WO2015181534A1 - Anti-bacterial lysate of probiotic bacteria - Google Patents

Anti-bacterial lysate of probiotic bacteria Download PDF

Info

Publication number
WO2015181534A1
WO2015181534A1 PCT/GB2015/051529 GB2015051529W WO2015181534A1 WO 2015181534 A1 WO2015181534 A1 WO 2015181534A1 GB 2015051529 W GB2015051529 W GB 2015051529W WO 2015181534 A1 WO2015181534 A1 WO 2015181534A1
Authority
WO
WIPO (PCT)
Prior art keywords
infection
aureus
bacteria
keratinocytes
composition
Prior art date
Application number
PCT/GB2015/051529
Other languages
English (en)
French (fr)
Inventor
Catherine O'neill
Andrew McBAIN
Original Assignee
The University Of Manchester
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 The University Of Manchester filed Critical The University Of Manchester
Priority to ES15727045T priority Critical patent/ES2842215T3/es
Priority to JP2016567543A priority patent/JP6741595B2/ja
Priority to CA2950510A priority patent/CA2950510C/en
Priority to EP15727045.5A priority patent/EP3148558B1/en
Priority to CN201580028700.4A priority patent/CN106794205B/zh
Priority to AU2015265698A priority patent/AU2015265698B2/en
Priority to US15/314,897 priority patent/US10702562B2/en
Publication of WO2015181534A1 publication Critical patent/WO2015181534A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/74Bacteria
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/74Bacteria
    • A61K35/741Probiotics
    • A61K35/744Lactic acid bacteria, e.g. enterococci, pediococci, lactococci, streptococci or leuconostocs
    • A61K35/747Lactobacilli, e.g. L. acidophilus or L. brevis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0014Skin, i.e. galenical aspects of topical compositions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K2035/11Medicinal preparations comprising living procariotic cells

