-aminosalicylic acid, Free Radic. Biol. Med. 21(3):367-73 [1996]). 5- Aminosalicylates may be able to act synergistically with endogenous antioxidants such as alpha- tocopherol to prevent the oxidative damage implicated in the pathogenesis of inflammatory bowel diseases. (E. Goncalves et al, Antioxidant activity of 5-aminosalicylic acid against peroxidation of phosphotidylcholine liposomes in the presence of alpha-tocopherol: a synergistic effect?, Free Radic. Res. 29(l):53-66 [1998]).
There are several reports that 5-aminosalicylic acid also inhibits fimbriae-mediated cellular adhesion by enteroaggregative Escherischia coli strains, associated with both acute and persistent diarrhea in infants and children . (G. Kang et al. , Salicylate inhibits fimbriae mediated Hep-2 cell adherence of and haemagglutination by enteroaggregative Escherischia coli, FEMS Microbiol. Lett. 166(2):257-65 [1998]; D. Law and H. Chart, Enteroaggregative Escherischia coli, J. Appl. Microbiol. 84(5):685-97 [1998] ; Y. Germani et al. , Prevalence ofenteropathogenic, enteroaggregative Escherischia coli among isolates from children with diarrhea in New Caledonia, J . Infect. Dis. 174(5): 124-26 [1996]; S. Knutton et al., Ability of enteroaggregative Escherischia coli strains to adhere in vitro to human intestinal mucosa, Infect. Immun. 60(5):2083-91 [1992]).
Antimicrobial growth inhibitory properties of the 5-aminosalicylates and other features and advantages of the present invention will be described herein.
SUMMARY OF THE INVENTION The method of the present invention employs the antimicrobial properties of 5-aminosalicylates in a method of inhibiting bacterial growth in a human or non-human vertebrate. By administering to a patient a pharmaceutical composition comprising a 5-aminosalicylate, the growth of bacterial species can be arrested, including the growth of anaerobic pathogens of the genus Clostridium, for example, C. perfringens, C. difficile, C. botulinum and C. tetani. The present method can be used to treat the gastrointestinal tract or any other non-gastrointestinal body site or tissue to inhibit bacterial growth.
The present method also has veterinary applications and can be used to treat a wide variety of non-human vertebrates, including wild, domestic and farm animals. The present invention is also related to an antimicrobial method for inhibiting the growth of a bacterial species in a foodstuff and to foodstuffs containing a 5-aminosalicylate compound, useful for preventing food poisoning or botulism. The present invention is also related to food containers and food- handling implements with bacteriostatic properties, intended for holding a foodstuff, and to cleansers, polishes, paints, sprays, soaps, and detergents for inhibiting the growth of a bacterial species on surfaces. The 5-aminosalicylates are generally well-tolerated and competitively priced compounds that can fill a definite need for an antimicrobial agent, which is selectively effective against gastrointestinal and non-gastrointestinal clostridial diseases.
These and other advantages and features of the present invention will be described more fully in a detailed description of the preferred embodiments which follows.
DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS
The present invention is directed to the use of 5-aminosalicylates as antimicrobial agents in a method of inhibiting bacterial growth in a human or non-human vertebrate, in the gastrointestinal tract or any other body site or tissue. The method involves administering to a human or non-human vertebrate subject a pharmaceutically acceptable composition of the present invention.
The pharmaceutically acceptable compositions in accordance with the present invention are formulated for acceptable delivery to a human or non-human vertebrate gastrointestinal tract or other body site or tissue. Topical delivery to gastrointestinal mucosa and/or systemic delivery thereto are intended. Also included are pharmaceutically acceptable formulations for systemic or topical delivery to a non-gastrointestinal body site or tissue subject to bacterial growth, and particularly subject to clostridial growth. The present invention is also related to pharmaceutically acceptable compositions containing a 5-aminosalicylate compound.
Five-aminosalicylate compounds include 5-aminosalicylic acid or any compound containing a 5-aminosalicylate moiety, or any conjugated derivative thereof, that is effective in inhibiting the growth of a bacterial species in a human or non-human vertebrate gastrointestinal tract or any other body site, limb, organ, such as heart, lung or kidney, or any other tissue of a vertebrate body.
A most preferred 5-aminosalicylate compound is 5-aminosalicylic acid, also known as mesalamine or mesalazine, and commercially available, for example, in pharmaceutical preparations known as Asacol® (Procter and Gamble), Pentasa® (Ferring A/S Vanlose), Claversal®, Mesasal®, Rowasa®, and Salofalk® (Dr. Falk GmbH). (R.N. Brogden and E.M. Sorkin, Mesalazine. A review of its pharmacodynamic andpharmacokinetic properties, and therapeutic potential in chronic inflammatory bowel disease, Drugs 38(4):500-23 [1989]).
Also preferred are 5-aminosalicylate salts, including the sodium or potassium salts of conjugated 5-aminosalicylates.
Other preferred 5-aminosalicylate compounds, for purposes of the present invention, are compounds containing a 5-aminosalicylic acid moiety, or a 5-aminosalicylate moiety, conjugated to another 5-aminosalicylic acid moiety or 5-aminosalicylate moiety, or conjugated to a pharmaceutically acceptable carrier molecule.
Preferred conjugated 5-aminosalicylate compounds include, but are not limited to, olsalazine or Poly-ASA. Olsalazine is known variously as disodium-diazosalicylate, azodisalicylate, or Dipentum® (Pharmacia). Olsalazine comprises two 5-aminosalicylate moieties linked through a diazobond. Poly- ASA comprises a water-soluble polymer that contains 5-aminosalicylate moieties linked through diazobonds to an inert sulfanilamide ethylene polymer. (F. Martin, Oral 5-aminosalicylic acid preparations in treatment of inflammatory bowel disease: an update, Digestive Dis. Sci. 32(12 Suppl.):57S-63S [1987]; H. Allgayer, Sulfasalazine and 5-ASA compounds, Gastrointestinal Pharmacol. 21(3):643-658 [1992]).
Balsalazide (balsalazine) is another preferred conjugated 5-aminosalicylate compound. (J.R. Green et al. , Balsalazide is more effective and better tolerated than mesalamine in the treatment of acute ulcerative colitis. The Abacus Investigator Group, Gastroenterol. 114(1): 15-22 [1998a]; J.R. Green et al, Maintenance and remission of ulcerative colitis: a comparison between balsalazide 3 g and mesalamine 1.2 g daily over 12 months. Abacus Investigator Group, Aliment. Pharmacol. Ther. 12(12): 1207-16 [1998b]). Another preferred conjugated 5-aminosalicylate compound is sulphasalazine, also known as sulfasalazine, salazosulfapyridine, or Azulfidine®, in which a 5-aminosalicylic acid moiety is linked through a diazobond to sulfapyridine, the carrier molecule. (H.P. Osterwald, Pharmaceutic development: mesalazine, Scand. J. Gastroenterol Suppl. 172:43-46 [1990]). The skilled practitioner will be aware that patients to whom sulphasalazine is administered should be monitored especially carefully for symptoms of drug intolerance or hypersensitivity reactions that can occur in some patients. These symptoms are related to the sulfapyridine moiety and may include headache, nausea, vomiting, fever, rash, epidermolysis, hemolytic anemia, pancreatitis, pulmonary fibrosis, agranulocytosis, or liver toxicity; the use of sulphasalazine should be discontinued immediately when these symptoms occur. (S.B. Hanauer and G. Stathopoulos, Risk-benefit assessment of drugs used in the treatment of inflammatory bowel disease, Drug Saf. 6(3): 192-219 [1992]; H. Allgayer [1992]). Impairment of male fertility is another possible side effect of sulphasalazine related to sulfapyridine. V.A. Botoman and G.F. Bonner, Management of inflammatory bowel disease, Am. Fam. Physician 57(l):57-68 [1998]).
Other preferred conjugated 5-aminosalicylate compounds are ipsalazine (ipsalazide) and salicylazobenzoic acid; these are also well tolerated molecules comprising a 5-aminosalicylic acid moiety or 5-aminosalicylate moiety linked by a diazobond to another pharmaceutically acceptable chemical unit.
(M.C. Rijk et al, Disposition of mesalazine-delivering drugs in patients with inflammatory bowel disease, with and without diarrhoea, Scand. J. Gastroenterol. 27(10):863-68 [1992]; S.N. Rasmussen,
Bioavailability of controlled release mesalazine [5-ASA] preparations, J. Gastroenterol. 30(Suppl.
8): 112-114 [1995]). Other preferred conjugated 5-aminosalicylate compounds for gastrointestinal use are bile acids conjugated with a 5-aminosalicylic acid moiety or 5-aminosalicylate moiety. For example, in ursodeoxycholic acid-5-aminosalicylic acid (UDCA-5-ASA), ursodeoxycholic acid is linked through an amide bond to 5-aminosalicylic acid. The conjugated 5-aminosalicylate compound is synthesized by adding a basic solution of 5-aminosalicylic acid into the mixed anhydride of ursodeoxycholic acid and ethyl chloroformate. (A.K. Batta et al, Synthesis and intestinal metabolism of ursodeoxycholic acid conjugate with an antiinflammatory agent, 5-aminosalicylic acid, J. Lipid Res. 39(8);1641-46 [1998]). Five-aminosalicylate bile acid conjugates are not substantially absorbed in the duodenum but can be partially hydrolyzed in the colon by intestinal bacteria, such as Clostridium perfringens, to ursodeoxycholic acid and 5-aminosalicylic acid. Id. As the skilled artisan is aware, bacterial azoreductases can reduce diazo bonds linking 5- aminosalicylic acid or 5-aminosalicylate moieties to another chemical moiety, thus releasing 5- aminosalicylic acid or 5-aminosalicylate. Similarly, certain hydrolases, e.g., cholylglycine hydrolase, can deconjugate 5-aminosalicylic acid moieties or 5-aminosalicylate moieties from other carriers, such as bile acids. Other particular varieties of 5-aminosalicylate conjugate can be subject to deconjugation by other enzyme systems. Whether or not a 5-aminosalicylic acid or 5-aminosalicylate moiety is actually deconjugated from a carrier molecule or from another 5-aminosalicylate moiety, in a human or non-human vertebrate gastrointestinal tract or other body site, does not limit the embodiments of a 5- aminosalicylate compound contemplated by the present invention. The present invention is not committed to any particular mechanism by which a particular 5-aminosalicylate compound operates to inhibit bacterial growth.
