EP3697414A1 - Verwendung von cloxacillin zur verhinderung/hemmung von biofilmbildung - Google Patents

Verwendung von cloxacillin zur verhinderung/hemmung von biofilmbildung

Info

Publication number
EP3697414A1
EP3697414A1 EP18762363.2A EP18762363A EP3697414A1 EP 3697414 A1 EP3697414 A1 EP 3697414A1 EP 18762363 A EP18762363 A EP 18762363A EP 3697414 A1 EP3697414 A1 EP 3697414A1
Authority
EP
European Patent Office
Prior art keywords
cloxacillin
biofilm
formation
concentration
bacterial
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP18762363.2A
Other languages
English (en)
French (fr)
Inventor
Christian Provot
Thierry Bernardi
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Biofilm Control SAS
Original Assignee
Biofilm Control SAS
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 Biofilm Control SAS filed Critical Biofilm Control SAS
Publication of EP3697414A1 publication Critical patent/EP3697414A1/de
Withdrawn legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/429Thiazoles condensed with heterocyclic ring systems
    • A61K31/43Compounds containing 4-thia-1-azabicyclo [3.2.0] heptane ring systems, i.e. compounds containing a ring system of the formula, e.g. penicillins, penems
    • A61K31/431Compounds containing 4-thia-1-azabicyclo [3.2.0] heptane ring systems, i.e. compounds containing a ring system of the formula, e.g. penicillins, penems containing further heterocyclic rings, e.g. ticarcillin, azlocillin, oxacillin
    • 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

  • the present invention relates to a pharmaceutical composition comprising cloxacillin for use as a medicament for preventing the formation of a bacterial biofilm.
  • the present invention also relates to the use of cloxacillin for the prevention and / or inhibition of the formation of a bacterial biofilm on a surface.
  • the present invention also relates to a medical device comprising on its surface cloxacillin.
  • the present invention finds application particularly in the pharmaceutical field, the medical field.
  • Biofilms are formed of different layers of bacteria or microorganisms, often contained in a solid matrix. They develop to form microbial communities, one of whose properties is to adhere to submerged surfaces. This adhesion is either nonspecific (adherence) or specific (adhesion itself) (Costerton et al., "Bacterial Biofilms: a common cause of persistent infections.” Science 1999; 284, 1318-1322):
  • - Adhesion or reversible adhesion The microorganisms present approach the surfaces by gravimetry, brownian movements or chemotaxis if they have flagella. During this first fixation step, involving only purely physical phenomena and weak physicochemical interactions, the microorganisms can still be easily unhooked.
  • - Adhesion This slower stage involves interactions of higher energy as well as microbial metabolism and cellular appendages of the microorganism (flagella, pilis, .). Membership is an active and specific phenomenon.
  • Some early colonizers will attach irreversibly to the surface thanks to the synthesis of exopolysaccharides (EPS). This process is relatively slow and depends on environmental factors and microorganisms. These biofilms are ubiquitous in many areas, where they pose health risks and cause relatively significant damage.
  • EPS exopolysaccharides
  • biofilms are responsible for infections that are increasingly difficult to control: over the ENT sphere (auditory canal, nasal mucosa, conjunctive eye %), on the teeth (appearance of tartar , caries, ...), on the bronchi, lungs (in patients with pneumopathies, cystic fibrosis %), in the urogenital tract, wounds.
  • Bacterial biofilms that develop on implants or during chronic infections are reservoirs of pathogens that cause many nosocomial infections.
  • biofilms are difficult to prevent and eradicate because of their characteristic tolerance to therapeutic doses of prescribed antibiotics, and even to high doses of antibiotics. This tolerance allows microorganisms to persist (hence the name of persisters assigned to them) and then to develop resistances within the biofilm (appearance of mutations, exchange of resistance genes) (H0iby, N. et al .; (2010) Antibiotic resistance of bacterial biofilms. International Journal of Antimicrobial Agents, 35, 322-332).
  • Osteoarticular infections are infections that affect the bones and / or joints that are frequently caused by staphylococci. There are also complex osteoarticular infections that appear more particularly in individuals with joint prosthesis or implantable medical devices. These infections are particularly difficult to treat in that they frequently have a tolerance (also called recalcitrance) to antibiotics, including the presence of biofilm, as well as resistances as described above.
  • the surface of prostheses, including articular is a favorable territory for the proliferation of bacteria and the formation of biofilm.
  • an antibiotic at a dose lower than its MIC (subCMI) can promote the attachment of bacteria.
  • aminoglycosides at subCMI doses induce adhesion of strains of Pseudomonas aeruginosa and Escherichia coli (Hoffman et al., 2005, Nature, 436: 1,171-1,175), metronidazole increases biofilm formation by Clostridium difficile (Vuotto et al, 2016, Pathod Dis, 74, doi: 10.1093 / femspd / ftv1 14), arnoxicillin at subCMI doses induces attachment of S. aureus strain USA 300 (Mlynek et al, 2016,
  • the present invention solves the disadvantages and obstacles of the prior art via the use of cloxacillin at a concentration greater than 4 mg / l, for example from 4 mg / l to 16 mg / l.
  • the present invention also solves the disadvantages and obstacles of the prior art via the use of cloxacillin at a concentration greater than 25 mg / kg, for example from 25 to 100 mg / kg, preferably from 25 to 50 mg / kg.
  • the inventors have surprisingly and unexpectedly demonstrated that use at a concentration greater than 4 mg / l, for example ranging from 4 mg / l to 16 mg / l, advantageously makes it possible to inhibit the formation and / or development of biofilms on a area.
  • the present invention also solves the disadvantages and obstacles of the prior art by using cloxacillin at a dose greater than 25 mg / kg, for example from 25 to 100 mg / kg, preferably from 25 to 50 mg / kg as a medicament thus allowing inhibit the formation and / or development of biofilms on a surface.
  • cloxacillin as a medicament at a dose greater than 25 mg / kg, for example ranging from 25 to 100 mg / kg, preferably from 25 to 50 mg / kg, advantageously makes it possible to inhibit training and / or development of biofilms.