Definitions

  • the present invention relates to probiotic bacteria and particularly, although not exclusively, to anti-bacterial compositions derived from probiotic bacteria.
  • probiotics are beneficial to gut health.
  • lactobacillus species inhibit the adhesion of Enterobacter sakazakii to intestinal mucus by competitive exclusion (32).
  • Probiotics are also able to produce antimicrobial peptides (bacteriocins) and acids.
  • probiotics may have positive impacts on the gut, their potential effects on other systems, such as the mouth (18) and the urogenital tract (44) have also begun to be investigated.
  • a study in 2001 examining the impact of oral administration of Lactobacilli in a clinical trial of women with bacterial vaginosis, showed that Lactobacilli could indeed inhibit the colonization of uro-epithelial cells by pathogens (44).
  • the topical application of probiotics to the skin has been investigated in a limited number of studies.
  • Topical application of sonicated Streptococcus salivarius strains to patients suffering from atopic dermatitis resulted in improved barrier function apparently through increasing the level of ceramides in the stratum corneum (13).
  • Topically applied L. plantarum for treatment of infected wounds resulted in improved tissue repair in a mouse burn model and prevention of infection in chronic leg ulcers and burns in humans (41 , 42).
  • the mechanisms underlying these effects are not well understood.
  • Staphylococcus aureus is both a transient coloniser of skin and a major opportunistic skin pathogen, causing diseases ranging from impetigo to life threatening conditions such as sepsis (25).
  • Lreuteri could protect epidermal keratinocytes from the toxic effects of S. aureus via competitive exclusion of the pathogen from keratinocyte binding sites (43).
  • the inventors have now identified Lrhamnosus GG as a second probiotic with the ability to protect skin cells from the effects of S. aureus.
  • Lrhamnosus GG uses multiple mechanisms to protect against infection including inhibition of S. aureus growth, competitive exclusion and displacement of the pathogen from keratinocytes.
  • Lactobacillus rhamnosus GG can inhibit Staphylococcus aureus infection of human primary keratinocytes in culture.
  • a probiotic bacterium Lactobacillus rhamnosus GG can inhibit Staphylococcus aureus infection of human primary keratinocytes in culture.
  • primary human keratinocytes were exposed to S. aureus, only 25% of the keratinocytes remained viable at 24 h afterwards.
  • the inventors have previously demonstrated that probiotic bacteria and lysates thereof in protecting cells against infection by pathogenic bacteria such as S. aureus (see
  • probiotic bacteria are able to protect cells from infection by at least two mechanisms. Firstly, the probiotic bacteria may be able to reduce or inhibit the growth of pathogenic bacteria through one or more agents contained within the probiotic bacterium that are able to directly inhibit growth and/or viability of the pathogenic bacteria. Secondly, and as identified herein, one or more agents that are secreted from the probiotic bacteria (and thus present in the culture media) are able to inhibit the ability of the pathogenic bacteria to infect the cells, possibly through preventing adhesion of the pathogenic bacteria to the cells. Material secreted by the probiotic bacteria is therefore protective against pathogenic bacterial infection. Thus, the secreted material has anti-bacterial, or anti-infective properties that can be harnessed in a variety of anti-bacterial compositions as described here.
  • the invention includes the combination of the aspects and preferred features described except where such a combination is clearly impermissible or expressly avoided.
  • probiotic bacteria are commonly defined as “live microorganisms which when administered in adequate amounts confer a health benefit on the host”. Studies in the gut have demonstrated the ability of probiotic bacteria to inhibit colonisation by pathogens through mechanisms including exclusion, competition and displacement of pathogen attachment to the host tissues. As used herein, the term “probiotic bacterium” may also refer to such bacteria when they are no longer alive, for example following inactivation by heat or radiation. Lactobacillus rhamnosus
  • the invention particularly relates to probiotic bacteria of the species Lactobacillus rhamnosus. Such bacteria were originally considered a subspecies of Lactobacillus casei, but later genetic research found it to be a species of its own. A number of probiotic bacteria of the species Lactobacillus rhamnosus. Such bacteria were originally considered a subspecies of Lactobacillus casei, but later genetic research found it to be a species of its own. A number of
  • strains 1-1720 Pieris 1-1720 (Pasteur collection
  • Lrhamnosus GG (also referred to herein as LGG) is deposited at ATCC (American Tissue Culture Collection) under accession number ATCC 53103. LGG was isolated in 1983 from the intestinal tract of a healthy human being by Gorbach and Goldin.
  • compositions according to the invention comprise or consist of secreted material from probiotic bacteria.
  • Secreted material refers to material secreted from a probiotic bacterium.
  • the secreted material may be a single agent. It may be a mixture of more than one agent.
  • the secreted material may include proteins, carbohydrates, nucleic acids or lipids.
  • Secreted material may include the secretome, which is all of the secreted proteins and secretory machinery of the probiotic bacterium. It may additionally encompass molecules that are not proteins, such as carbohydrates, lipids and nucleic acid.
  • Some compositions described herein contain secreted material in a carrier.
  • the carrier is usually a solution in which the secreted material is dissolved, suspended, diluted or admixed.
  • the carrier may be the medium which has been in contact with the probiotic bacterium during culturing.
  • the composition of the medium will have changed during the culture, for example by the secretion of material from the probiotic bacterium.
  • the compositions may consist or comprise culture medium in which the probiotic bacteria have been growing in.
  • Media suitable for culturing probiotic bacteria is well known to those of skill in the art.
  • the terms "media” and “medium” encompasses any nutrient containing liquid in which microorganisms such as bacteria may be supported, kept alive, grown and/or expanded.
  • the media may contain the minimal nutrients to support bacterial life, and optionally other nutrients. Exemplary nutrients contained within the broth include sugar, magnesium, phosphate, phosphorous and sulphur.
  • the media may be made to, or modified from, a combination of nutrients that is well known in the art, such as Wilkins- Chalgren Broth.
  • Media may be obtained pre-mixed from a commercial source, or may be made in-house.
  • the probiotic bacterium may have been in contact with the media for at least six hours, at least twelve hours, at least eighteen hours, at least twenty four hours, at least 3 days, at least 4 days, at least 5 days, at least 6 days, at least 7 days, at least 8 days, at least 9 days, at least 10 days, at least two weeks or longer.
  • the probiotic bacteria may have been cultured in the media, or in contact with the media, under aerobic or anaerobic conditions.
  • the probiotic bacteria Preferably have been cultured under anaerobic conditions.
  • the culture may be performed under 10% H 2 , 10% C0 2 , 80% N 2 .
  • the probiotic bacteria may have been cultured in the media under conditions that facilitate growth and expansion of the probiotic bacteria. Such conditions are well known to those of skill in the art.