The 5-aminosalicylate compound is administered by any suitable method. Representative methods include giving, providing, feeding or force-feeding, dispensing, inserting, prescribing, furnishing, treating with, taking, swallowing, eating or applying a pharmaceutical composition of the present invention.
As well as a 5-aminosalicylate compound, the pharmaceutical compositions of the present invention can optionally contain pharmaceutically acceptable solvent(s), adjuvant(s) or non-medicinal carrier(s), binder(s), thickener(s), or filler substance(s) that are known to the skilled artisan. Common fillers include, but are not limited to, sucrose or lactose, or polymeric substances like starch. Also contemplated are additional medicinal or nutritive additives in combination with a 5-aminosalicylate compound, as may be desired to suit the more particular needs of the practitioner.
A dose effective to inhibit the growth of a bacterial species is such dose as sufficient to prevent cellular proliferation of a bacterial species, by either killing bacterial cells or by preventing or slowing cellular growth or reproduction of a bacterial species, compared to the rate of growth or reproduction in the absence of a 5-aminosalicylate compound. By way of example, 12.5 mg or more of 5-aminosalicylic acid have been found sufficient to inhibit the growth of Clostridium perfringens on 10-mL blood agar plates. The effective dose for each human or non-human vertebrate subject will depend upon the size and physiologic reactions of the subject to whom or to which the pharmaceutical preparations of the present invention are administered. And these reactions and the antimicrobial activity of the administered dose can be monitored by the prescribing physician or veterinarian. The pharmaceutical compositions of the present invention can be formulated and manufactured at more than one concentration, such that modular incremental amounts of 5-aminosalicylate compound are easily administered.
For example, currently available preparations of 5-aminosalicylate compounds can be obtained in variable dosage units, including, for example: 250 mg (e.g., Salofalk®, Pentasa®, Azulfadine®, Dipentum®, or olsalazine); 400 mg (e.g., Asacol®); 0.5, 1.5 g or 3.0 g (e.g., Pentasa®, Rowasa®, Azulfadine®, balsalazide). Suppositories containing 5-aminosalicylic acid in dosage units ranging from 0.2 to 1.0 grams are also available; enema solutions containing 1 to 4 g of 5-aminosalicylic acid are also known. (M. A. Peppercorn, Advances in drug therapy for inflammatory bowel disease, Ann. Intern. Med. 112:50-60 [1990]).
The foregoing are merely illustrative of the possible dosage units that can be employed in the pharmaceutical compositions of the present invention, and smaller or larger dosage units than these are also contemplated. Larger dosage units are especially useful for large non-human vertebrates, such as, but not limited to, bovine animals, horses, pachyderms, or large marine mammals; smaller dosage units are especially useful for pediatric application and for small vertebrates, such as, but not limited to, mice or chickens.
A minimum effective dose is as little as between 6.25 and 12.5 mg of a 5-aminosalicylate compound per day. Effective doses of a 5-aminosalicylate compound at 12.5 to 150 mg per day and 150 to 250 mg per day are sufficient for smaller human adults, children, and smaller non-human vertebrates, such as rodents, canines, chickens and turkeys. A higher effective dose for an adult human is 250-6400 mg of a 5-aminosalicylate per day. Pediatric doses are typically 10-20% of effective doses for adult humans. Higher effective doses, from 6,450 mg/day, up to 1,000 mg/kg body mass per day can be used for large non-human vertebrates, for example, for sheep and larger animals such as cattle, horses, and elephants.
Antimicrobial activity by 5-aminosalicylate compounds against a specific bacterial species of interest is determined by routine means well known to the skilled practitioner. For example, a "lawn" of a bacterial species can be plated on an appropriate solid medium, and zones of growth inhibition around sterile cellulose disks impregnated with a 5-aminosalicylate compound of interest can be measured. Similarly, inhibition assays in liquid media are also routinely accomplished.
Inhibition of bacterial growth in a gastrointestinal tract is measured by conventional means well known to the skilled artisan. Many fermentative bacterial species found in the gastrointestinal tract produce detectable quantities of hydrogen or methane gas in the presence of certain sugars, which gases enter the blood stream of the host and are exhaled. This is the basis for intestinal bacterial growth detection means, such as, but not limited to, the lactulose, glucose, or lactose breath hydrogen tests. (E.g., P. Kerlin and L. Wong, Breath hydrogen testing in bacterial overgrowth of the small intestine, Gastroenterol. 95(4):982-88 [1988]; A. Strocchi et al, Detection of malabsorption of low doses of carbohydrate: accuracy of various breath H2 criteria, Gastroenterol. 105(5): 1404-1410 [1993]).
Alternatively, bacterial growth in a gastrointestinal tract is measured by detection of 13CO2 or 14C02 breath emissions after administering an isotope-labeled sugar that is metabolizable by gastrointestinal bacteria but non-digestible by the host, such as, but not limited to, xylose or lactulose in humans. (E.g., G.R. Swart and J.W. van den Berg, I3C breath test in gastrointestinal practice, Scand. J. Gastroenterol. [Suppl.] 225: 13-18 [1998]; C.E. King and P.P. Toskes, Breath tests in the diagnosis of small intestinal bacterial overgrowth, Crit. Rev. Lab. Sci. 21(3):269-81 [1984]; C.S. Chang et al, Increased accuracy of the carbon-14 D-xylose breath test in detecting small-intestinal bacterial overgrowth by correction with the gastric emptying rate, Eur. J. Nucl. Med. 22(10): 1118-22 [1995]; A. Schneider et al, Value of the l4C-D-xylose breath test in patients with intestinal bacterial overgrowth, Digestion 32(2):86-91 [1985]).
Direct gastrointestinal sampling or biopsy from any body site or tissue can also be used to measure the inhibition of bacterial growth in a gastrointestinal tract or other body site or tissue. As the skilled artisan is aware, direct sampling at time intervals provides information about the growth inhibition of specific bacterial species of interest, to which breath testing is not well-suited. Samples are diluted and bacterial numbers can be assessed by conventional microbiological means such as, but not limited to colony plating or Most Probable Number (MPN) techniques, or direct counting of bacterial cells. For direct bacterial cell counts, cells can optionally be labeled with specific markers, and counts can be accomplished manually or by devices such as fluorescence activated cell sorting (FACS). Alternatively, evidence of inhibition of bacterial growth can be inferred by the practitioner treating a bacterial infection or intestinal bacterial overgrowth in a human or non-human vertebrate subject with observation of an improvement in various infection-or overgrowth-related symptoms in response to the administration of an antimicrobial composition of the present invention.
Among the bacterial species inhibited in accordance with the present inventive method are obligate anaerobes such as, but not limited to, Clostridium species. It is a particular advantage of the present invention that 5-aminosalicylic acid is an antimicrobial agent that does not affect many beneficial or commensal gastrointestinal bacteria but selectively inhibits potentially pathogenic clostridial species, such as, but not limited to, C. perfringens, C. difficile, C. tetani and C. botulinum.
The method of the present invention is also useful for veterinary purposes. An antimicrobial composition comprising a 5-aminosalicylate compound can be administered to a non-human vertebrate including, but not limited to, a wild, domestic or farm animal. The present method is useful for treating a mammal such as a non-human primate, mouse, rat, rabbit, gerbil, hamster, canine, feline, ovine, bovine, swine, pachyderm, equine, or marine mammal. Also, the method is useful to inhibit the growth of bacteria in a bird (avian) or poultry, such as a duck, chicken, goose, turkey, ostrich, emu, dove, pigeon, quail, pheasant, peafowl, or guinea fowl.
In one embodiment, the pharmaceutically acceptable composition is administered by a non- systemic delivery route to the site of bacterial infection or overgrowth that is not primarily by way of the blood stream of a human or non-human vertebrate.
Some non-systemic delivery routes and pharmaceutically acceptable non-systemic delivery systems that could be employed by one of skill in the art in practicing the methods and compositions of the present invention are now exemplified. The following are presented merely to illustrate and in no way to limit the possible delivery means contemplated.
For gastrointestinal bacterial infection or bacterial overgrowth, suitable non-systemic delivery routes include, but are not limited to, an ingestive delivery route or a colonic delivery route. A most preferred delivery route is an ingestive delivery route, whereby the antimicrobial agent enters the gastrointestinal or digestive tract by way of voluntary or forced ingestion through the mouth. The organs of a gastrointestinal tract include the esophagus, stomach, large intestine, small intestine, or rectum. The skilled artisan will be aware that in a non-human vertebrate the digestive tract may include a rumen, crop, gullet, cecum, or other specialized organ as pertains to a particular vertebrate species.