  • per mg / kg is meant a dose in milligrams per kilogram of the individual to be treated.
  • the present invention also solves the disadvantages and obstacles of the prior art by providing a pharmaceutical composition comprising cloxacillin at a dose greater than 4 mg / l, for example ranging from 4 mg / l to 16 mg / l. for use as a medicament for preventing and / or inhibiting the formation of a bacterial biofilm.
  • the inventors are the first to have surprisingly demonstrated inhibition of biofilm formation by cloxacillin.
  • the inventors are the first to have surprisingly demonstrated inhibition of biofilm formation by cloxacillin at a concentration / amount used which is totally different and independent of the concentration / amount known and / or useful for its use as as bactericidal or "killer of bacteria".
  • the inventors have notably demonstrated an inhibition of the biofilm formation under in vitro conditions used comprising in a short incubation (4h) of clinical strains of Staphylococcus aureus, in the presence of cloxacillin from the beginning of the incubation.
  • the inventors demonstrated and characterized the effect of cloxacillin on initial biofilm formation, adhesion and adherence steps.
  • a dose corresponding to the Minimum Inhibitory Concentration (MIC) the inventors have demonstrated that above a particular concentration and or in a particular concentration range, cloxacillin inhibits the formation of biofilms.
  • the minimum concentration to inhibit biofilm formation is also referred to in the text as Minimum Biofilm Inhibitory Concentration (MICb).
  • MICb Minimum Biofilm Inhibitory Concentration
  • the inventors have also demonstrated that the MICI may vary, like the MIC, depending on the strains, however, they have also demonstrated that the variation may be within a range of values advantageously allowing the use of cloxacillin to inhibit the formation of biofilm , without need for characterization, for example determination of the MIC and / or MICB, of the strain, for example by using a concentration and / or dose corresponding to a frequently found MIC.
  • Cloxacillin is an antibiotic of the ⁇ -lactam family of the group M penicillins. Its action is bactericidal, that is to say, it will kill bacteria, and it is used in this way.
  • Cloxacillin is a lactam of the following formula (I):
  • cloxacillin is used against Streptococcus pyogenes, a Gram + aerobic bacterium that is usually susceptible, and against other Gram + aerobic bacteria such as Staphylococcus aureus and coagulase negative staphylococci, but some staphylococci are now resistant.
  • cloxacillin is used orally in the management of staphylococcal and / or streptococcal skin infections. Injectable, cloxacillin is used in the management of pathologies largely associated with a staphylococcal infection such as: osteoarticular infections, endocarditis, respiratory infections, urogenital infections. Staphylococcal dermatologic infections can also be treated by injection.
  • Cloxacillin is also known in a galenic form of capsule, for example marketed under the trademark ORBENINE 500 mg capsule (cloxacillin), and is indicated in adults and children in the treatment of mild skin infections due to staphylococci and / or susceptible streptococci.
  • cloxacillin is used and described as a bactericidal antibiotic, that is, a "killer molecule of bacteria".
  • cloxacillin Panpharma 500mg is intended to be administered, after solubilization, intravenously, in curative or preventive treatment.
  • the dosage is, for an adult without kidney or liver problems, 8 to 12 g / day, divided into 4 to 6 administrations daily.
  • prophylactic antibiotic prevention of postoperative infections in surgery is generally 2g to anesthesia, continued by injections of 1g every 2h.
  • cloxacillin is known and used as a killer of bacteria in the treatment of osteoarticular infections, particularly when the causative organism identified is a strain of methicillin-sensitive Staphylococcus aureus.
  • antibiotic therapy seems adapted because of the sensitivity of the strain, (determined by tests of sensitivity to conventional antibiotics, such as Etest, Vitek® or Vitek® 2 from bioMérieux, antibiogram), a failure rate therapeutic is not negligible, namely more than 20% failure (Valor et al., BMC Infect Dis. 2014 Aug 16; 14:44)).
  • Biofilm herein means any biofilm known to those skilled in the art formed by a microorganism, for example it may be a bacterial biofilm.
  • surface is meant any surface known to those skilled in the art on which a biofilm can be formed. It may be for example a biotic or abiotic surface.
  • biotic surface any biotic surface known to those skilled in the art. It can be for example a biotic surface selected from the group consisting of skin, mucous membranes, teeth, any surface of the vascular system, for example blood vessels, walls of the endocardium, any surface of the lymphatic system, any surface of the digestive system, any surface of the respiratory system, for example the walls of the bronchopulmonary system, any surface of the Oto-Rhino-Laryngeal sphere.
  • the biotic surface may be a healthy surface and / or exhibiting at least one lesion and / or infection. It may be for example a biotic surface comprising a wound, an infected wound, a wound in the process of healing, a sutured wound, a scar, a burn.
  • wound any wound known to those skilled in the art. It may be for example a superficial wound, for example wound of the epidermis and / or dermis, for example less than 5 cm in size. It can also be deep wound, for example a wound of the epidermis and dermis with exposure of the subcutaneous tissue. It can also be burns, bedsores. It can also be any wounds susceptible to infection, for example any wounds likely to be infected, the infection delaying or compromising the healing of the wound.
  • infection any infection known to those skilled in the art for which biofilms are likely to form.
  • infections selected from the group consisting of osteoarticular infections, diabetic foot infections, skin wound infections such as burns and bedsores, infections of the respiratory system, for example bronchopulmonary system, infections of the Oto-Rhino-Laryngeal sphere, infections of the cardiovascular system, infections of the digestive system.
  • abiotic surface is meant any abiotic surface known to those skilled in the art. It may also be any device surface known to those skilled in the art on which a bacterium is capable of forming a biofilm.
  • a medical device for example a catheter, a needle, a catheter, implantable chamber (or Porth-a-Cath), a valve, for example a heart valve, a prosthesis, for example a joint prosthesis, a ligament prosthesis, a dental implant, a urinary tract prosthesis, the peritoneal membrane, catheters of peritoneal dialysis, synthetic vascular grafts, stents, internal fixation devices, percutaneous and / or tracheal sutures, ventilation tubes.