  • the culture may be incubated at 37°C.
  • the composition does not contain any probiotic bacteria.
  • the probiotic bacteria may have been removed from the media, for example by centrifugation and/or filtration.
  • the bacteria may be removed by sedimenting them from the media in a centrifuge at 15,000 x g for a period of time sufficient for substantially all of the bacteria to sediment from the media.
  • the media may be filtered using a microporous filter with pores of a suitable size to remove substantially all of the bacteria from the media. These methods may remove intact bacteria, and may also remove bacterial debris, such as the remains of any bacteria that have undergone cell lysis such as by apoptosis.
  • the media containing secreted material has not been obtained from a culture that has undergone a lysis process, and thus is not, and has not been obtained from, a lysate.
  • the composition may be sterile. That is to say that the secreted material has been subject to a sterilisation process, such as irradiation, heat, chemicals, pressure or filtration, or any combination thereof. This may include autoclaving, x-ray sterilization or UV-light sterilisation.
  • a sterilisation process such as irradiation, heat, chemicals, pressure or filtration, or any combination thereof. This may include autoclaving, x-ray sterilization or UV-light sterilisation.
  • the media may have been sterilised before the probiotic bacteria were introduced and cultured, and also after the bacteria had been removed from that media.
  • the composition comprising secreted material contains substantially no intact bacteria.
  • the composition may also be substantially free from lysed bacteria or bacterial fragments, such as bacteria that have undergone apoptosis.
  • the intact bacteria and/or lysed bacteria or bacterial fragments may have been separated from the secreted material. Separation may occur by any suitable means known in the art, such as centrifugation or filtration.
  • substantially free from we mean that the secreted material contains no or minimal contamination of non-secreted bacterial components, such as whole bacteria, lysed bacteria, or bacterial fragments.
  • the composition may contain 100% secreted material, at least 99% secreted material, at least 95% secreted material, at least 90% secreted material, at least 85% secreted material, at least 80% secreted material, at least 75% secreted material or at least 70% secreted material.
  • the secreted material may comprise additional components of non-bacterial origin, such as carrier solutions, other active agents, or preservatives, as described herein.
  • compositions as described herein may be prepared by cu!turing a probiotic bacteria in media, separating the probiotic bacteria from the media, and preparing a composition from the media.
  • the probiotic bacteria may be cultured under anaerobic conditions.
  • the probiotic bacteria may be cultured at a temperature above the normal temperature of the human body.
  • the probiotic bacteria may be cultured at 30°C, 31 °C, 32°C, 33°C, 34°C, 35°C, 36°C, 37°C, 38°C, 39°C, 40°C or 41 °C.
  • the probiotic bacteria are cultured at 37°C.
  • the probiotic bacteria may be cultured in the media for 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days or 14 days.
  • the probiotic bacteria or lysed bacteria or fragments of bacteria may be separated from the media by centrifugation, such as centrifugation at 5000 x g.
  • the media may be separated from the probiotic bacteria, lysed bacteria or fragments of bacteria by filtration.
  • the media may be separated by a combination of filtration and centrifugation.
  • the media may be subject to sterilisation, before or after the probiotic bacteria are removed. For example, following separation of the media from the whole bacteria, lysed bacteria or bacterial fragments, the media may be subject to sterilisation.
  • the media may be subject to concentration, such that the proportion of secreted material increases relative to the total volume of media. Concentration may occur by any method known in the art, such as evaporation.
  • Secreted material may be separated from the media. Any method of separating material from a carrier solution may be used. For example the secreted material may be separated from the media by chromatography, crystallisation, distillation, drying, electrophoresis or precipitation. Once isolated from the media, or concentrated in the media, the secreted material may be dissolved or diluted in a carrier, or otherwise formulated into a composition as disclosed herein.
  • the compounds and compositions of the present invention are useful in the treatment of a wide range of diseases and conditions. In particular they are useful in the treatment and prevention of skin infections, including bacterial infections. In particular, the compounds and compositions are useful in the treatment or prevention of S. aureus infections. The compounds and compositions are particularly useful in the treatment of soft tissue bacterial infections, such as skin infections. The compounds and compositions of the present invention are particularly useful in the prevention or treatment of S.aureus skin infections.
  • the invention relates to the prevention or treatment of infections.
  • the probiotic compositions of the present invention exhibit anti-infection activity.
  • anti- adhesion activity including preventing the adhesion of S.aureus to cells.
  • the compositions are useful for the prevention or treatment of infections including bacterial infections, such as the prevention or treatment of multi-drug resistant bacterial infections, hospital acquired bacterial infections, antibiotic resistant bacterial infections, infections by gram negative and/or gram positive bacterial infections.
  • the compositions of the invention are useful in the prevention of infections by
  • Staphylococcus spp. such as S. saprophytics, S.xylosus, S.lugdenensis, S.schleiferi, S.caprae, S.epidermidis, S. saprophytics, S.warneri, S.aureus, S.hominis, Methicilin resistant S.aureus (MRSA), S.pyrogenes, S.saiivariu, S.mutans and S. pneumonia.
  • MRSA Methicilin resistant S.aureus
  • the compositions of the invention exhibit anti-Staphylococcus adhesion activity, and are therefore useful in the prevention or treatment of Staphylococcus infection.
  • the compositions of the invention exhibit anti-Staphylococcus aureus activity, and are therefore useful in the prevention or treatment of S.aureus infections.
  • Infections occur where disease causing microorganisms invade the tissues of the body. Multiplication of those microorganisms and the toxins that they produce react with the tissues of the body, often causing immune reactions by the infected host. Infections may be caused by bacteria, viruses, viroids, fungi and other parasites. Infections may occur via any of the tissues of the body, such as the skin, gut or membranes.
  • the probiotic bacteria or lysates of the invention are used to treat infection of tissues other than the gut, for example in some embodiments the probiotic bacterium or lysate according to the invention is not used for the treatment of infection of the alimentary canal, esophagus, stomach, intestines, rectum or anus.
  • the invention relates to the treatment or prevention of infection of the external surface of the body, and particularly the skin.
  • the compositions according to the invention may be used in the prevention or treatment of skin infections.
  • the infection may be due to a bacterium, such as a Staphylococcus bacteria, including S. aureus.