Another non-systemic delivery route is useful for non-gastrointestinal bacterial infections, particularly infections of the skin or externally accessible wounds; this delivery route is topical application to the affected area of an antimicrobial cream, gel, or ointment.
Another preferred embodiment of the pharmaceutical compositions of the present invention, employing a non-systemic delivery route, is a suppository or foam for delivery of a composition comprising a 5-aminosalicylate compound via anus or rectum to the colon. Once delivered, a 5- aminosalicylate compound of the present invention will act topically at the intestinal mucosa. Such suppository or foam delivery systems are known in the art; a commercially available example is Rowasa® mesalamine suppositories for anti-inflammatory purposes. Together with a 5-aminosalicylate compound the pharmaceutical preparation of the present invention, such as a suppository, can employ a variety of conventional thickeners, such as alginate, xanthan gum, or petrolatum. Also contemplated are suppositories or foams comprising hydrophilic polymers, such as hydroxypropyl cellulose, hydroxypropyl methylcellulose, hydroxyethylcellulose, dextran, pectin, poly vinyl pyrrolidone, starch, gelatin, or any of a number of other polymers known to be useful for this purpose.
Another preferred embodiment of the compositions of the present invention is a gel for non- systemic delivery of a composition comprising a 5-aminosalicylate compound via the colon, similar to gels commonly used for the delivery of other chemotherapeutic agents. Hydrogel matrices are known for this purpose. (Feijen, Biodegradable hydrogel matrices for the controlled release of pharmacologically active agents, U.S. Pat. No. 4,925,677). Such biodegradable gel matrices may be formed, for example, by cross-linking a proteinaceous component and a polysaccharide or mucopolysaccharide component, then loading with a 5-aminosalicylate compound, for deli verability over an extended period. Another preferred embodiment of the present pharmaceutical composition formulated for a colonic non-systemic delivery system is a composition comprising a 5-aminosalicylate compound in an osmotically suitable enema solution. Commercially available preparations include Rowasa® mesalamine enemas for anti-inflammatory purposes. (See, R.N. Brogden and E.M. Sorkin, Mesalazine. A review of its pharmacodynamic andpharmacokinetic properties, and therapeutic potential in chronic inflammatory bowel disease, Drugs 38(4):500-24 [1989]).
A most preferred embodiment of the pharmaceutical composition of the present invention is formulated for a non-systemic ingestive delivery system, such as, but not limited to a tablet, capsule, or caplet.
Preferably, the non-systemic ingestive delivery system comprises an enteric coating to prevent esophageal or gastric release of 5-aminosalicylate compound. Such enteric-coated pharmaceuticals disintegrate after leaving the stomach, resulting in drug dispersion in the small intestine or colon where 5-aminosalicylates will act topically at the intestinal mucosa. As the skilled artisan will be aware, enteric-coated drug delivery systems are typically pH-sensitive, polymer-coated tablets, capsules, or caplets. A polymer coating can be selected that will direct release of a 5-aminosalicylate compound to a particular region of the gut. Such polymers include, but are not limited acrylic polymers such as Eudragit-L or Eudragit-S, and cellulosic polymers, such as ethylcellulose. Commercially available examples of enteric-coated 5-aminosalicylates are mesalamine preparations known as Asacol® and Claversal®, used as anti-inflammatory agents. The acrylic polymer coating of Claversal®, starts to dissolve at pH 6.0 after passing through the far more acidic milieu of the stomach, and as a result, mesalamine is reliably released in the distal small intestine (ileum) and colon of a human patient. (H.P. Osterwald, Pharmaceutic development: mesalazine, Scand. J. Gastroenterol. Suppl. 172:43-46 [1990]; D. Claussen, Asacol [mesalamine], Gastroenterol. Nurs. 15(l):33-34 [1992]). The acrylic polymer coating (i.e., Eudragit-S) of Asacol® is degraded at above pH 7.0. Thus, it is carried to the right side of the human colon where intraluminal pH is elevated above 7.0. (M.J. Dew et al, An oral preparation to release drug in the human colon, Br. J. Clin. Pharmacol. 14:405-08 [1982]). Salofalk® is another example; it is coated first with a semipermeable layer of ethylcellulose and second with an acrylic polymer (i.e., Eudragit-L), which is degraded at above pH 5.6 in the distal small intestine and colon to release mesalamine there. (F. Martin [1987]). Pentasa® contains 5-aminosalicylic acid microgranules coated with a semipermeable membrane of amphionic ethylcellulose, which can dissolve at either acidic or basic pH to release the microgranules. (F. Martin [1987]).
In addition, such tablets, capsules, or caplets can be formulated in a multi-layer configuration for slow release over an extended period. Again, Pentasa® is an example of a commercially available ingestible slow release form of a 5-aminosalicylate compound. (See P. Fockens et al, Comparison of the efficacy and safety of 1.5 compared with 3.0 g oral slow release mesalazine [Pentasa] in the maintenance treatment of ulcerative colitis. Dutch Pentasa Study Group, Eur. J. Gastroenterol. Hepatol. 7(11): 1025-30 [1995]). Another preferred ingestive delivery system comprises a lavage system, whereby a patient will ingest a large volume of an osmotically balanced flushing solution, containing 5-aminosalicylic acid, or a conventional flushing solution in conjunction with another ingestible form of a 5-aminosalicylate antimicrobial agent. Such a lavage system can virtually eliminate bacterial populations from the intestine. This may be especially desirable in refractory cases of bacterial overgrowth or in preparing a patient for abdominal surgery.
Another preferred ingestive delivery system is especially, but not exclusively, useful for veterinary applications. In this embodiment a pharmaceutical preparation of the present invention is formulated and prepared to be ingested by an animal along with its food, as part of a pharmaceutically acceptable feed mixture. A pharmaceutically acceptable food additive for humans is also contemplated. For some applications, a preferred embodiment of the antimicrobial method of the present invention involves a systemic delivery route, i.e., a route whereby delivery of a 5-aminosalicylate compound to the site of infection or bacterial growth is primarily via the blood stream. This embodiment can be used to inhibit bacterial growth in any body site or tissue, including the gastrointestinal tract. A systemic delivery route is also particularly, but not exclusively useful for gastrointestinally infected patients who are unable to effectively ingest a non-systemic formulation of the composition of the present invention due to persistent nausea, difficulty in swallowing, or other ingestion-preventing conditions, or who, due to resection or other condition of the bowel cannot accept a colonic delivery system.
Alternatively, a systemic delivery route can be employed to deliver a 5-aminosalicylate compound to body sites or tissues other than those of the gastrointestinal tract, including, but not limited to, skin, heart, lung, blood, kidney, liver, brain, arms, legs, digits, sexual organs, trunk, head, neck, or tail. Applications can include but are not limited to treating or preventing clostridial infections at any body site or tissue of a vertebrate. Such clostridial infections include, but are not limited to, gangrene or tetanus, caused, respectively, by C. perfringens and C. tetani, when these species grow in wounds and damaged tissues with low oxygen tension.
Entry of a 5-aminosalicylate compound into the blood stream of a human or non-human vertebrate patient can occur by any route, system, device, method or mechanism. For the purposes of the present invention, a systemic delivery route can also include delivery through the skin, mucosa or epithelium of the mouth including the sublingual epithelium, the rectum, or the vaginal epithelium.
Systemic delivery systems that are contemplated by the present invention include, but are not limited to, ingestion, injection, or intravenous drip, most conventionally. Other useful systemic delivery systems are known and include, but are not limited to, implant; adhesive transdermal patches; topical creams, gels or ointments for transdermal delivery; transmucosal delivery matrices or suppositories or gels. It is contemplated that the compositions of the present invention are formulated to deliver an effective dose of a 5-aminosalicylate compound by these or any other pharmaceutically acceptable systemic delivery system. A preferred embodiment of the compositions of the present invention employing a systemic delivery route is a topical cream, ointment or gel to be applied to the skin. In this embodiment, a composition of the present invention comprises a 5-aminosalicylate compound in a pharmaceutically acceptable delivery system comprising a permeation or penetration enhancer, such as polyethylene glycol monolaurate, dimethyl sulfoxide, N-vinyl-2-pyrrolidone, N-(2-hydroxyethyl)-pyrrolidone, or 3-hydroxy- N-methyl-2-pyrrolidone. A variety of conventional thickeners often used in creams, ointments and gels, such as, but not limited to, alginate, xanthan gum, or petrolatum, may also be employed in this embodiment of a composition of the present invention.
Another preferred embodiment of the compositions of the present invention is a formulation for systemic transmucosal delivery of a 5-aminosalicylate compound. A variety of pharmaceutically acceptable systems for transmucosal delivery of therapeutic agents are known in the art and are compatible with the practice of the present invention. (Heiber et al, Transmucosal delivery of macromolecular drugs, U.S. Pat. Nos. 5,346,701 and 5,516,523; Longenecker et al, Transmembrane formulations for drug administration, U.S. Pat. No. 4,994,439). Transmucosal delivery devices may be in free form, such as a cream, gel, or ointment, or may comprise a determinate form such as a tablet, patch, or troche. For example, delivery of a 5-aminosalicylate compound may be via a transmucosal delivery system comprising a laminated composite of, for example, an adhesive layer, a backing layer, a permeable membrane defining a reservoir containing a 5-aminosalicylate compound a peel seal disc underlying the membrane, one or more heat seals, and a removable release liner. (Ebert et al,