  • a medical device for example a catheter, a needle, a catheter, implantable chamber (or Porth-a-Cath), a valve, for example a heart valve, a prosthesis, for example a joint prosthesis, a ligament prosthesis, a dental implant, a urinary tract prosthesis,
  • This may include, for example, any area in medical rooms / emergency rooms, medical treatments, operating rooms, operating theaters, rooms / rooms of intensive care units (UTIs), rooms / rooms infectious disease units, rooms / rooms of oncology units and / or other units receiving severely immunocompromised patients, isolation rooms / rooms, aircraft cabins, It may also be any surface of rooms / parts of laboratory, biological laboratory, laboratory of biological analyzes, room of incubation and another closed volume of any kind in which a biofilm is likely to be formed.
  • UTIs intensive care units
  • UTIs intensive care units
  • rooms / rooms infectious disease units rooms / rooms of oncology units and / or other units receiving severely immunocompromised patients
  • isolation rooms / rooms aircraft cabins
  • It may also be any surface of rooms / parts of laboratory, biological laboratory, laboratory of biological analyzes, room of incubation and another closed volume of any kind in which a biofilm is likely to be formed.
  • the abiotic surface may be formed by any material known to those skilled in the art. It may be for example a biocompatible material or not.
  • the material may be any biocompatible material known to those skilled in the art, for example a prosthesis, for example an osteoarticular prosthesis, a heart valve, a dental implant.
  • the material may be any material used in pipes or any container allowing the passage of a fluid, for example in production units of the food industry, pharmaceutical.
  • the abiotic surface may be for example a metal surface, a surface formed by an alloy, a polymeric surface.
  • the metal may be any metal known to those skilled in the art, for example a metal selected from the group consisting of titanium, copper, iron, aluminum, nickel, tungsten, silver, nickel, and the like. gold, palladium, vanadium, molybdenum.
  • the alloy can be any alloy known to those skilled in the art, for example an alloy selected from the group consisting of steel, brass, copper-nickel, copper-palladium, silver-gold, silver-palladium, molybdenum-vanadium, molybdenum-tungsten.
  • the polymer may be any polymer known to those skilled in the art likely to constitute and / or cover a surface. It may be for example a polymer selected from the group comprising polytetrafluoroethylene (PTFE), polysiloxanes, polyurethanes, functionalized polymers.
  • PTFE polytetrafluoroethylene
  • the use of cloxacillin to inhibit the formation of a bacterial biofilm on a surface can be carried out at a dose of 25 to 100 mg / kg of individual, preferably 25 to 50 mg / kg of individual.
  • cloxacillin when cloxacillin is used at a dose of 25 to 100 mg / kg of individual, preferably 25 to 50 mg / kg of individual, it makes it possible to obtain serum concentrations of cloxacillin ranging from 4 to 16 mg / L. preferentially at serum concentrations of 4 to 8 mg / L.
  • the concentration of cloxacillin used may be a function of the surface.
  • cloxacillin when the surface is an abiotic surface, cloxacillin may be used at a dose of 25 to 100 mg / kg of individual, for example 25 to 50 mg / kg, and / or at a concentration of 4 to 16 mg / L, preferably 4 to 8 mg / L.
  • cloxacillin when the surface is a biotic surface, cloxacillin can be used at a dose of 25 to 100 mg / kg of individual, for example 25 to 50 mg / kg of individual.
  • cloxacillin when cloxacillin is applied directly to a surface, for example biotic or abiotic, cloxacillin may be used at a concentration of 4 to 16 mg / l, preferably 4 to 8 mg / l.
  • the inventors have surprisingly demonstrated that cloxacillin allows particular and unusual concentrations to inhibit the formation of biofilm and in particular to inhibit the first step essential to this formation, namely the adhesion and fixation, also referred to as adhesion and adhesion, respectively, of bacteria on a surface.
  • adhesion and fixation also referred to as adhesion and adhesion
  • the present invention also relates to the use of cloxacillin at a concentration greater than 4 mg / L of 4 to 16 mg / L to prevent the formation of biofilms on a surface.
  • the present invention also relates to the use of cloxacillin at a concentration greater than 25 mg / kg, for example 25 to 100 mg / kg, for example 25 to 50 mg / kg to prevent the formation of biofilms on a surface .
  • the surface may be a surface as defined above.
  • "preventing the formation of biofilms” is intended to prevent adhesion and / or attachment of microorganisms to said surface in the presence of microorganisms.
  • cloxacillin When used for preventing the formation of a biofilm on a surface, cloxacillin may be contacted and / or applied to said surface by any suitable method and / or device known to those skilled in the art.
  • the bringing into contact and / or application can be carried out by sprinkling the cloxacillin on the surface, by soaking the surface in a composition comprising cloxacillin, by spraying or soaking cloxacillin in combination with a component that is supposed to act also against the formation of biofilm, such as copper.
  • a component that is supposed to act also against the formation of biofilm such as copper.
  • cloxacillin when used for the prevention and / or inhibition of the formation of a bacterial biofilm on an abiotic surface, for example a medical device, it can be applied to the outer surface of said device by any known method. of the skilled person.
  • the placing in contact or application can be carried out by sprinkling cloxacillin on said surface, by soaking the medical device in a solution comprising cloxacillin, by spraying or soaking said solution.
  • the solution may further comprise one or more active agents, for example a compound known to inhibit the formation of biofilm, for example copper.
  • cloxacillin When cloxacillin is used for the prevention and / or inhibition of bacterial biofilm formation on an abiotic surface, cloxacillin may be in any suitable form known to those skilled in the art. For example, cloxacillin may be used in liquid form and / or in powder form.
  • cloxacillin when used for the prevention and / or inhibition of the formation of a bacterial biofilm on an abiotic surface, for example a medical device, it may be applied prior to implantation and / or use of said, for example, medical device.