  • the composition may be applied separately, sequentially or simultaneously with exposure to the infective agent. Preferably, the composition is applied before exposure to the infective agent.
  • compositions of the invention are preferably used for the prevention of bacterial infection. They are preferentially administered to a subject before that subject is exposed to the infective agent, such as S. aureus.
  • the subject may have been identified as being at risk of infection by the infective agent.
  • Subjects may be identified as being at risk of infection by an infective agent because of their environment, for example being situated in an environment where the inventive agent is known to exist, or due to the health of the subject, such as the existence of an open wound or poor immune health.
  • the compositions may be used in a hospital or other clinical environment in which a pathological bacteria is known to, or suspected to, be present.
  • the patient is about to undergo, or has recently undergone, surgery.
  • the compositions described herein may be used to prevent infection of an open wound such as a surgical incision or graft by a pathogenic bacteria.
  • the subject is determined not to have an infection by the infective agent.
  • the subject may be determined not to have a S. aureus infection.
  • Methods for determining whether a subject has an infection are well known in the art, and may include the analysis of a sample obtained from the subject for the presence of the infective agent.
  • a composition may be administered alone or in combination with other treatments, either simultaneously or sequentially dependent upon the condition to be treated.
  • the secreted material may be dissolved in, suspended in, or admixed with one or more other pharmaceutically acceptable ingredients.
  • the probiotic bacterium or lysate thereof may be presented in a liposome or other microparticulate.
  • the secreted material may be provided as a suspension in a pharmaceutically acceptable excipient, diluent or carrier.
  • probiotic bacterium may be provided as a lyophilisate.
  • the invention also provides antibacterial compositions in the form of cleaning products, washes, surface coatings or other compositions which are not for medical treatment of the human or animal body. Such agents may be useful for removing, killing, or preventing the accumulation of bacteria on a surface, or inhibiting the action or growth of the bacteria.
  • the secreted material is formulated as an antibacterial composition.
  • Anti-bacterial compositions according to the invention may be useful for treating biomaterials, implants and prosthesis (including stents, valves, eyes, hearing aids, gastric bands, dentures, artificial joint replacements etc), surgical instruments or other medical devices prior to administration to, or treatment of, or use with, a patient or subject.
  • the antibacterial compositions may be useful for treating surfaces prone to colonisation or exposure to bacterial, such as handrails, food preparation surfaces, kitchen surfaces or equipment, tables, sinks, toilets or other bathroom hardware,
  • Antibacterial compositions may comprise agents in addition to the lysate, such as cleaning agents, stabilisers, anionic surfactants, perfumes, chelating agents, acids, alkalis, buffers or detergents. Such agents may facilitate or enhance the antibacterial properties of the agent, such as killing or inhibiting bacteria, or preventing the
  • the present invention also gives rise to a method of preparing a surface comprising applying secreted material to the surface.
  • the method may result in reduced colonisation of the surface by pathogenic microorganisms.
  • the secreted material Whilst it is possible for the secreted material to be used alone, it is preferable to present it as a formulation comprising the material and a carrier.
  • the secreted material may be dissolved in, suspended in, or admixed with one or more other ingredients. In some cases the secreted material is presented in a liposome or other microparticulate.
  • Formulations disclosed herein include skin care, wound care, respiratory care and oral care formulations, including medical, personal care and consumer products.
  • Formulations may suitably be in the form of liquids, solutions (e.g., aqueous, non- aqueous), suspensions (e.g., aqueous, non-aqueous), emulsions (e.g., oil-in-water, water-in-oil), elixirs, syrups, electuaries, mouthwashes, drops, tablets (including, e.g., coated tablets), granules, powders, losenges, pastilles, capsules (including, e.g., hard and soft gelatin capsules), cachets, pills, ampoules, boluses, suppositories, pessaries, tinctures, gels, pastes, ointments, creams, lotions, oils, foams, sprays, mists, or aerosols.
  • solutions e.g., aqueous, non- aqueous
  • suspensions e.g., aqueous, non-aqueous
  • Formulations may suitably be provided as a patch, adhesive plaster, bandage, dressing, or the like which is impregnated with one or more active compounds and optionally one or more other pharmaceutically acceptable ingredients, including, for example, penetration, permeation, and absorption enhancers. Formulations may also suitably be provided in the form of a depot or reservoir.
  • the secreted material is formulated with one or more
  • Pharmaceutically acceptable ingredients are well known to those skilled in the art, and include, but are not limited to, pharmaceutically acceptable carriers, adjuvants, excipients, diluents, fillers, buffers, preservatives, antioxidants, lubricants, stabilisers, solubilisers, surfactants (e.g., wetting agents), masking agents, colouring agents, flavouring agents, and sweetening agents.
  • the formulation may further comprise other active agents, for example, other therapeutic or prophylactic agents.
  • Skin care means topical personal care and/or health care products including products useful for the treatment of adult or infant skin to maintain or improve the health of the skin or improve the appearance of the skin.
  • Wild care includes products for the treatment of a wound to assist in the closure or healing of the wound, and/or to reduce the pain or scarring associated with the wound, maintaining or improving the health of such tissue or skin, repairing such tissue or skin, and reducing irritation, itching and/or redness of such tissue or skin.
  • the secreted material according to the invention is formulated for topical administration, particularly for use or application to, or on, the skin.
  • Formulations suitable for topical administration include gels, pastes, ointments, creams, lotions, and oils, as well as patches, adhesive plasters, bandages, dressings, depots, cements, glues, and reservoirs.
  • Ointments are typically prepared from the secreted material and a paraffinic or a water- miscible ointment base.
  • Creams are typically prepared from the probiotic bacterium or lysate and an oil-in-water cream base.
  • the aqueous phase of the cream base may include, for example, at least about 30% w/w of a polyhydric alcohol, i.e., an alcohol having two or more hydroxyl groups such as propylene glycol, butane-1 ,3-diol, mannitol, sorbitol, glycerol and polyethylene glycol and mixtures thereof.
  • the topical formulations may desirably include a compound which enhances absorption or penetration of the active compound through the skin or other affected areas. Examples of such dermal penetration enhancers include dimethylsulfoxide and related analogues.
  • Emulsions are typically prepared from the probiotic bacterium or lysate and an oily phase, which may optionally comprise merely an emulsifier (otherwise known as an emulgent), or it may comprises a mixture of at least one emulsifier with a fat or an oil or with both a fat and an oil.