Transdermal delivery system with adhesive overlay and peel seal disc, U.S. Pat No. 5,662,925; Chang et al, Device for administering an active agent to the skin or mucosa, U.S. Pat. Nos. 4,849,224 and
4,983,395). Alternatively, a tablet or patch for delivery through the oral mucosa can comprise an inner layer containing the therapeutic agent of choice, a permeation enhancer, such as a bile salt or fusidate, and a hydrophilic polymer, such as hydroxypropyl cellulose, hydroxypropyl methylcellulose, hydroxyethylcellulose, dextran, pectin, poly vinyl pyrrolidone, starch, gelatin, or any of a number of other polymers known to be useful for this purpose. This inner layer can have one surface adapted to contact and adhere to the moist mucosal tissue of the oral cavity and may have an opposing surface adhering to an overlying non-adhesive inert layer. Optionally, such a transmucosal delivery system can be in the form of a bilayer tablet, in which the inner layer also contains additional binding agents, flavoring agents, or fillers. Some useful systems employ a non-ionic detergent along with a permeation enhancer. These examples are merely illustrative of available transmucosal delivery technology and are not limiting of the present invention.
Another preferred embodiment of the compositions of the present invention is a gel for systemic delivery of a 5-aminosalicylate via the rectal or vaginal mucosa, similar to gels commonly used for the delivery of various other therapeutic agents. Hydrogel matrices are known for this purpose. (Feijen, Biodegradable hydrogel matrices for the controlled release of pharmacologically active agents, U.S. Pat. No. 4,925,677). Such biodegradable gel matrices can be formed, for example, by cross-linking a proteinaceous component and a polysaccharide or mucopolysaccharide component, then loading with a 5-aminosalicylate compound to be delivered.
Another preferred embodiment of the compositions of the present invention is the systemic delivery of a 5-aminosalicylate compound via a biodegradable matrix implanted within the body or under the skin of a human or non-human vertebrate. The implant matrix may be a hydrogel similar to those described above. Alternatively, it may be formed from a poly-alpha-amino acid component. (Sidman, Biodegradable, implantable drug delivery device, and process for preparing and using same, U.S. Pat. No. 4,351,337).
A preferred embodiment of the composition of the present invention employing a systemic delivery route is a transdermal delivery system of a kind known in the art for delivering various drugs. Transdermal delivery systems can be of any number of varieties known in the art. For example, delivery of a 5-aminosalicylate compound can be via a transdermal delivery system comprising a laminated composite of an adhesive layer, a backing layer, a permeable membrane defining a reservoir containing a 5-aminosalicylate compound, a peel seal disc underlying the membrane, one or more heat seals, and a removable release liner. (Ebert et al , Transdermal delivery system with adhesive overlay and peel seal disc, U.S. Pat No. 5,662,925; Chang et al. Device for administering an active agent to the skin or mucosa, U.S. Pat. Nos. 4,849,224 and 4,983,395).
Alternatively, a transdermal delivery device can be a matrix type transdermal patch. (Chien et al, Transdermal estrogen/progestin dosage unit, system and process, U.S. Pat. Nos. 4,906,169 and 5,023,084; Cleary et al, Diffusion matrix for transdermal drug administration and transdermal drug delivery devices including same, U.S. Pat. No.4,911,916; Teillaud et al, EVA-based transdermal matrix system for the administration of an estrogen and/or a progestogen, U.S. Pat. No. 5.605,702; Venkateshwaran et al, Transdermal drug delivery matrix for coadministering estradiol and another steroid, U.S. Pat. No.5,783,208; Ebert etal, Methods for providing testosterone and optionally estrogen replacement therapy to women, U.S. Pat. No. 5,460,820). The matrix of the patch can comprise a basal support layer, such as an acrylic or ethylene/vinyl acetate copolymer or a polyurethane foam or cellulosic material, and an adhesive, such as, but not limited to, polysiloxane. In the compositions of the present invention, the polymer matrix also contains a 5-aminoslaicylate compound, as described above, and optionally, a penetration-enhancing vehicle or carrier, such as N-vinyl-2-pyrrolidone, N-(2- hydroxyethyl)-pyrrolidone, or 3-hydroxy-N-methyl-2-pyrrolidone. The adhesive patch may be pressure- sensitive, to release the 5-aminosalicylate compound across the skin of the patient when the patch matrix has been applied to the skin. The patch may optionally comprise an inert backing layer in addition to a matrix layer, or can comprise multiple dosage units or layers. The present invention is also related to a method of inhibiting the growth of a bacterial species on a foodstuff. A foodstuff for purposes of the present invention is any food or beverage that can be ingested. This method is intended for bacteriostatic food packaging or handling and relies on the antimicrobial properties of 5-aminosalicylates. The present method involves treating a food-contacting surface of a material that is pharmaceutically acceptable for food packaging or food handling purposes, with a 5-aminosalicylate compound in an amount effective to inhibit the growth of a bacterial species on a food contacting the surface.
Acceptable materials for food packaging or handling include, but are not limited to, paper, wood, cardboard, or other cellulosic polymers, including transparent and non-transparent cellulosic polymers; plastic polymers; waxes; glass or silica; pottery, earthenware, or other ceramic; or metallic materials.
A food-contacting surface of a suitable polymeric material is treated with a 5-aminosalicylate compound by enmeshing, implanting, or impregnating the compound within the polymeric material, by means known to the artisan skilled in food packaging and handling materials.
Alternatively, a suitable polymeric material is treated with a 5-aminosalicylate compound by covalent linkage, such as by conjugation of the 5-aminosalicylate compound to the polymeric material through, for example, diazo bonds or amide bonds.
Alternatively, a polymeric or non-polymeric material is treated with a 5-aminosalicylate compound by coating the material with a coating formulation suitable for a desired coating means, such as, but not limited to, dipping, spraying or brushing onto a surface intended for food contact. A suitable coating formulation contains, in addition to a 5-aminosalicylate compound, an appropriate carrier(s), which carriers are known in the art. For example, the skilled practitioner can employ as a carrier a non- toxic polymeric resin, additionally containing an effective amount of a 5-aminosalicylate compound, which resin can be used to coat the food-contacting surface of the material, hardening in place upon it. An effective amount of a 5-aminosalicylate compound for treating the food-contacting surface of the material is between 0.1 and 10 mg per cm2 of the surface. Most preferably, the amount of 5- aminosalicylate compound is 0.4 to 2 mg per cm2 of the surface. This is sufficient concentration to inhibit the growth of a food-poisoning or botulism-causing bacterial species as demonstrated in the detailed examples described herein. While the amount of the 5-aminosalicylate compound on the food- contacting surface of the material is relatively low, it is preferable not to use sulphasalazine as the 5- aminosalicylate compound, due to the possibility of sulfapyridine-related reactions in a minority of food consumers, as described above. Also, it is preferable not to use a 5-aminosalicylate-conjugated bile acid, as this may adversely affect the stability or taste of the food contacting the surface
The present invention is also related to food containers and food-handling implements for holding a foodstuff, which includes containing, packaging, covering, storing, displaying, processing, cutting, chopping, impaling, kneading, manipulating or otherwise handling the foodstuff, such that a surface of the food container or implement comes in contact with the foodstuff.
The present food containers and food-handling implements comprise a material suitable for contact with food and have a food-contacting surface treated with a 5-aminosalicylate compound, as described above, in an amount effective to inhibit the growth of a bacterial species. The containers and implements are in any suitable disposable (i.e., single-use) or non-disposable (i.e., multi-use) configuration capable of holding a foodstuff. These configurations include, but are not limited to, foils, shear wraps, sheets, papers, waxed papers, bags, cartons, trays, plates, bowls, covered and uncovered storage vessels, serving dishes, cups, cans, jars, bottles, or any other suitable container configuration for a particular foodstuff. Additional configurations especially useful for food handling purposes include, but are not limited to, gloves or mitts; utensils such as forks, spoons, knives, slicers, processors, juicers, grinders, chippers, hooks, presses, screws, openers, cutters, peelers, tongs, ladles, scoops, cups, chutes or spatulas; and cutting boards, kneading boards, mixing bowls, drying or cooling racks, or shelves.