  • cloxacillin When cloxacillin is used for the prevention and / or inhibition of bacterial biofilm formation on a biotic surface, cloxacillin may be in any suitable form known to those skilled in the art.
  • cloxacillin may be used in liquid form, for example parenterally administered, and / or in an ingestible form, for example administered orally, or in a form suitable for topical application, for example an ointment, a cream, a dressing / woven or impregnated woven support.
  • form refers to the pharmaceutical formulation of the drug for its practical use.
  • the inventors have also surprisingly demonstrated that the use of cloxacillin according to the invention advantageously makes it possible to inhibit the formation of biofilm and in particular inhibits in particular the first step required for this formation, namely the deposition and fixation, also referred to respectively adhesion and adhesion, bacteria, for example on wounds and / or at the level of osteoarticular infections.
  • the inventors have notably demonstrated this inhibition, as described in the examples, under in vivo conditions via a inoculation on a subcutaneous implanted catheter in the thigh of a mouse and measuring the effect, in examples of pharmacokinetic / pharmacodynamic conditions defined according to the effects of cloxacillin on the biofilm installation on the catheter of same clinical strains used in vitro.
  • the inventors have also surprisingly demonstrated that cloxacillin, administered at a therapeutic dose established on the basis of the MIC of a strain, is ineffective on biofilm formation, hence on the installation of an infection.
  • the use of antibiotics at known therapeutic doses does not prevent and / or inhibit the formation / development of a biofilm a medical device, for example an implanted catheter.
  • the present invention therefore also relates to the use of cloxacillin at a dose greater than 25 mg / kg of 25 to 100 mg / kg of individual as a medicament for inhibiting the formation and / or development of a biofilm on a surface.
  • the present invention therefore also relates to the use of cloxacillin at a dose greater than 25 mg / kg, for example of 25 to 100 mg / kg, of 25 to 50 mg / kg of individual as a medicament for inhibiting formation and / or development of a biofilm on a surface.
  • the surface may be a surface as defined above.
  • the surface may be an abiotic surface, for example an outer surface of a medical prosthesis.
  • the surface may also be a biotic surface, for example the skin, mucous membranes, teeth, any surface of the vascular system, the walls of the endocardium, any surface of the lymphatic system, any surface of the Oto-Rhino-Laryngeal sphere.
  • the use of cloxacillin as a drug according to the invention can be carried out simultaneously on several surfaces, for example in the same mammal.
  • the use can be carried out on a biotic and abiotic surface, for example on a prosthetic surface and on a surface of biological tissue surrounding said prosthesis.
  • the use can be performed on at least two biotic and / or abiotic surfaces.
  • inhibition of biofilm formation is meant inhibition in the presence of microorganisms, for example microorganisms forming biofilms, adhesion and / or attachment thereof to said surface.
  • inhibition of development of a biofilm on a surface is meant inhibition in the presence of microorganisms, for example microorganisms forming biofilms, adhesion and / or attachment thereof on said surface .
  • the medicament may be intended for a mammal, for example a mammal chosen from the group comprising the order Monotremata, Didelphimorphia, Paucituberculata, Microbiotheria, Notoryctemorphia, Dasyuromorphia, Peramelemorphia, Diprotodontia,
  • Tubulidentata for example a human or an animal. This may be for example a farm animal, a pet, an endangered animal or any other animal.
  • the farm animal may be selected from the group consisting of cattle, pigs, sheep, goats, camels, canines, horses, murines.
  • the pet may be selected from the group consisting of canines and felines.
  • the term "medicament” is intended to mean a medicament for human use or a veterinary medicinal product.
  • a mammal for example a mammal selected from the group consisting of the order Monotremata, Didelphimorphia, Paucituberculata, Microbiotheria, Notoryctemorphia, Dasyuromorphia, Peramelemorphia, Diprotodontia, Tubulidentata, Sirenia, Afrosoricida, Macroscelidea, Hyracoidea, Proboscidea, Cingulata, for example the armadillo, Pilosa, Scandentia, Dermoptera, Primates, Rodentia, Lagomorpha, Erinaceomorpha, Soricomorpha, Chiroptera, Pholidota, Carnivora, Perissodactyla, Artiodactyla and Cetacea.
  • It can be for example a human or an animal. This may be for example a farm animal, a pet, an endangered animal or any other animal.
  • the farm animal may be selected from the group consisting of cattle, pigs, sheep, goats, camels, canines, horses, murines.
  • the pet may be selected from the group consisting of canines and felines.
  • cloxacillin may be in any form that can be administered to a human or an animal. Administration may be directly, i.e., pure or substantially pure, or after mixing cloxacillin with a pharmaceutically acceptable carrier and / or carrier.
  • the medicament may be in powder form, for example for solution for injection, or for oral administration to be swallowed in capsule form.
  • the medicament may be a medicament for oral administration.
  • the medicament when the medicament is a medicament for oral administration, it may for example be in the form of a capsule.
  • the medicament may be a medicament for parenteral administration, for example intravenous administration.
  • the medicament may be a medicament for topical administration, for example for a cutaneous application, for example an ointment, a cream, an impregnated dressing, a galenic patch or a spray.
  • cloxacillin when used as a medicament, it may be at a dose of 25 to 100 mg / kg of individual, for example 25 to 50 mg / kg of individual and / or at a concentration of between 4 and 16 mg / L, for example from 4 to 8 mg / L.
  • cloxacillin when the surface is a biotic surface, cloxacillin may be used at a concentration of 4 to 16 mg / l, for example from 4 to 8 mg / l.
  • cloxacillin when the surface is a biotic surface, cloxacillin may be used at a concentration of 25 to 100 mg / kg, for example 25 to 50 mg / kg of individual, and / or at a concentration of between 4 and 16 mg / L, for example from 4 to 8 mg / L.
  • cloxacillin as a medicament may be carried out at a concentration such that its bioavailability at the surface may be from 4 to 16 mg / l.
  • the inventors have surprisingly demonstrated that the use of cloxacillin at particular quantities / concentrations advantageously makes it possible to inhibit the formation and / or the development of a biofilm on the surface of medical devices and in particular of medical devices. implanted in a mammal.