  • an emulsifier also known as an emulgent
  • a hydrophilic emulsifier is included together with a lipophilic emulsifier which acts as a stabiliser. It is also preferred to include both an oil and a fat.
  • the emulsifier(s) with or without stabiliser(s) make up the so-called emulsifying wax
  • the wax together with the oil and/or fat make up the so-called emulsifying ointment base which forms the oily dispersed phase of the cream formulations.
  • Suitable emulgents and emulsion stabilisers include Tween 60, Span 80, cetostearyl alcohol, myristyl alcohol, glyceryl monostearate and sodium lauryl sulphate.
  • the choice of suitable oils or fats for the formulation is based on achieving the desired cosmetic properties, since the solubility of the active compound in most oils likely to be used in pharmaceutical emulsion formulations may be very low.
  • the cream should preferably be a non-greasy, non-staining and washable product with suitable consistency to avoid leakage from tubes or other containers.
  • Straight or branched chain, mono- or dibasic alkyl esters such as di-isoadipate, isocetyl stearate, propylene glycol diester of coconut fatty acids, isopropyl myristate, decyl oleate, isopropyl palmitate, butyl stearate, 2-ethylhexyl palmitate or a blend of branched chain esters known as Crodamol CAP may be used, the last three being preferred esters. These may be used alone or in combination depending on the properties required. Alternatively, high melting point lipids such as white soft paraffin and/or liquid paraffin or other mineral oils can be used.
  • Oral care means products for use and/or uses of materials in the oral cavity or any portion thereof, including products for use on the teeth, mucosa, tongue, and the like.
  • Products and uses in the field of oral care include those intended for tooth aesthetics including, for example, tooth whitening, stain prevention, and the like, as well as anti-plaque, anti-gingivitis, anti- sensitivity, anti-caries, breath freshening, dry mouth relief, erosion repair and prevention, active delivery and retention, sensory enhancement and mouth feel alteration, and the like.
  • Formulations for oral care include dental sprays, mouthwashes, toothpastes, lozenges, antibacterial washes, drinks (e.g. milk, yoghurt), food items (such as yoghurt, ice cream, candy bars), or powdered foods (such as powdered milk).
  • Formulations suitable for oral care include formulations suitable for oral and/or buccal administration.
  • Formulations suitable for oral administration include liquids, solutions (e.g., aqueous, non-aqueous), suspensions (e.g., aqueous, non-aqueous), emulsions (e.g., oil-in-water, water-in-oil), elixirs, syrups, electuaries, tablets, granules, powders, capsules, cachets, pills, ampoules, boluses.
  • Formulations suitable for buccal administration include mouthwashes, losenges, pastilles, as well as patches, adhesive plasters, depots, and reservoirs.
  • Losenges typically comprise the active compound in a flavored basis, usually sucrose and acacia or tragacanth.
  • Pastilles typically comprise the active compound in an inert matrix, such as gelatin and glycerin, or sucrose and acacia.
  • Mouthwashes typically comprise the active compound in a suitable liquid carrier.
  • formulations disclosed herein are suitably provided as a patch, adhesive plaster, bandage, dressing, or the like which is impregnated with, or coated with, one or more secreted material according to the invention and optionally one or more other
  • probiotic bacteria, lysates or culture media may also be provided in the form of coatings for medical devices such as implants, prosthetics, surgical instruments, gloves, catheters, valves, pacemakers and the like.
  • compositions and formulations disclosed herein are suitable for respiratory care.
  • “Respiratory care” means products for the treatment of conditions including prevention and treatment of rhinitis, sinusitis, seasonal allergies, nasal congestion and colds.
  • the compositions may be useful for preventing a bacterial infection of the respiratory tract, including the sinuses, airways, throat or lungs.
  • such formulations are formulated for intranasal administration or pulmonary administration.
  • Formulations suitable for intranasal administration, where the carrier is a liquid include, for example, nasal spray, nasal drops, or by aerosol administration by nebuliser, include aqueous or oily solutions of the active compound.
  • Formulations suitable for intranasal administration include, for example, those presented as a coarse powder having a particle size, for example, in the range of about 20 to about 500 microns which is administered in the manner in which snuff is taken, i.e., by rapid inhalation through the nasal passage from a container of the powder held close up to the nose.
  • Formulations suitable for pulmonary administration include those presented as an aerosol spray from a pressurised pack, with the use of a suitable propellant, such as dichlorodifluoromethane, trichlorofluoromethane, dichoro-tetrafluoroethane, carbon dioxide, or other suitable gases.
  • Compositions and formulations according to the invention may further comprise other active agents, for example other anti-bacterial agents such as bactericidal agents.
  • a formulation for use according to the present invention may comprise at least about 0.01 %, about 0.05%, about 0.1 %, about 0.2%, about 0.3%, about 0.4%, about 0.5%, about 0.6%, about 0.7%, about 0.8%, about 0.9%, about 1.0%, about 1.5%, about 2.0%, about 3.0%, about 4.0%, about 5.0%, about 6.0%, about 7.0%, about 8.0%, about 9.0%, about 1 0.0%, about 11.0%, about 12.0%, about 13.0%, about 14.0%, about 15.0%, about 16.0%, about 17.0%, about 18.0%, about 19.0%, about 20.0%, about 25.0%, about 30.0%, about 35.0%, about 40.0%, about 45.0%, about 50.0% by weight of secreted material.
  • the formulation may comprise, one of at least about 0.01% to about 30%, about 0.01 % to about 20%, about 0.01 % to about 5%, about 0.1% to about 30%, about 0.1 % to about 20%, about 0.1% to about 15%, about 0.1% to about 10%, about 0.1 % to about 5%, about 0.2% to about 5%, about 0.3% to about 5%, about 0.4% to about 5%, about 0.5% to about 5%, about 1% to 10 about 5%, by weight of secreted material.
  • probiotic preparations according to the invention may be formulated as
  • compositions for clinical use may comprise a pharmaceutically acceptable carrier, diluent or adjuvant. They may be formulated for topical administration.
  • Administration is preferably in a prophylactically or therapeutically effective amount, this being an amount sufficient to show benefit to the individual.
  • the actual amount administered, and rate and time-course of administration will depend on the nature and severity of the disease being treated. Prescription of treatment, e.g. decisions on dosage etc., is within the responsibility of general practitioners and other medical doctors, and typically takes account of the disorder to be treated or prevented, the condition of the individual patient, the site of delivery, the method of administration and other factors known to practitioners. Examples of the techniques and protocols mentioned above can be found in Remington's Pharmaceutical Sciences, 20 th Edition, 2000, pub. Lippincott, Williams & Wilkins. It will be appreciated by one of skill in the art that appropriate dosages of the active compounds and compositions comprising the active compounds can vary from patient to patient.
  • compositions of the present invention may be formulated as medicaments, that is to say formulated as a medicine.