The present method, containers, and implements are especially useful in inhibiting the growth of bacterial species that can release pathogenic exotoxins, such as, but not limited to, Clostridium perfringens, or Clostridium botulinum, the exotoxins of which cause botulism. They are especially, but not exclusively, useful in commercial and institutional food preparation contexts where food is handled and packaged in bulk, such as, but not limited to, food processing factories, canning plants, slaughterhouses, restaurants, cafeterias, salad bars, grocery outlets, and hospitals. The present method, containers and implements are useful for any kinds of foodstuff. They are particularly useful in situations where food is processed, packaged, stored, or displayed at a temperature, at or above room temperature, but insufficiently hot to kill bacterial cells and spores, for example, when food is kept under a warming light. But the present method, containers and implements are also useful in the home kitchen. The present invention is also related to antimicrobial cleansers, polishes, paints, sprays, soaps, or detergents formulated for application to surfaces to inhibit the growth of a bacterial species thereon. These surfaces include, but are not limited to surfaces, such as, countertops, desks, chairs, laboratory benches, tables, floors, bed stands, tools or equipment, doorknobs, and windows. The present cleansers, polishes, paints, sprays, soaps, or detergents contain a 5-aminosalicylate compound that provides a bacteriostatic property to them. They can optionally contain suitable solvent(s), carrier(s), thickeners, pigments, fragrances, deodorizers, emulsifiers, surfactants, wetting agents, waxes, or oils. A preferred embodiment is a formulation for external use as a pharmaceutically acceptable skin cleanser, particularly for the surfaces of human hands.
In the present cleansers, polishes, paints, sprays, soaps, and detergents, the concentration of the 5-aminosalicylate compound is between 0.625 and 200 mg per mL. Most preferably the concentration of 5-aminosalicylate compound is 1.25 to 50 mg per mL. The present cleansers, polishes, paints, sprays, soaps, and detergents are useful in homes and institutions, particularly but not exclusively in hospital settings for the prevention of nosocomial infections.
The present invention is also related to an antimicrobial method for inhibiting the growth of a bacterial species in a foodstuff and to foodstuffs containing a 5-aminosalicylate compound. Bacterial growth in foodstuff, if uninhibited, can result in the release of bacterial exotoxins that can cause illness or death in a human or non-human vertebrate that consumes the foodstuff. The present method and foodstuffs are particularly useful in preventing clostridial food poisoning by, for example, Clostridium perfringens, or by Clostridium botulinum, the exotoxins of which cause botulism.
The present method employs a 5-aminosalicylate compound added to the foodstuff. Any foodstuff can be treated using the present method, but foods for which the present method is especially useful include non-acidic foods, such as mayonnaise or other egg products, potato products, and other vegetable or meat products. Five-aminosalicylates are autoclavable and thus may be used effectively in canned foods.
The 5-aminosalicylate compound for adding to the foodstuff can be part of any comestible formulation that can also include a suitable medium or carrier for convenient mixing or dissolving into a particular foodstuff. The medium or carrier is preferably one that will not interfere with the familiar flavor of the food of interest, such as are known by the artisan skilled in food processing techniques. An effective amount of a 5-aminosalicylate compound to be added to a foodstuff is less than that required for administration to a vertebrate subject. The present foodstuffs contain a concentration of a 5-aminosalicylate compound between 0.625 and 10 mg per gram of the foodstuff. Most preferably the concentration of 5-aminosalicylate compound is 1.25 to 5 mg per gram of foodstuff. This is sufficient concentration to inhibit the growth of a food-poisoning or botulism-causing bacterial species as demonstrated in the detailed examples described herein.
While the concentration of the 5-aminosalicylate compound in the foodstuff is relatively low, it is again, preferable not to use sulphasalazine as the 5-aminosalicylate compound, due to the possibility of sulfapyridine-related reactions in a minority of food consumers, as described above. Also, it is preferable not to use a 5-aminosalicylate-conjugated bile acid, as this may adversely affect the stability or taste of the foodstuff before consumption.
The present antimicrobial method for inhibiting bacterial growth in a foodstuff and foodstuffs employing the antimicrobial properties of 5-aminosalicylates are useful alternative means of preventing food poisoning or botulism, instead of nitrite preservatives or gamma irradiation of food, which are disfavored by a sizeable number of consumers.
The methods and pharmaceutical compositions of the present invention provide a much needed addition to the antimicrobial armamentarium against bacterial overgrowths and infections, especially clostridial infections that affect great numbers of humans and animals worldwide. Five-aminosalicylate compounds are modestly priced relative to other antibiotics and are conveniently administered. The administration of 5-aminosalicylates is well tolerated by the vast majority of patients with only rare side effects, which, of course, the practitioner should monitor in each individual patient. For example, 5-aminosalicylic acid is known to pose no substantial teratogenic risk in humans and can be administered safely to pregnant women. (O. Diav-Citrin et al, The safety of mesalamine in human pregnancy: aprospective controlled cohort study , Gastroenterol. 114(l):23-28 [1998]; P. Marteau et al. , Foetal outcome in women with inflammatory bowel disease treated during pregnancy with oral mesalazine microgranules, Aliment. Pharmacol. Ther. 12(11): 1101-08 [1998]; CM. Bell and F.M. Habal, Safety of topical 5-aminosalicylic acid in pregnancy, Am. J. Gastroenterol. 92(12): 2201-02 [1997]). The use of sulphasalazine requires closer monitoring, as mentioned above.
The foregoing applications for the methods and compositions of the present invention are illustrative and by no means exhaustive. The invention will now be described in greater detail by reference to the following non-limiting examples.
EXAMPLES
The antimicrobial properties of a 5-aminosalicylate compound, mesalamine, were tested using five organisms, commonly, but not exclusively, found in gastrointestinal tracts. Example 1. Bacterial Cultures
Bacteroides fragilis (Gram negative anaerobe), Clostridium perfringens (Gram positive obligate anaerobe), Escherichia coli (Gram negative facultative anaerobe), a Lactobacillus isolate (Gram positive aerotolerant anaerobe), and Enterococcus faecalis (Gram negative facultative anaerobe) were examined, because each produces a detectable gaseous fermentation product under certain anaerobic growth conditions. All cultures were maintained on standard blood agar plates ([g/L distilled H20]: pancreatic digest of casein, 15.0; papale digest of soybean meal, 5.0; NaCl, 5.0; agar, 20.0; yeast extract, 5.0; hemin, 0.003; vitamin Kl, 0.01; L-cystine, 0.4; defibrinated sheeps' blood, 5.0 %) at 37 °C in an anaerobic C02 chamber (Bacteroides, Lactobacillus and Clostridium) or aerobically (Escherichia and Enterococcus). Inocula for plating experiments were taken aseptically by probe from distinct colonies on culture plates that were no more than 72 hours (anaerobic) or 24 hours (aerobic) old.
Example 2. Plating and Colony Counting
Mesalamine was obtained commercially in the form of Asacol (Procter and Gamble). The enteric coating on 400 mg Asacol tablets was removed with acetone. After the coating was dissolved, intact tablets were air dried and sterilized by autoclaving at 121 °C, 15 psi, for 15 minutes. The sterile tablets were aseptically dissolved in sterile distilled water.
One milliliter-aliquots of serial dilutions containing 0, 3.125, 6.25, 12.5, 25, 50, and 100 mg/mL of mesalamine in sterile distilled water were placed onto anaerobic and aerobic blood 10-mL agar plates. To facilitate impregnation into the agar, the plates were allowed to dry for about 6 hours. Aliquots of 0.5 McFarland units of cell suspensions of each bacterial species were separately diluted 1: 106 in sterile saline (0.15 M NaCl), and I mL of each diluted cell suspension was plated on 4 replicate plates at each concentration of mesalamine. All plates were incubated at 37 °C Plates inoculated with E. coli or Enterococcus faecalis were incubated aerobically for 24 hours, and plates inoculated with C. perfringens, B. fragilis, or Lactobacillus were allowed to incubate anaerobically in GasPak chambers for 48 hours. Colony counts were assessed from each of the four plates of each concentration of mesalamine for each species. Paired t-test was used to compare groups of readings. Four replicate control plates at each concentration of mesalamine were incubated as described above after plating with 1 mL of sterile saline. (All controls were negative for bacterial colonies.)
Example 3. Results Resulting colony counts for E. coli and a C. perfringens isolate are tabulated in Table 1. In the concentration range between 12.5 and 100 mg/mL, mesalamine inhibited the growth of the C. perfringens isolate but had no effect on E. coli colony counts. Similar growth inhibition by mesalamine was detected for Clostridium perfringens ATCC 13124 and Clostridium difficile ATCC 9689. (Data not shown.) But mesalamine did not affect the colony numbers of Lactobacillus, Enterococcus, or Bacteroides. O 00/45803
22
Table 1. Growth inhibition of a Clostridium perfringens isolate by mesalamine.
Results: (mean colony count ± SE)
Mesalamine (mg/mL) 0 3.125 6.25 12.5 25 50 100
E. coli 28.5±2.3 29.3±1.9 29.0±2.2 24.0±4.8 28.3±4.6 33.3±2.6 27.8±2.5 C. perfringens 29.5±1.3 38.7±33.2 24.8±13.9 l.O±O.δ* 2.0±4.0* 1.8±2.4* 1.3±2.5*
*p,0.0001
These results demonstrate that 5-aminosalicylates exert a selective antimicrobial action, for example on the growth of Clostridium. This selectivity of action is one of the advantages of the present invention, a feature which is useful in fighting the serious problem of clostridial infections and botulism food poisoning.