  • the use of cloxacillin according to the invention advantageously makes it possible to inhibit the formation and / or the development of a biofilm on the surface, in particular of an implanted catheter, while the use of cloxacillin at a usual therapeutic dose, that is to say that used for the treatment of bacterial infections, does not prevent the implantation and / or the formation of the biofilm or inhibit its development.
  • cloxacillin advantageously makes it possible to prevent / inhibit the formation of a biofilm and also the installation of an infection / a bacterial colony contrary to known uses. .
  • the inventors have therefore demonstrated that the use according to the invention advantageously makes it possible to prevent tolerance and / or resistance and / or multi-bacterial resistance related to the formation and / or the development of a biofilm on a surface.
  • the inventors have also demonstrated that the use according to the invention advantageously makes it possible to prevent a bacterial tolerance linked to the formation and / or the development of a biofilm on a surface.
  • the inventors have also demonstrated that the use according to the invention advantageously makes it possible to prevent bacterial resistance linked to the formation and / or the development of a biofilm on a surface.
  • the present invention therefore also relates to Cloxacillin for use as a medicament for preventing bacterial multi-resistance related to the formation and / or development of a biofilm on a surface.
  • the present invention therefore also relates to the use of cloxacillin at a dose greater than 25 mg / kg of individual, for example from 25 to 100 mg / kg of individual and / or at a concentration of between 4 and 16. mg / L, for example from 4 to 8 mg / L as a medicament for to prevent bacterial multi-resistance related to the formation and / or development of a biofilm on a surface.
  • the subject of the present invention is also the use of cloxacillin at a dose greater than 25 mg / kg of individual, for example ranging from 25 to 100 mg / kg, from 25 to 50 mg / kg of individual as a medicine for preventing multi-bacterial resistance related to the formation and / or development of a biofilm on a surface.
  • cloxacillin when cloxacillin is used at a dose greater than 25 mg / kg of individual, for example from 25 to 100 mg / kg of individual as a drug to prevent bacterial multi-resistance related to the formation and / or development of a biofilm on a surface, it makes it possible to obtain a serum concentration of cloxacillin ranging from 4 to 16 mg / l advantageously to prevent a multi-bacterial resistance linked to the formation and / or the development of a biofilm on a surface .
  • bacterial multi-resistance means the ability of bacteria to resist several antibiotic treatments, in particular the ability of bacteria to resist at least two antibiotic treatments.
  • the present invention therefore also relates to Cloxacillin for use as a medicament for preventing bacterial tolerance related to the formation and / or development of a biofilm on a surface.
  • the subject of the present invention is also the use of cloxacillin at a dose greater than 25 mg / kg of individual, for example ranging from 25 to 100 mg / kg of individual and / or at a concentration of between 4 and 16 mg. / L, for example 4 to 8 mg / L as a drug to prevent bacterial tolerance related to the formation and / or development of a biofilm on a surface.
  • the subject of the present invention is also the use of cloxacillin at a dose greater than 25 mg / kg of individual, for example ranging from 25 to 100 mg / kg, from 25 to 50 mg / kg of d as a drug to prevent bacterial tolerance related to the formation and / or development of a biofilm on a surface.
  • cloxacillin when cloxacillin is used at a dose greater than 25 mg / kg of individual, for example from 25 to 100 mg / kg of individual as a medicament for preventing a bacterial tolerance related to the formation and / or development of a biofilm on a surface, it makes it possible to obtain a serum concentration of cloxacillin of between 4 and 16 mg / l, advantageously making it possible to prevent a bacterial tolerance linked to the formation and / or the development of a biofilm on a surface.
  • bacterial tolerance is meant the ability of bacteria to resist naturally, that is to say without being identified (s) mutation (s) or gene (s) conferring one or more resistance (s) , one or more antibiotic treatment (s).
  • the present invention also relates to Cloxacillin for use as a medicament for preventing bacterial resistance related to the formation and / or development of a biofilm on a surface.
  • the subject of the present invention is also the use of cloxacillin at a dose greater than 25 mg / kg of individual, for example of 25 to 100 mg / kg, of an individual and / or at a concentration of between 4 and 16. mg / L, for example 4 to 8 mg / L as a drug to prevent bacterial resistance related to the formation and / or development of a biofilm on a surface.
  • the subject of the present invention is also the use of cloxacillin at a dose greater than 25 mg / kg of individual, for example ranging from 25 to 100 mg / kg, from 25 to 50 mg / kg of d 'individual as a drug to prevent bacterial resistance related to the formation and / or development of a biofilm on a surface.
  • cloxacillin when cloxacillin is used at a dose greater than 25 mg / kg of individual, for example between 25 and 100 mg / kg of individual as a drug to prevent bacterial resistance related to the formation and / or development of a biofilm on a surface, it makes it possible to obtain a serum concentration of cloxacillin of between 4 and 16 mg / L, advantageously making it possible to prevent a bacterial resistance linked to the formation and / or the development of a biofilm on a surface.
  • Bacterial resistance herein refers to the ability of mutant or acquired bacteria to have one or more resistance gene (s) to resist one or more antibiotic (s) treatment (s).
  • mutation and / or acquisition of the resistance gene allows the bacterium to resist one or more antibiotic treatment (s).
  • the conditions, amounts and routes of administration of the cloxacillin used according to the invention may be as described herein, for example, above.
  • the skilled person with this general knowledge will adapt the conditions and forms used depending on the surface.
  • the inventors have demonstrated that the use according to the invention makes it possible to inhibit or prevent the formation of a biofilm on a surface and thus a possible tolerance (also called recalcitrance) and / or resistance to antibiotics, in particular by the presence of biofilm.
  • the use according to the present invention allows both to inhibit or prevent the formation of a biofilm and possibly to treat the infection.
  • the use according to the invention of cloxacillin on biotic surfaces advantageously makes it possible to prevent and / or inhibit the formation of biofilm.