  • the medicament may include other pharmaceutically acceptable ingredients well known to those skilled in the art, including , but not limited to, pharmaceutically acceptable carriers, adjuvants, excipients, diluents, fillers, buffers, preservatives, anti-oxidants, lubricants, stabilisers, solubilisers, surfactants (e.g. wetting agents), masking agents, colouring agents, flavouring agents, and sweetening agents.
  • the formulation may further comprise other active agents, for example other therapeutic or prophylactic agents.
  • FIG. 1 Lrhamnosus GG protects keratinocytes from the toxic effects of S.
  • FIG. 2 Lysate and spent culture fluid (CM) from Lrhamnosus GG protect keratinocytes from the effects of S.aureus.
  • Figure 3 Lrhamnosus GG protects keratinocytes from infection with S. aureus.
  • FIG. 4 L.rhamnosus GG but not its spent culture fluid rescuesd keratinocytes from S. aureus mediated toxicity.
  • FIG. 6 Live Lrhamnosus GG, lysate or spent culture fluid inhibited S. aureus from adhering to keratinocytes by competitive exclusion from binding sites.
  • SA S. aureus
  • FIG. 7 Live Lrhamnosus GG or lysate inhibited S. aureus from adhering to keratinocytes by competitive displacement to binding sites.
  • SA S. aureus
  • NHEK normal human epidermal keratinocytes
  • keratinocyte basal medium Promocell, Heidelberg, Germany
  • a supplement mix bovine pituitary extract 0.004mg/ml
  • epidermal growth factor recombinant human
  • Lactobacillus rhamnosus Goldin and Gorbach (Lrhamnosus GG) (ATCC 53103.ATCC, Middlesex, UK ) was grown routinely in Wilkins-Chalgren Broth or Agar (Oxoid,
  • Bacteria (10 8 CFU/ml of probiotics and10 6 CFU/ml of S. aureus) were centrifuged at 15,000 x g, washed twice in 0.85% NaCI and resuspended in keratinocyte basal medium. This suspension was added directly to 5 x 10 3 cells/cm 2 of NHEK growing in 24 well plates.
  • keratinocytes 5 x 10 3 cells/cm 2 .
  • cells were sedimented in a centrifuge at 15,000 x g for 5 minutes and the cell-free supernatant (spent culture fluid) collected and filtered using a 0.22pm pore filter (Millipore, Billerica, USA) to remove any whole bacteria remaining.
  • keratinocytes monolayers were co-infected with pathogen plus probiotics or lysates simultaneously.
  • cells were exposed to Lrhamnosus GG lysate 2, 4, 6, 8 and 12 hours after S. aureus infection had consumed.
  • keratinocytes were detached and cell viability was determined using trypan blue exclusion assays as described in (43).
  • Lrhamnosus GG lysates 2, 4, 6, 8 and 12hours post infection with S. aureus. The total number of viable staphylococci was determined by counting the colonies as described previously (43).
  • PBS phosphate buffered saline
  • Lrhamnosus GG protects keratinocytes from the pathogenic effects of S. aureus.
  • the ability of Lrhamnosus GG to protect keratinocytes from the effects of S. aureus was investigated. In agreement with our previous findings (43) 24h exposure of keratinocytes to 10 6 CFU/ml S. aureus resulted in significant keratinocyte cell death.
  • Lrhamnosus GG lysates and spent culture fluid protect keratinocytes from the effects of S. aureus.
  • Lrhamnosus GG lysate but not spent culture fluid rescues keratinocytes from S. aureus toxicity.
  • L.rhamnosus GG inhibits adhesion of S. aureus to keratinocytes.
  • CFCS cell-free culture supernatants
  • Keratinocyte protection by the lactobacillus lysate may involve at least two mechanisms. Firstly, the lysate may be able to reduce the growth of S aureus. Competition assays demonstrated that Lrhamnosus GG lysate reduced the total number of viable
  • Lrhamnosus GG contains bacteriostatic substances, then this may also, at least partially explain the protective effect of the probiotic in keratinocyte survival assays.
  • Probiotics, especially lactobacilli have previously been shown to exert a strong inhibitory effect on S. aureus growth.
  • Certain Lactobacillus strains have been reported to be highly antagonistic to biofilm-forming S. aureus (28, 30).
  • Other studies have reported that probiotics can improve gut health by inhibiting growth of pathogens through production of bacteriocins (16, 48).
  • Lrhamnosus GG has been shown to inhibit the growth of Salmonella enterica through production of lactic acid (29).
  • a second mechanism by which live bacterium or lysate of Lrhamnosus GG could protect the keratinocytes is by inhibition of pathogenic adhesion. Indeed, our data demonstrated a reduction in adhesion of S. aureus to keratinocytes in the presence of Lrhamnosus GG or its lysate. This data suggests a mechanism of exclusion as we have observed previously for L. reuteri (43). However, interestingly, viable L .rhamnosus GG or its lysate also inhibited adhesion of S. aureus when added to existing infections demonstrating another mechanism of protection i.e. that Lrhamnosus GG can displace pathogen from keratinocytes (Figure7).
  • Lrhamnosus GG has been shown to displace pathogens from the intestinal cells in the gut (47).
  • our data demonstrated that the presence of live bacterium is not necessary for displacement of S. aureus from keratinocytes.
  • our data demonstrated species dependent differences in the mechanisms used by lactobacilli to reduce pathogen toxicity.
  • Our previous work highlighted L reuteri as an organism capable of excluding S. aureus form keratinocyte binding sites (43).
  • Lrhamnosus GG can, not only, exclude pathogens but can also reduce pathogen growth and displace pathogen from keratinocytes.
  • this displacement activity may be related to the ability of Lrhamnosus GG to inhibit growth and further studies will be required to clarify this point.
  • Lrhamnosus GG is a potential new agent to inhibit the pathogenicity of S. aureus. Furthermore, our data shows that the utility of L .rhamnosus GG on skin will not be limited by whether it can grow and survive on skin because a lysate of the organisms is just as efficacious at preventing S. aureus colonization as live bacteria. Furthermore, the lysate could be useful as prophylaxis e.g. in hand washes, but potentially as an adjunct or even an alternative to antibiotics in existing infection.
  • Lactobacillus crispatus strain ZJ001 is responsible for competitive exclusion against Escherichia co// 0157:H7 and Salmonella typhimurium. Int. J. Food Microbiol. 115:307-312.
  • Lactobacilli Adv. Appl. Microbiol. 70:127-152. Di Marzio, Cinque LB, De Simone C, Cifone MG. 1999. Effect of the Lactic Acid Bacterium Streptococcus thermophilus on Ceramide Levels in Human
  • Staphylococcus aureus Epidemiology, underlying mechanisms, and associated risks. Clin. Microbiol. Rev. 10:505-520.
  • Typhimurium reveals a role for lactic acid and other inhibitory compounds.
  • MapA Mediates the Adhesion of Lactobacillus reuteri to Caco-2 Human Intestinal Epithelial Cells. Biosci. Biotechnol. Biochem. 70:1622-1628.
  • Lactobacillus reuteri in bovine milk fermented decreases the oral carriage of mutans streptococci. Int. J. Food Microbiol. 95:219-223.
PCT/GB2015/051529 2014-05-29 2015-05-26 Anti-bacterial lysate of probiotic bacteria WO2015181534A1 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
ES15727045T ES2842215T3 (es) 2014-05-29 2015-05-26 Lisado antibacteriano de bacterias probióticas
JP2016567543A JP6741595B2 (ja) 2014-05-29 2015-05-26 プロバイオティック細菌の抗菌性溶解物
CA2950510A CA2950510C (en) 2014-05-29 2015-05-26 Anti-bacterial lysate of probiotic bacteria
EP15727045.5A EP3148558B1 (en) 2014-05-29 2015-05-26 Anti-bacterial lysate of probiotic bacteria
CN201580028700.4A CN106794205B (zh) 2014-05-29 2015-05-26 益生菌的抗菌裂解物
AU2015265698A AU2015265698B2 (en) 2014-05-29 2015-05-26 Anti-bacterial lysate of probiotic bacteria
US15/314,897 US10702562B2 (en) 2014-05-29 2015-05-26 Anti-bacterial lysate of probiotic bacteria