Claims

1. A method of inhibiting the growth of a bacterial species in a human subject, comprising: administering to a human subject having a bacterial infection or overgrowth a pharmaceutically acceptable composition containing a 5-aminosalicylate compound in a dose effective to inhibit the growth of a bacterial species in the human subject.
2. A method of inhibiting the growth of a bacterial species in a human gastrointestinal tract, comprising: administering to a human subject having a gastrointestinal bacterial infection or overgrowth a pharmaceutically acceptable composition containing a 5-aminosalicylate compound in a dose effective to inhibit the growth of a bacterial species in the gastrointestinal tract of the human subject.
3. A method of inhibiting growth of a bacterial species in a non-human vertebrate, comprising: administering to a non-human vertebrate subject having a bacterial infection or overgrowth a pharmaceutically acceptable composition containing a 5-aminosalicylate compound in a dose effective to inhibit the growth of a bacterial species in the non-human vertebrate subject.
4. A method of inhibiting the growth of a bacterial species in a non-human vertebrate gastrointestinal tract, comprising: administering to a non-human vertebrate subject having a gastrointestinal bacterial infection or overgrowth a pharmaceutically acceptable composition containing a 5-aminosalicylate compound in a dose effective to inhibit the growth of a bacterial species in the gastrointestinal tract of said non-human vertebrate subject.
5. The method of any of Claims 1, 2, 3, or 4, wherein said 5-aminosalicylate compound is mesalamine, sulphasalazine, olsalazine, ipsalazine, salicylazobenzoic acid, or balsalazide.
6. The method of any of Claims 2 or 4, wherein said 5-aminosalicylate compound is a conjugated bile acid.
7. The method of Claim 6, wherein said conjugated bile acid is ursodeoxycholic acid-5- aminosalicylic acid.
8. The method of any of Claims 1, 2, 3, or 4, wherein said composition is administered by a non-systemic delivery route.
9. The method of Claim 8, wherein said non-systemic delivery route is a colonic delivery route.
10. The method of Claim 8, wherein said non-systemic delivery route is an ingestive delivery route.
11. The method of any of Claims 1 or 3, wherein said composition is administered by a non- systemic delivery route comprising a topical application of a cream, gel, or ointment.
12. The method of any of Claims 1, 2, 3, or 4, wherein said composition is administered by a systemic delivery route.
13. The method of Claim 12, wherein said systemic delivery route is by ingestion, injection, intravenous drip, or implant.
14. The method of Claim 12, wherein said systemic delivery route is a transdermal delivery route.
15. The method of Claim 12, wherein said systemic delivery route is a transmucosal delivery route.
16. The method of any of Claims 1, 2, 3, or 4, wherein said bacterial species is an obligate anaerobe.
17. The method of Claim 16, wherein said bacterial species is a Clostridium species.
18. The method of Claim 17, wherein said bacterial species is Clostridium perfringens, Clostridium difficile, Clostridium tetani or Clostridium botulinum.
19. The method of any of Claims 3 or 4, wherein said non-human vertebrate is a domestic or farm animal.
20. The method of Claim 19, wherein said non-human vertebrate is a mammal or bird.
21. The method of Claim 19, wherein said non-human vertebrate is a non-human primate, mouse, rat, rabbit, gerbil, hamster, canine, feline, ovine, bovine, swine, pachyderm, equine, or marine mammal.
22. The method of Claim 19, wherein said non-human vertebrate is a duck, chicken, goose, turkey, ostrich, emu, dove, pigeon, quail, pheasant, peafowl, or guinea fowl.
23. A method of inhibiting the growth of a bacterial species in a foodstuff, comprising: adding to a foodstuff a 5-aminosalicylate compound in an amount effective to inhibit the growth of a bacterial species in said foodstuff.
24. The method of Claim 23, wherein said bacterial species is Clostridium perfringens, Clostridium difficile, Clostridium botulinum, or Clostridium tetani.
25. The method of Claim 23, wherein said 5-aminosalicylate compound is mesalamine, sulphasalazine, olsalazine, ipsalazine, salicylazobenzoic acid, or balsalazide.
26. An antimicrobial pharmaceutical composition comprising a 5 -aminosalicylate compound formulated in a delivery system to deliver a dose between 6.25 and 150 mg/day.
27. The antimicrobial pharmaceutical composition of Claim 26, wherein said dose is between 12.5 and 150 mg/day.
28. An antimicrobial pharmaceutical composition comprising a 5-aminosalicylate compound formulated in an ingestive delivery system to deliver a dose between 6.25 and 200 mg/day.
29. The antimicrobial pharmaceutical composition of Claim 28, wherein said dose is between 12.5 and 200 mg/day.
30. An antimicrobial pharmaceutical composition comprising a 5-aminosalicylate compound formulated in a delivery system to deliver a dose over 6,450 mg/day.
31. An antimicrobial pharmaceutical composition comprising a 5-aminosalicylate compound formulated in an ingestive delivery system to deliver a dose over 6,450 mg/day.
32. The antimicrobial pharmaceutical composition of any of Claims 26, 28, 30, or 31, wherein said 5-aminosalicylate compound is mesalamine, sulphasalazine, olsalazine, ipsalazine, salicylazobenzoic acid, or balsalazide.
33. The antimicrobial pharmaceutical composition of any of Claims 26, 28, 30, or 31, wherein said 5-aminosalicylate compound is a conjugated bile acid.
34. The antimicrobial pharmaceutical composition of Claim 33, wherein said conjugated bile acid is ursodeoxycholic acid-5-aminosalicylic acid.
35. The antimicrobial pharmaceutical composition of any of Claims 26 or 30, wherein said delivery system is an injection, intravenous drip, or implant delivery system.
36. The antimicrobial pharmaceutical composition of any of Claims 26 or 30, wherein said delivery system is a transdermal delivery system.
37. The antimicrobial pharmaceutical composition of Claim 36, wherein said delivery system comprises an adhesive patch.
38. The antimicrobial pharmaceutical composition of any of Claims 26 or 30, wherein said delivery system comprises a gel or cream.
39. The antimicrobial pharmaceutical composition of any of Claims 26 or 30, wherein said delivery system is a transmucosal delivery system.
40. The antimicrobial pharmaceutical composition of Claim 39, wherein said delivery system is an oral transmucosal delivery system.
41. The antimicrobial pharmaceutical composition of Claim 39, wherein said delivery system is a vaginal transmucosal delivery system.
42. The antimicrobial pharmaceutical composition of any of Claims 26 or 30, formulated for a colonic delivery system.
43. The antimicrobial pharmaceutical composition of Claim42, wherein the colonic delivery system is an enema.
44. The antimicrobial pharmaceutical composition of Claim 42, wherein the colonic delivery system is a suppository, gel, or foam.
45. The antimicrobial pharmaceutical composition of any of Claims 28 or 31, wherein the ingestive delivery system is a tablet, capsule, caplet, or food additive.
46. The antimicrobial pharmaceutical composition of Claim 45, wherein the ingestive delivery system comprises an enteric coating to prevent esophageal or gastric release of mesalamine.
47. The antimicrobial pharmaceutical composition of any of Claims 28 or 31 , wherein the ingestive delivery system comprises a lavage system.
48. The antimicrobial pharmaceutical composition of any of Claims 26, 28, 30, or 31, formulated for treating a human.
49. The antimicrobial pharmaceutical composition of Claim 48, formulated for pediatric use.
50. The antimicrobial pharmaceutical composition of any of Claims 26, 28, 30, or 31, formulated for veterinary use.
51. The antimicrobial pharmaceutical composition of Claim 50, formulated for use in a domestic or farm animal.
52. The antimicrobial pharmaceutical composition of Claim 50, formulated for use in a non- human mammal or bird.
53. The antimicrobial pharmaceutical composition of Claim50, formulated for use in anon- human primate, mouse, rat, rabbit, gerbil, hamster, canine, feline, ovine, bovine, swine, pachyderm, equine, or marine mammal.
54. The antimicrobial pharmaceutical composition of Claim 50, formulated for use in a duck, chicken, goose, turkey, ostrich, emu, dove, pigeon, quail, pheasant, peafowl, or guinea fowl.
55. A foodstuff, containing: a 5-aminosalicylate compound in a concentration between 0.625 and 10 mg per gram of the foodstuff.
56. The foodstuff of Claim 55, wherein the concentration of said 5-aminosalicylate compound is between 0.625 and 5 mg per gram of the foodstuff.
57. The foodstuff of Claim 55, wherein the concentration of said 5-aminosalicylate compound is between 0.625 and 2.5 mg per gram of the foodstuff.
58. The foodstuff of Claim 55, wherein the concentration of said 5-aminosalicylate compound is between 0.625 and 1.25 mg per gram of the foodstuff.
59. The foodstuff of Claim 55, wherein the concentration of said 5-aminosalicylate compound is 1.25 mg to 5 mg per gram of the foodstuff.
60. The foodstuff of Claim 55, wherein said 5-aminosalicylate compound is mesalamine, sulphasalazine, olsalazine, ipsalazine, salicylazobenzoic acid, or balsalazide.
61. A method of inhibiting the growth of a bacterial species on a foodstuff, comprising: treating a food-contacting surface of a material for food packaging or food handling with a 5- aminosalicylate compound; and contacting a foodstuff with said surface, whereby the growth of a bacterial species on said foodstuff is inhibited.
62. The method of Claim 61, wherein said food packaging or handling material is a cellulosic polymer.
63. The method of Claim 61, wherein said food packaging or handling material is paper, wood, or cardboard.
64. The method of Claim 61 , wherein said food packaging or handling material is a plastic polymer, wax, glass, silica, ceramic, earthenware, or metallic material.
65. The method of Claim 61, wherein said food-contacting surface comprises a surface belonging to a foil, a shear plastic wrap, a cellophane, a wrapping paper, a waxed paper, a bag, a carton, a box, a tray, a plate, a bowl, a food storage vessel, a serving dish, a cup, a can, ajar, or a bottle.
66. The method of Claim 61, wherein said food-contacting surface comprises a surface belonging to a glove, a mitt, a fork, a spoon, a knife, a sheer, a tong, a ladle, a scoop, a cup, a processor, a juicer, a grinder, a press, a hook, a chipper, a peeler, a cutter, a screw, an opener, a chute, a spatula, a cutting board, a kneading board, a bowl, a rack, or a shelf.
67. The method of Claim 61, wherein said 5-aminosalicylate compound is mesalamine, sulphasalazine, olsalazine, ipsalazine, salicylazobenzoic acid, or balsalazide.
68. The method of Claim 61. wherein said bacterial species is an obligate anaerobe.
69. The method of Claim 61, wherein said bacterial species is a Clostridium species.
70. The method of Claim 61, wherein said bacterial species is Clostridium perfringens, Clostridium difficile, Clostridium botulinum, or Clostridium tetani.
71. A food container or food-handling implement, said container or implement having a food-contacting surface, said surface treated with a 5-aminosalicylate compound in an amount effective to inhibit the growth of a bacterial species on a foodstuff contacting said surface.
72. The food container or food-handling implement of Claim 71 , wherein said container or implement is a foil, a shear plastic wrap, a cellophane, a wrapping paper, a waxed paper, a bag, a carton, a box, a tray, a plate, a bowl, a food storage vessel, a serving dish, a cup, a can, a jar, a bottle, a glove, a mitt, a fork, a spoon, a knife, a sheer, a tong, a ladle, a scoop, a cup, a processor, a juicer, a grinder, a press, a hook, a chipper, a screw, a cutter, a peeler, an opener, a chute, a spatula, a cutting board, a kneading board, a bowl, a rack, or a shelf.
73. The food container or food-handling implement of Claim 71, having an amount of a 5- aminosalicylate compound between 0.1 and 10 mg per cm2 of said food-contacting surface.
74. The food container or food-handling implement of Claim 71, having an amount of a 5- aminosalicylate compound between 0.4 and 2 mg per cm2 of said food-contacting surface.
75. The food container or food-handling implement of Claim 71, wherein said 5- aminosalicylate compound is mesalamine, sulphasalazine, olsalazine, ipsalazine, salicylazobenzoic acid, or balsalazide.
76. An antimicrobial cleanser, polish, paint, spray, soap, or detergent for applying to a surface, containing a 5-aminosalicylate compound in an amount effective to inhibit the growth of a bacterial species on said surface.
77. The antimicrobial cleanser, polish, paint, spray, soap, or detergent of Claim 76, wherein said 5-aminosalicylate compound is mesalamine, sulphasalazine, olsalazine, ipsalazine, salicylazobenzoic acid, balsalazide, or a conjugated bile acid.
78. The antimicrobial cleanser, polish, paint, spray, soap, or detergent of Claim 76, formulated for applying to an inanimate surface.
79. The antimicrobial cleanser, polish, paint, spray, soap, or detergent of Claim 76, formulated as a pharmaceutically acceptable skin cleanser.
80. The antimicrobial cleanser, polish, paint, spray, soap, or detergent of Claim 76, wherein the amount of 5-aminosalicylate compound is between 0.625 and 200 mg per mL.
81. The antimicrobial cleanser, polish, paint, spray, soap, or detergent of Claim 76, wherein the amount of 5-aminosalicylate compound is between 1.25 and 50 mg per mL.
PCT/US2000/002802 1999-02-08 2000-02-04 Use of 5-aminosalicylates as antimicrobial agents WO2000045803A2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU35879/00A AU3587900A (en) 1999-02-08 2000-02-04 Use of 5-aminosalicylates as antimicrobial agents

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US09/246,645 US6326364B1 (en) 1999-02-08 1999-02-08 Use of 5-aminosalicylates as antimicrobial agents
US09/246,645 1999-02-08

Publications (2)

Publication Number Publication Date
WO2000045803A2 true WO2000045803A2 (en) 2000-08-10
WO2000045803A3 WO2000045803A3 (en) 2001-04-05

Family

ID=22931573

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2000/002802 WO2000045803A2 (en) 1999-02-08 2000-02-04 Use of 5-aminosalicylates as antimicrobial agents

Country Status (3)