  • the use according to the invention makes it possible to prevent and / or inhibit an additional infection and / or superinfection by bacteria forming biofilms, for example at the level of wounds.
  • the subject of the present invention is therefore also the use of cloxacillin at a concentration greater than 25 mg / kg of individual, for example ranging from 25 to 100 mg / kg of individual and / or at a concentration of between 4 and 16. mg / L, for example from 4 to 8 mg / L as a medicament for preventing and / or inhibiting an additional infection and / or superinfection by bacteria forming biofilms of an infected biotic surface.
  • the subject of the present invention is also the use of cloxacillin at a dose greater than 25 mg / kg of individual, for example ranging from 25 to 100 mg / kg, from 25 to 50 mg / kg of d as a medicament for preventing and / or inhibiting an additional infection and / or superinfection by bacteria forming biofilms of an infected biotic surface.
  • prevention and / or inhibition of additional infection and / or superinfection by bacteria is meant inhibition in the presence of microorganisms, for example microorganisms forming biofilms, adhesion and / or attaching them to said infected surface.
  • biotic surface infected any biotic surface known to those skilled in the art that may have a bacterial infection.
  • bacterial infection is meant any bacterial infection known to those skilled in the art likely to affect a biotic or abiotic surface.
  • bacterial infections for example of biotic surfaces, for example skin may be abscesses, folliculitis, boils, furunculosis, anthrax, paronychia, phlegmon, perleche, intertrigo, stye, erysipelas, impetigo, prurigo.
  • cloxacillin when used as a medicament for preventing and / or inhibiting an additional infection and / or superinfection by biofilm forming bacteria of an infected biotic surface, it may be used at a dosage of 25%. at 100 mg / kg of individual, for example from 25 to 50 mg / kg of individual and / or at a concentration of between 4 and 16 mg / l, for example from 4 to 8 mg / l.
  • cloxacillin when used as a medicament for preventing and / or inhibiting an additional infection and / or superinfection by bacteria forming biofilms of an infected biotic surface, it may be used at a dose of 25 to 100 mg / kg of individual, for example 25 to 50 mg / kg of individual.
  • cloxacillin when cloxacillin is used at a dose greater than 25 mg / kg of individual, for example from 25 to 100 mg / kg of individual as a medicament for preventing and / or inhibiting an additional infection and / or superinfection by biofilms forming bacteria of an infected biotic surface, it makes it possible to obtain a serum concentration of cloxacillin of between 4 and 16 mg / l, for example from 4 to 8 mg / l, advantageously making it possible to prevent bacterial multi-resistance related to the formation and / or development of a biofilm on a surface.
  • cloxacillin is used as a medicament for preventing and / or inhibiting additional infection and / or superinfection by biofilm-forming bacteria of an infected biotic surface, it may be used at a concentration of 25 to 100 mg / ml. kg of individual, for example from 25 to 50 mg / kg of individual and / or at a concentration of between 4 and 16 mg / l, for example from 4 to 8 mg / l.
  • cloxacillin when used as a medicament for preventing and / or inhibiting an additional infection and / or superinfection of a biotic surface infected with bacteria forming biofilms, it can be used at a dose of 25 to 100 mg / kg of individual, for example 25 to 50 mg / kg of individual.
  • cloxacillin when cloxacillin is used at a dose greater than 25 mg / kg of individual, for example from 25 to 100 mg / kg of individual as a medicament for preventing and / or inhibiting an additional infection and / or superinfection by biofilms forming bacteria of an infected biotic surface, it makes it possible to obtain a serum concentration of cloxacillin of between 4 and 16 mg / l, for example from 4 to 8 mg / l, advantageously making it possible to prevent bacterial multi-resistance related to the formation and / or development of a biofilm on a surface.
  • the use of cloxacillin as a medicament may be carried out at a concentration such that its bioavailability at the level of the surface may be from 4 to 16 mg / l.
  • cloxacillin For its use as a medicament for preventing and / or inhibiting further infection and / or superinfection by biofilm-forming bacteria of an infected biotic surface, cloxacillin may be in any suitable form known to those skilled in the art. It may be of a form as mentioned above.
  • the present invention also relates to a pharmaceutical composition
  • a pharmaceutical composition comprising cloxacillin at a dose greater than 0.4%, for example ranging from 0.4% to 1.6% by weight percentage relative to the total weight of the composition. intended to be used as a medicament for preventing the formation of a biofilm on a surface.
  • the subject of the present invention is also a pharmaceutical composition comprising cloxacillin at a dose greater than 4 mg / l, for example ranging from 4 to 16 mg / l intended to be used as a medicament for the prevention of the formation of a biofilm on a area.
  • composition according to the invention can be carried out on any surface known to those skilled in the art. It can be for example a biotic or abiotic surface as defined above. It may be for example one or more surfaces as defined above.
  • the concentration / amount of cloxacillin may be greater than 0.4%, for example, from 0.4% to 1.6% by weight relative to the total weight of the composition
  • the concentration / amount of cloxacillin may be greater than 4 mg / L example of 4 to 16mg / L.
  • compositions according to the invention may be as described herein, for example above.
  • skilled person with this general knowledge will adapt the conditions and channels of administration of the composition.
  • compositions of the present invention may further comprise a pharmaceutically acceptable carrier, adjuvant or vehicle.
  • a pharmaceutically acceptable carrier, adjuvant or carrier includes any solvent, diluent or other liquid vehicle, dispersion or suspension aid, surfactant, isotonic agent, thickening or emulsifying agent, preservative, binder solid, lubricant and the like, adapted to the particular dosage form desired.
  • Remington Pharmaceutical Sciences, Sixteenth Edition, EW Martin (Mack Publishing Co., Easton, Pa., 1980) describes various carriers used in the formulation of pharmaceutically acceptable compositions and known techniques for their preparation.