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GB1409541.8 2014-05-29
GBGB1409541.8A GB201409541D0 (en) 2014-05-29 2014-05-29 Probiotic Bacteria

Publications (1)

Publication Number Publication Date
WO2015181534A1 true WO2015181534A1 (en) 2015-12-03

Family

ID=51214406

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2015/051529 WO2015181534A1 (en) 2014-05-29 2015-05-26 Anti-bacterial lysate of probiotic bacteria

Country Status (9)

Country Link
US (1) US10702562B2 (zh-TW)
EP (1) EP3148558B1 (zh-TW)
JP (1) JP6741595B2 (zh-TW)
CN (1) CN106794205B (zh-TW)
AU (1) AU2015265698B2 (zh-TW)
CA (1) CA2950510C (zh-TW)
ES (1) ES2842215T3 (zh-TW)
GB (1) GB201409541D0 (zh-TW)
WO (1) WO2015181534A1 (zh-TW)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017199022A2 (en) 2016-05-18 2017-11-23 Skinbiotherapeutics Plc Compositions and uses thereof
WO2018209457A1 (es) * 2017-05-17 2018-11-22 Universidad San Sebastián Aerosol orofaríngeo compuesto por un hidrolizado derivado de un probiótico que tiene impacto en la inmunidad del tracto genitourinario, método de obtención del hidrolizado, composiciones farmacéuticas que lo contienen y uso de las mismas
WO2019215446A1 (en) 2018-05-09 2019-11-14 Skinbiotherapeutics Plc Compositions and uses thereof
US10702562B2 (en) 2014-05-29 2020-07-07 Skinbiotherapeutics Plc Anti-bacterial lysate of probiotic bacteria
WO2022195109A1 (en) * 2021-03-19 2022-09-22 Eligo Bioscience Therapeutic use of engineered postbiotics comprising bacteriocins and/or endolysins
WO2022195108A3 (en) * 2021-03-19 2022-11-10 Eligo Bioscience Cosmetic use of engineered postbiotics comprising bacteriocins and/or endolysins
WO2023035633A1 (zh) * 2021-09-07 2023-03-16 青岛蔚蓝生物股份有限公司 一株对皮肤损伤具有保护作用的鼠李糖乳杆菌及其应用

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB2544481B (en) * 2015-11-16 2019-12-11 Century International Enterprises Ltd A wearable article and a method for producing a wearable article
CN112957404A (zh) * 2021-02-08 2021-06-15 广东广汇农牧有限公司 一种用于母猪阴道护理的环保型清洗液组合物及其制备方法

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009155711A1 (en) * 2008-06-27 2009-12-30 University Of Guelph Analysis of lactobacillus acidophilus la-5 signal interference molecules
WO2013153358A1 (en) * 2012-04-13 2013-10-17 The University Of Manchester Probiotic bacteria

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MXPA06015184A (es) * 2004-06-23 2007-11-22 Nestec Sa Procedimiento y composiciones utiles para prevenir y/o tratar pieles sensibles y/o secas.
US20110039765A1 (en) * 2008-04-17 2011-02-17 Connor Ruth I Bioactive Molecules Produced by Probiotic Bacteria
DK2704704T3 (en) * 2011-05-03 2019-01-14 Dupont Nutrition Biosci Aps PROBIOTIC BACTERIES FOR TOPICAL TREATMENT OF SKIN DISORDERS
GB201409541D0 (en) 2014-05-29 2014-07-16 Univ Manchester Probiotic Bacteria

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009155711A1 (en) * 2008-06-27 2009-12-30 University Of Guelph Analysis of lactobacillus acidophilus la-5 signal interference molecules
WO2013153358A1 (en) * 2012-04-13 2013-10-17 The University Of Manchester Probiotic bacteria