Country Link
US (1) US6326364B1 (en)
AU (1) AU3587900A (en)
WO (1) WO2000045803A2 (en)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1931354A2 (en) * 2005-08-24 2008-06-18 Salix Pharmaceuticals, Inc. Balsalazide formulations and manufacture and use thereof
EP2096912A2 (en) * 2006-11-03 2009-09-09 Salix Pharmaceuticals, Inc. Formulations and uses of 2-hydroxy-5-phenylazobenzoic acid derivatives
WO2011023573A1 (en) * 2009-08-26 2011-03-03 Basf Se Antimicrobial amino-salicylic acid derivatives
RU2454230C2 (en) * 2000-10-18 2012-06-27 Эко Энимал Хелт Лтд Treatment and prevention of diseases and infections in pigs and poultry
JP2014504650A (en) * 2011-02-04 2014-02-24 オプティマー ファーマシューティカルズ,インコーポレーテッド Treatment of bacterial infection
US9192616B2 (en) 2005-08-24 2015-11-24 Salix Pharmaceuticals, Inc. Formulations and uses of 2-hydroxy-5-phenylazobenzoic acid derivatives
AU2013228061B2 (en) * 2006-11-03 2015-11-26 Salix Pharmaceuticals, Inc. Formulations and uses of 2-hydroxy-5-phenylazobenzoic acid derivatives
US9937190B2 (en) 2004-02-06 2018-04-10 Salix Pharmaceuticals, Ltd Use of aminosalicylates in diarrhoea-predominent irritable bowel syndrome
US11122821B2 (en) 2016-12-15 2021-09-21 Societe Des Produits Nestle S.A. Compositions and methods that modulate bacteria in a companion animal

Families Citing this family (40)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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
US7048906B2 (en) 1995-05-17 2006-05-23 Cedars-Sinai Medical Center Methods of diagnosing and treating small intestinal bacterial overgrowth (SIBO) and SIBO-related conditions
US7884090B2 (en) 1999-03-17 2011-02-08 Ernest L. Bonner, Jr. Compositions and methods for the treatment of arthritis
US6765000B2 (en) * 1999-03-17 2004-07-20 Bonner Jr Ernest L Treatment for reactive arthritis or bursitis
US7691831B2 (en) 1999-03-17 2010-04-06 Ernest L. Bonner, Jr. Pharmaceutical combination and method for treatment of reactive arthritis or bursitis
EP1642885B1 (en) * 2000-08-29 2009-11-11 Biocon Limited Use of a pharmaceutical composition containing a para-aminophenyl acetic acid derivative for treating inflammatory conditions of the gastrointestinal tract
CA2359812C (en) * 2000-11-20 2004-02-10 The Procter & Gamble Company Pharmaceutical dosage form with multiple coatings for reduced impact of coating fractures
US8048924B2 (en) 2001-08-29 2011-11-01 Biocon Limited Methods and compositions employing 4-aminophenylacetic acid compounds
US20080306029A1 (en) * 2004-05-28 2008-12-11 Salix Pharmaceuticals, Inc. Prevention, Treatment, and Amelioration of Radiation Induced Enteritis
US7932366B2 (en) 2004-07-07 2011-04-26 Biocon Limited Synthesis of azo bonded immunoregulatory compounds
ITRM20050390A1 (en) * 2005-07-22 2007-01-23 Giuliani Spa COMPOUNDS AND THEIR SPECIFIC SALTS FOR PPAR RECEPTORS AND RECEPTORS FOR EGF AND THEIR USE IN MEDICAL FIELDS.
ITRM20050389A1 (en) 2005-07-22 2007-01-23 Giuliani Spa COMPOUNDS AND THEIR SPECIFIC SALTS FOR PPAR RECEPTORS AND RECEPTORS FOR EGF AND THEIR USE IN MEDICAL FIELDS.
US7178823B1 (en) * 2005-09-23 2007-02-20 Jen-Lung David Tai Portable trailer
MX2009002785A (en) * 2006-09-13 2009-03-30 Procter & Gamble Methods of treatment for ulcerative colitis.
CA2662430A1 (en) * 2006-09-13 2008-03-20 The Procter & Gamble Company Methods of treatment for ulcerative colitis using aminosalicylate
US20080159987A1 (en) * 2007-01-03 2008-07-03 Leonard Weinstock Use of Rifaximin for the Treatment of Restless Legs Syndrome
US20080161337A1 (en) * 2007-01-03 2008-07-03 Leonard Weinstock Use of Rifaximin for the Treatment of Chronic Prostatitis
US7645801B2 (en) * 2007-01-29 2010-01-12 Alaven Pharmaceutical Llc Reduced irritant enema for treatment of inflammatory bowel disease (IBD)
IE20070129A1 (en) * 2007-02-28 2008-12-24 Giuliani Int Ltd Ppar-gamma agonists stimulate enteric defensin expression
US7541384B2 (en) 2007-06-08 2009-06-02 Axcan Pharma Inc. Mesalamine suppository
US8436051B2 (en) * 2007-06-08 2013-05-07 Aptalis Pharma Canada Inc. Mesalamine suppository
US8217083B2 (en) * 2007-06-08 2012-07-10 Aptalis Pharma Canada Inc. Mesalamine suppository
US8974825B2 (en) * 2007-07-06 2015-03-10 Lupin Limited Pharmaceutical compositions for gastrointestinal drug delivery
AU2009320350B2 (en) * 2008-11-03 2015-09-24 Tufts University Methods and compositions for inhibiting Clostridium difficile spore germination and outgrowth
UA107562C2 (en) 2008-12-05 2015-01-26 METHOD OF TREATMENT OF PSORIASIS
JP5645851B2 (en) 2009-02-16 2014-12-24 ノグラ ファーマ リミテッド How to treat hair-related conditions
US20110155749A1 (en) * 2009-12-24 2011-06-30 Kathryn Jeanne Neiheiser Germ guard label and methods for using germ guard label
BR112012027520A2 (en) 2010-04-26 2015-09-15 Salix Pharmaceuticals Ltd Formulations and uses of 2-hydroxy-5-phenylazobenzoic acid derivatives for the treatment of males
EP2603198A4 (en) 2010-08-09 2014-01-08 Itesm Antimicrobial, antibacterial and spore germination inhibiting activity from an avocado extract enriched in bioactive compounds
EA030762B1 (en) 2012-02-09 2018-09-28 Ногра Фарма Лимитед Methods of treating fibrosis
US9055889B2 (en) 2012-03-20 2015-06-16 Commonwealth Laboratories, Inc. Method and apparatus for breath testing
AU2013248397A1 (en) 2012-04-18 2014-10-02 Nogra Pharma Limited Methods of treating lactose intolerance
US20140141075A1 (en) 2012-11-21 2014-05-22 Warner Chilcott Company, Llc 5-aminosalicylic acid capsule formulation
IN2014MU00097A (en) * 2014-01-10 2015-08-21 Cadila Healthcare Ltd
US20160045442A1 (en) * 2014-08-13 2016-02-18 Cadila Healthcare Limited Stable pharmaceutical compositions of mesalamine
US20180000793A1 (en) 2015-02-06 2018-01-04 Ernesto Abel-Santos Inhibiting Germination of Clostridium Perfringens Spores to Reduce Necrotic Enteritis
WO2017051208A1 (en) 2015-09-23 2017-03-30 Instituto Tecnológico y de Estudios Superiores de Monterrey Acetogenin molecules having antiplatelet and/or antithrombic activities, and methods and compositions thereof
US10932484B2 (en) 2016-10-19 2021-03-02 Instituto Tecnologico Y De Estudios Superiores De Monterrey Inhibitory activity of acetogenins against Listeria monocytogenes
US11091597B2 (en) * 2017-05-23 2021-08-17 The Research Foundation For The State University Of New York Packaging material and methods of using the same
CN113825739A (en) 2019-02-08 2021-12-21 诺格拉制药有限公司 Process for preparing 3- (4' -aminophenyl) -2-methoxypropionic acid and analogs and intermediates thereof

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3412090A (en) * 1964-07-10 1968-11-19 Geigy Chem Corp Organotin-substituted s-triazines
FR1605124A (en) * 1967-01-27 1973-03-16 Benzamide fungicides - applied by vaporisation with a cyanamide
US4863744A (en) * 1984-09-17 1989-09-05 Alza Corporation Intestine drug delivery
EP0468555A1 (en) * 1990-07-27 1992-01-29 GIULIANI S.p.A. Pharmaceutical composition for rectal administration of active principles exhibiting a prevalently topical medication action at the colon level
WO1992006690A1 (en) * 1990-10-22 1992-04-30 Borody Thomas J Treatment of non-inflammatory and non-infectious bowel disorders
WO1994028911A1 (en) * 1993-06-08 1994-12-22 Farmaceutisk Laboratorium Ferring A/S COMPOSITIONS FOR USE IN THE REGULATION OF SUBNORMAL pH VALUES IN THE INTESTINAL TRACT AND FOR TREATMENT OF BOWEL DISEASES
WO1997028795A1 (en) * 1996-02-07 1997-08-14 Joseph Chege Use of 3-amino-4-hydroxybenzoic acid for the treatment of retroviral infections
WO1998007458A1 (en) * 1996-08-19 1998-02-26 Korea Institute Of Science And Technology Surface coating method for metal implants
EP0882461A2 (en) * 1997-06-04 1998-12-09 Unitika Ltd. Medical device and production method thereof

Family Cites Families (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS57500432A (en) 1980-03-20 1982-03-11
US4960765A (en) 1980-03-20 1990-10-02 Farmaceutisk Laboratorium Ferring A/S Pharmaceutical composition and method for the treatment of colitis ulcerosa and Crohn's disease by oral administration
USRE33239E (en) 1983-09-06 1990-06-26 Farmaceutisk Laboratorium Ferring A/S Packaged stable enema solution or suspension containing 5-aminosalicyclic acid
US4657900A (en) 1983-09-27 1987-04-14 Rowell Laboratories Pharmaceutical article of manufacture comprising a bisulfite stabilized aqueous solution of 5-aminosalicylic acid and method
GB8909559D0 (en) 1989-04-26 1989-06-14 Smith Kline French Lab Pharmaceutical compositions
US5010069A (en) 1989-05-15 1991-04-23 Marion Laboratories, Inc. Stable liquid form of 5-aminosalicylic acid
US5120306A (en) 1990-03-21 1992-06-09 Gosselin Leon F Direct delivery of anti-inflammatories to the proximal small bowel
US5519014A (en) 1990-10-22 1996-05-21 Borody; Thomas J. Treatment of non-inflammatory and non-infectious bowel disorders
JPH04300801A (en) * 1991-03-29 1992-10-23 Nippon Paint Co Ltd Intercalate composition of antibacterial substance
CA2110376C (en) 1991-06-07 2003-02-11 Adrianus J. Wittebrood Pharmaceutical enema preparation
JPH0638591A (en) * 1992-07-10 1994-02-10 Ricoh Co Ltd Drive control method for stepping motor
IT1277663B1 (en) 1995-09-28 1997-11-11 Crinos Industria Farmaco STABLE AQUEOUS SUSPENSIONS OF MESALAZINE FOR TOPICAL USE