  • constituents which may serve as pharmaceutically acceptable carriers include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins such as human serum albumin, buffering substances such as phosphates, glycine, sorbic acid, or potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes such as protamine sulfate, disodium phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinylpyrrolidone, polyacrylates, waxes, polyethylene-polyoxypropylene polymers, sugars such as lactose glucose and sucrose; starches such as corn and potato starch; cellulose and its derivatives such as sodium carboxymethylcellulose, ethylcellulose and cellulose acetate; tragacanth powder; malt; gelatin; talc; excipients such as cocoa butter and sup
  • Ringer ethyl alcohol, and solution phosphate buffer, as well as other nontoxic compatible lubricants such as sodium lauryl sulphate and magnesium stearate, as well as coloring agents, mold release agents, coating agents sweetening, flavoring and perfuming agents, preservatives and antioxidants may also be present in the composition, in the judgment of galenist.
  • nontoxic compatible lubricants such as sodium lauryl sulphate and magnesium stearate
  • coloring agents such as sodium lauryl sulphate and magnesium stearate
  • coating agents sweetening, flavoring and perfuming agents, preservatives and antioxidants may also be present in the composition, in the judgment of galenist.
  • compositions of the present invention can be administered to humans and other animals orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically (as by powders, ointments or drops), orally, as a oral or nasal spray, or the like, depending on the severity of the infection to be treated.
  • Liquid dosage forms for oral administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs.
  • the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (in particular, cottonseed peanut, corn, seed, olive, and sesame), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and sorbitan fatty acid esters, and mixtures thereof.
  • the oral compositions may also include adjuvants such as wetting agents, emuls
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders and granules.
  • the active compound is mixed with at least one inert carrier, excipient or pharmaceutically acceptable carrier such as sodium citrate or dicalcium phosphate and / or a) fillers or diluents such as starches, lactose, sucrose, glucose, mannitol and silicic acid, b) binders such as, for example, carboxymethylcellulose, alginate, gelatin, polyvinylpyrrolidone, sucrose and acacia, c) humectants such as glycerol, d) disintegrating agents such as agar-agar, calcium carbonate, potato or cassava starch, alginic acid, certain silicates and sodium carbonate, e) absorption accelerators such as quaternary ammonium compounds, f) wetting agents such as, for example, cetyl alcohol and glycerol monostearate, g) absorbents or
  • Solid compositions of a similar type can also be employed as fillers in soft or hard gelatin capsules using excipients such as lactose or milk sugar as well as polyethylene glycols of high molecular weight polyethylene and the like.
  • excipients such as lactose or milk sugar as well as polyethylene glycols of high molecular weight polyethylene and the like.
  • the solid dosage forms of tablets, dragees, capsules, pills and granules can be prepared with coatings and envelopes such as enteric coatings and other coatings well known in the pharmaceutical art formulating. They may optionally contain opacifying agents and may also be of a composition such that they release the active ingredient (s) only, or preferentially, in a certain part of the intestinal tract, optionally in a delayed manner.
  • coating compositions that can be used include polymeric substances and waxes.
  • compositions of a similar type can also be employed as fillers in soft gelatin capsules and hard using excipients such as lactose or milk sugar as well as polyethylene glycols of high molecular weight polyethylene and the like.
  • the uses and / or compositions according to the invention can be combined with conventional antibacterial agents, such as antibiotics, for example bactericidal and / or bacteriostatic.
  • the conventional antibacterial agents may be used or administered previously, simultaneously, with the use and / or the composition according to the invention.
  • the uses and / or compositions according to the invention can be combined with conventional antifungal agents, for example commercially available antifungal agents, for example niconazole.
  • conventional antifungal agents for example commercially available antifungal agents, for example niconazole.
  • the conventional antifungal agents may be used or administered beforehand, simultaneously, with the use and / or the composition according to the invention.
  • cloxacillin may be used according to the invention and / or the pharmaceutical composition may be used according to the invention according to the invention in combination with at least one of the antibiotics selected from the group comprising aminoglycosides, fluoroquinolones, macrolides , novobiocin, rifampicin, oxazolidinones, fusidic acid, mupirocin, pleuromutilins, daptomycin, vancomycin, tetracyclines, sulfonamides, chloramphenicol, trimethoprim, fosfomycin, cycloserine and polymyxin.
  • the antibiotics selected from the group comprising aminoglycosides, fluoroquinolones, macrolides , novobiocin, rifampicin, oxazolidinones, fusidic acid, mupirocin, pleuromutilins, daptomycin, vancomycin, tetracyclines,
  • Figure 1 comprises a scanned image (scan) of 96-well plate and a diagram showing the evolution of the biofilm index (BFI) (ordinate) as a function of the concentration of cloxacillin in g / mL.
  • FIG. 1A is a digitized image of the plate after magnetization and represents the crude result of the Antibiofilmogram of strain S14 and the strain
  • lines E and A correspond to strains S14 and S39 cultured in the presence of different concentrations of cloxacillin.
  • Lines B, C and D correspond to other strains not mentioned here.
  • Line F corresponds to the controls in absence of strain (normal presence of a spot), the wells at H1 and H2 correspond to strain S14 alone, and wells H9 and H10 correspond to strain S39 alone.
  • Each column from 1 to 12 corresponds to different concentrations in g / ml of cloxacillin, ie from column 1 to column 12): 32/16/8/7/6/5/4/3/2/1/0 , 5 / 0.25 ⁇ g / ml.
  • FIG. 2 represents images acquired by confocal microscopy of the bottom well surface in which bacteria have been cultured, the light spots on these photographs correspond to the bacteria forming the biofilm.
  • the diagram represents the percentage of surface coverage by a biofilm (ordinate).
  • FIG. 2A corresponds to the results obtained with bacterial strain S14 cultivated without antibiotic (control) with a concentration of cloxacillin lower than the MIC ( ⁇ MIC), a concentration of cloxacillin equal to its MIC (MIC), a concentration between the MIC and the concentration inhibiting biofilm formation ( ⁇ cmib) and a concentration equal to the concentration inhibiting biofilm formation (cmib).