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
DORON S ET AL: "Probiotics: Their role in the treatment and prevention of disease", EXPERT REVIEW OF ANTI-INFECTIVE THERAPY, FUTURE DRUGS, LONDON, GB, vol. 4, no. 2, 1 April 2006 (2006-04-01), pages 261 - 275, XP009107657, ISSN: 1478-7210, DOI: 10.1586/14787210.4.2.261 *
LU R ET AL: "M1208 Six Small Bioactive Peptides Identified from Lactobacillus GG Cultured Supernatant", GASTROENTEROLOGY, ELSEVIER, PHILADELPHIA, PA, vol. 134, no. 4, 1 April 2008 (2008-04-01), pages A - 361, XP023433480, ISSN: 0016-5085, [retrieved on 20080401], DOI: 10.1016/S0016-5085(08)61683-2 *
SIGRID C.J. DE KEERSMAECKER ET AL: "Strong antimicrobial activity of Lactobacillus rhamnosus GG against Salmonella typhimurium is due to accumulation of lactic acid", FEMS MICROBIOLOGY LETTERS, vol. 259, no. 1, 1 June 2006 (2006-06-01), pages 89 - 96, XP055088376, ISSN: 0378-1097, DOI: 10.1111/j.1574-6968.2006.00250.x *

Cited By (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10702562B2 (en) 2014-05-29 2020-07-07 Skinbiotherapeutics Plc Anti-bacterial lysate of probiotic bacteria
WO2017199022A2 (en) 2016-05-18 2017-11-23 Skinbiotherapeutics Plc Compositions and uses thereof
WO2017199022A3 (en) * 2016-05-18 2018-02-22 Skinbiotherapeutics Plc Lactobacillus rhamnosus lysate compositions and uses thereof
JP2019523219A (ja) * 2016-05-18 2019-08-22 スキンバイオセラピューティクス ピーエルシー 組成物およびその使用
JP7189024B2 (ja) 2016-05-18 2022-12-13 スキンバイオセラピューティクス ピーエルシー 組成物およびその使用
WO2018209457A1 (es) * 2017-05-17 2018-11-22 Universidad San Sebastián Aerosol orofaríngeo compuesto por un hidrolizado derivado de un probiótico que tiene impacto en la inmunidad del tracto genitourinario, método de obtención del hidrolizado, composiciones farmacéuticas que lo contienen y uso de las mismas
WO2019215446A1 (en) 2018-05-09 2019-11-14 Skinbiotherapeutics Plc Compositions and uses thereof
US11759484B2 (en) 2018-05-09 2023-09-19 Skinbiotherapeutics Plc Compositions and uses thereof
WO2022195112A1 (en) * 2021-03-19 2022-09-22 Eligo Bioscience Therapeutic use of engineered postbiotics comprising bacteriocins and/or endolysins for treating acneiform rash
WO2022195118A1 (en) * 2021-03-19 2022-09-22 Eligo Bioscience Therapeutic use of engineered postbiotics comprising bacteriocins and/or endolysins for treating cutaneous lymphoma
WO2022195108A3 (en) * 2021-03-19 2022-11-10 Eligo Bioscience Cosmetic use of engineered postbiotics comprising bacteriocins and/or endolysins
WO2022195115A1 (en) * 2021-03-19 2022-09-22 Eligo Bioscience Therapeutic use of engineered postbiotics comprising bacteriocins and/or endolysins for treating subacute cutaneous lupus erythematosus
US11541106B2 (en) 2021-03-19 2023-01-03 Eligo Bioscience Method for killing bacteria with an engineered postbiotic
US11633348B2 (en) 2021-03-19 2023-04-25 Eligo Bioscience Cosmetic use of engineered postbiotics comprising bacteriocins and/or endolysins
WO2022195109A1 (en) * 2021-03-19 2022-09-22 Eligo Bioscience Therapeutic use of engineered postbiotics comprising bacteriocins and/or endolysins
US11839645B2 (en) 2021-03-19 2023-12-12 Eligo Bioscience Therapeutic use of engineered postbiotics comprising bacteriocins and/or endolysins
WO2023035633A1 (zh) * 2021-09-07 2023-03-16 青岛蔚蓝生物股份有限公司 一株对皮肤损伤具有保护作用的鼠李糖乳杆菌及其应用

Also Published As

Publication number Publication date
US10702562B2 (en) 2020-07-07
AU2015265698B2 (en) 2020-10-22
CA2950510C (en) 2023-10-24
EP3148558B1 (en) 2020-10-28
GB201409541D0 (en) 2014-07-16
EP3148558A1 (en) 2017-04-05
ES2842215T3 (es) 2021-07-13
CN106794205A (zh) 2017-05-31
AU2015265698A1 (en) 2016-12-22
US20170196919A1 (en) 2017-07-13
CA2950510A1 (en) 2015-12-03
CN106794205B (zh) 2021-10-01
JP6741595B2 (ja) 2020-08-19
JP2017519730A (ja) 2017-07-20

Similar Documents

Publication Publication Date Title
AU2015265698B2 (en) Anti-bacterial lysate of probiotic bacteria
US20230088050A1 (en) Probiotic Bacteria
JP7308875B2 (ja) 口腔組成物
US20080107699A1 (en) Method of using topical probiotics for the inhibition of surface contamination by a pathogenic microorganism and composition therefor
JP7189024B2 (ja) 組成物およびその使用
US20070238652A1 (en) Neutralization of bacterial spores
WO2021154954A1 (en) Compositions and methods for treating biofilms, infections and periodontitis

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 15727045

Country of ref document: EP

Kind code of ref document: A1

REEP Request for entry into the european phase

Ref document number: 2015727045

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2015727045

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2016567543

Country of ref document: JP

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2950510

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 15314897

Country of ref document: US

ENP Entry into the national phase

Ref document number: 2015265698

Country of ref document: AU

Date of ref document: 20150526

Kind code of ref document: A