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3412090A (en) * 1964-07-10 1968-11-19 Geigy Chem Corp Organotin-substituted s-triazines
FR1605124A (en) * 1967-01-27 1973-03-16 Benzamide fungicides - applied by vaporisation with a cyanamide
US4863744A (en) * 1984-09-17 1989-09-05 Alza Corporation Intestine drug delivery
EP0468555A1 (en) * 1990-07-27 1992-01-29 GIULIANI S.p.A. Pharmaceutical composition for rectal administration of active principles exhibiting a prevalently topical medication action at the colon level
WO1992006690A1 (en) * 1990-10-22 1992-04-30 Borody Thomas J Treatment of non-inflammatory and non-infectious bowel disorders
WO1994028911A1 (en) * 1993-06-08 1994-12-22 Farmaceutisk Laboratorium Ferring A/S COMPOSITIONS FOR USE IN THE REGULATION OF SUBNORMAL pH VALUES IN THE INTESTINAL TRACT AND FOR TREATMENT OF BOWEL DISEASES
WO1997028795A1 (en) * 1996-02-07 1997-08-14 Joseph Chege Use of 3-amino-4-hydroxybenzoic acid for the treatment of retroviral infections
WO1998007458A1 (en) * 1996-08-19 1998-02-26 Korea Institute Of Science And Technology Surface coating method for metal implants
EP0882461A2 (en) * 1997-06-04 1998-12-09 Unitika Ltd. Medical device and production method thereof

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
DATABASE WPI Week 197145 Derwent Publications Ltd., London, GB; AN 1971-72342S XP002154689 & JP 46 038591 A (TAKEDA CHEM IND LTD) *
DATABASE WPI Week 199249 Derwent Publications Ltd., London, GB; AN 1992-403352 XP002154690 & JP 04 300801 A (NIPPON PAINT CO LTD), 23 October 1992 (1992-10-23) *
KANG, G. ET AL: "Salicylate inhibits fimbriae mediated HEp-2 cell adherence of and hemagglutination by enteroaggregative Escherichia coli" FEMS MICROBIOL. LETT. (1998), 166(2), 257-265 , XP000972470 *
SANDBERG-GERTZEN, H. ET AL: "In vitro effects of sulphasalazine, azodisal sodium, and their metabolites on Clostridium difficile and some other fecal bacteria" SCAND. J. GASTROENTEROL. (1985), 20(5), 607-12 , XP000972467 *
TRIADAFILOPOULOS, GEORGE ET AL: "Comparative study of Clostridium difficile toxin A and cholera toxin in rabbit ileum" GASTROENTEROLOGY (1989), 97(5), 1186-92 , XP000972469 *

Cited By (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
RU2454230C2 (en) * 2000-10-18 2012-06-27 Эко Энимал Хелт Лтд Treatment and prevention of diseases and infections in pigs and poultry
US10328088B2 (en) 2004-02-06 2019-06-25 Salix Pharmaceuticals, Inc. Use of aminosalicylates in diarrhoea-predominent irritable bowel syndrome
US9937190B2 (en) 2004-02-06 2018-04-10 Salix Pharmaceuticals, Ltd Use of aminosalicylates in diarrhoea-predominent irritable bowel syndrome
EP1931354A4 (en) * 2005-08-24 2010-03-24 Salix Pharmaceuticals Inc Balsalazide formulations and manufacture and use thereof
EP1931354A2 (en) * 2005-08-24 2008-06-18 Salix Pharmaceuticals, Inc. Balsalazide formulations and manufacture and use thereof
US9192616B2 (en) 2005-08-24 2015-11-24 Salix Pharmaceuticals, Inc. Formulations and uses of 2-hydroxy-5-phenylazobenzoic acid derivatives
JP2010509326A (en) * 2006-11-03 2010-03-25 サリックス ファーマシューティカルズ, インコーポレイテッド Formulation and use of 2-hydroxy-5-phenylbenzoic acid derivatives
JP2013032395A (en) * 2006-11-03 2013-02-14 Salix Pharmaceuticals Inc Formulation and use of 2-hydroxy-5-phenylazobenzoic acid derivative
AU2007322362B2 (en) * 2006-11-03 2013-06-20 Salix Pharmaceuticals, Inc. Formulations and uses of 2-hydroxy-5-phenylazobenzoic acid derivatives
US8921344B2 (en) 2006-11-03 2014-12-30 Salix Pharmaceuticals, Inc. Formulations and uses of 2-hydroxy-5-phenylazobenzoic acid derivatives
AU2013228061B2 (en) * 2006-11-03 2015-11-26 Salix Pharmaceuticals, Inc. Formulations and uses of 2-hydroxy-5-phenylazobenzoic acid derivatives
EP2096912A4 (en) * 2006-11-03 2010-01-06 Salix Pharmaceuticals Inc Formulations and uses of 2-hydroxy-5-phenylazobenzoic acid derivatives
EP2096912A2 (en) * 2006-11-03 2009-09-09 Salix Pharmaceuticals, Inc. Formulations and uses of 2-hydroxy-5-phenylazobenzoic acid derivatives
JP2013503122A (en) * 2009-08-26 2013-01-31 ビーエーエスエフ ソシエタス・ヨーロピア Antimicrobial aminosalicylic acid derivatives
WO2011023573A1 (en) * 2009-08-26 2011-03-03 Basf Se Antimicrobial amino-salicylic acid derivatives
US9644175B2 (en) 2009-08-26 2017-05-09 Basf Se Antimicrobial amino-salicylic acid derivatives
KR101759360B1 (en) 2009-08-26 2017-07-18 바스프 에스이 Antimicrobial amino-salicylic acid derivatives
JP2014504650A (en) * 2011-02-04 2014-02-24 オプティマー ファーマシューティカルズ,インコーポレーテッド Treatment of bacterial infection
US11122821B2 (en) 2016-12-15 2021-09-21 Societe Des Produits Nestle S.A. Compositions and methods that modulate bacteria in a companion animal

Also Published As

Publication number Publication date
AU3587900A (en) 2000-08-25
WO2000045803A3 (en) 2001-04-05
US6326364B1 (en) 2001-12-04

Similar Documents

Publication Publication Date Title
US6326364B1 (en) Use of 5-aminosalicylates as antimicrobial agents
US7375080B1 (en) Immobilized lactoferrin (Im-LF) antimicrobial agents and uses thereof
KR101925508B1 (en) Pharmaceutical compositions containing pediococcus and methods for reducing the symptoms of gastroenterological syndromes
Aarestrup Association between decreased susceptibility to a new antibiotic for treatment of human diseases, everninomicin (SCH 27899), and resistance to an antibiotic used for growth promotion in animals, avilamycin
US20080107699A1 (en) Method of using topical probiotics for the inhibition of surface contamination by a pathogenic microorganism and composition therefor
CN105307654A (en) Compositions and methods for treating microbiota-related psychotropic conditions and diseases
KR20040018319A (en) Probiotic compounds derived from lactobacillus casei strain ke01
US11590182B2 (en) Methods and compositions to modulate antibiotic resistance and gastrointestinal microbiota
US20220347257A1 (en) Quorum-sensing inhibitors and/or postbiotic metabolites and related methods
Traub-Dargatz et al. Clostridia-associated enterocolitis in adult horses and foals
Ammar et al. Campylobacter as a major foodborne pathogen: A review of its characteristics, pathogenesis, antimicrobial resistance and control
TWI337079B (en) Compositions comprising oligosaccharides
Grønstøl et al. Experimental Salmonella infection in calves. 1. The effect of stress factors on the carrier state
Jayne‐Williams et al. The relationship between the intestinal microflora and the effects of diets containing raw navy beans (Phaseolus vulgaris) on the growth of Japanese quail (Coturnix coturnix japonica)
Black et al. Mecillinam, a new prophylactic for travellers' diarrhoea: a prospective double-blind study in tourists travelling to Egypt and the Far East
Ghosh et al. Antibiotic resistance in Enterococci: A food safety perspective
Levent et al. In vitro effects of Thymol-β-D-glucopyranoside on Salmonella enterica serovar Typhimurium and Escherichia coli K88
Goodman et al. Effects of erythromycin and ciprofloxacin on chronic fecal excretion of Campylobacter species in marmosets
Keeling et al. Medical management of the rhesus monkey
Hines et al. Pseudomembranous enteritis associated with ciprofloxacin and Clostridium difficile in a penguin (Eudyptes chrysolophus)
Bryan Diseases Transmitted by Foods:(a Classification and Summary)
KR102208837B1 (en) Composition for inhibiting adhesion, invasion of bacteria or antibacterial resistance comprising methyl gallate and fluoroquinolone antibacterial agent
EL-PRINCE et al. Investigation of milk and some dairy products for fecal pollution indicators
Meng et al. Shiga Escherichia Toxin-Producing coli
Hamdan Campylobacter infection

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AL AM AT AU AZ BA BB BG BR BY CA CH CN CR CU CZ DE DK DM EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
AK Designated states

Kind code of ref document: A3

Designated state(s): AE AL AM AT AU AZ BA BB BG BR BY CA CH CN CR CU CZ DE DK DM EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A3

Designated state(s): GH GM KE LS MW SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

122 Ep: pct application non-entry in european phase