  • FIG. 2B corresponds to the results obtained with bacterial strain S39 cultivated without antibiotic (control) with a concentration of cloxacillin less than the MIC ( ⁇ MIC), a concentration of cloxacillin equal to its MIC (MIC), a concentration between the MIC and the concentration inhibiting biofilm formation ( ⁇ cmib) and a concentration equal to the concentration inhibiting biofilm formation (cmib).
  • Figure 3 shows diagrams corresponding to the enumeration of bacteria attached to an implanted catheter in mice based on the presence or absence of cloxacillin.
  • the ordinate corresponds to the decimal log of colony forming units (Log10 CFU) per gram of catheter (Logio CFU / Gram of catheter).
  • the abscissa at time after infection with the strain and depending on treatment with cloxacillin: without cloxacillin (Control) after 12 hours (H12), 24 hours (H24), after 30 hours (H30) or with treatment with cloxacillin (Cloxa) at different concentrations ( ⁇ or>).
  • FIG. 3A represents the diagram obtained with strain S14, cloxacillin being administered at a concentration of 3 mg / kg ( ⁇ 2) or 15 mg / kg (> 2).
  • Figure 3B shows the diagram obtained with strain S39, cloxacillin being administered at a concentration of 4 mg / kg ( ⁇ 4) or 25 mg / kg (> 4).
  • FIG. 4A shows the measurement of the scarring score in infected mice, treated either with cloxacillin corresponding to the MIC of strain S14 (3 mg / kg), or with cloxacillin corresponding to the dose CMIb (15 mg / kg), or by physiological saline (vehicle).
  • FIG. 4B shows the measurement of the scarring score in uninfected mice (control), treated either with cloxacillin corresponding to the MIC of strain S14 (lnfected / 3 mg / kg), or with cloxacillin corresponding to the CMIb dose ( lnfected / 15mg / Kg), either with physiological saline (Infected / control).
  • Figure 4C shows the count of bacteria installed in the wound. Their number is conventionally expressed in CFU (Colony-Forming Units) corresponding to the number of colonies counted on a petri dish.
  • CFU Cold-Forming Units
  • an Antibiofilmogram was performed to demonstrate the inhibition of cloxacillin biofilm formation and to determine doses of cloxacillin effective against biofilm formation in a polystyrene microplate well.
  • This example clearly demonstrates that Minimal Inhibitory Concentrations of biofilm formation (MICb) are different from Minimum Inhibitory Concentrations.
  • MIC sensitivity to the antibiotic in relation to its activity bactericide.
  • These MICs can differ between 2 strains whereas the MICs for these strains are close (0.125 and 0.25 g / ml, or mg / L) and on the other hand are very far from the MICs.
  • the bacterial inoculum was 4.10 6 bacteria / ml, in 200 ⁇ of BHI (Brain Heart Infusion) medium, in the presence of magnetic microbeads (Toner 4, reference TON004) at the concentration of 2 ⁇ for 200 ⁇ , ie ⁇ ⁇ / ml).
  • the microplate was incubated for 4 hours at 37 ° C., then was coated with 120 ⁇ of contrast liquid (contrast liquid, reference LIC001) and was placed 1 minute on a magnet block (reference BKTMB) designed so that each mini-magnet is centered with respect to the 96 wells.
  • the microplate was then placed on a scanner and the image was acquired and analyzed by the BFCE3 software which quantifies the aggregation of the magnetic microbeads in the center of the wells, and provides a BFI value (Biofilm Training Index).
  • a BFI of 20, or close to 20 corresponds to a maximum aggregation, therefore an absence of biofilm formation, thus an anti-adherence efficacy of cloxacillin.
  • a BFI of 0 corresponds to a total immobility of the microbeads completely blocked by the adhered bacteria. All intermediate values, allowing a quantitative measure of the effectiveness of cloxacillin are possible.
  • FIG. 1A is a digitized image (scan) of the plate after magnetization and represents the raw result of the Antibiofilmogram of the strain S14 and FIG. 1B is a diagram representing the evolution of the
  • BFI (ordinate) as a function of cloxacillin concentration in ⁇ g mL.
  • lines E and A correspond to strains S14 and S39 grown in the presence of varying concentrations of cloxacillin, respectively.
  • Lines B, C and D correspond to other strains not mentioned here.
  • Line F corresponds to controls in absence of strain (normal presence of a spot) and the wells at H1 and H2 correspond to strain S39 alone, and wells H9 and H10 correspond to strain S39. Strains are tested at the following concentrations, starting from column 1 to column 12: 32/16/8/7/6/5/4/3/2/1/05 / 0,25Mg / ml,
  • Figure 1 B represents the curves corresponding to the BFIs measured in the wells of FIG.
  • MIC Minimal Inhibitory Concentration
  • the MIC was measured in Mueller-Hinton medium (MH, reference medium for antibiotic susceptibility testing) and Brain Heart Broth medium (BHI).
  • the MICs in MH and BHI media were very similar, showing that the culture medium in which the bacteria were cultured for the cloxacillin susceptibility test had no effect.
  • strains S02 and S83 have MICs in MH medium of 0.125 g / ml while their MICs were respectively 2 and 16 mg / ml.
  • strains S38, S58 and S83 have a MICr of 16 g / ml and have MICs of 0.5, 0.25 and 0.125 g / ml respectively.
  • cloxaciline as a medicament for inhibiting the formation and / or development of biofilm on a surface is carried out at concentrations and / or doses very different from those known for an antibiotic effect.
  • concentrations and / or doses very different from those known for an antibiotic effect.
  • the administration of cloxacillin at a dose for example 49 mg / kg of said subject advantageously allows to inhibit the formation and / or development of biofilm by said bacterium.
  • Example 2 confirms by confocal microscopy the interpretations of the Antibiofilmogram. It is clearly shown that the cloxacillin antibiotic, used at the MIC dose (a dose defined as being sufficient for the bactericidal activity of cloxacillin on the strain in question), namely in particular strain S14 and S39, is not active on the formation of biofilm on the plastic. On the other hand, at the CMIb dose (defined as effective in preventing biofilm formation), the strain did not adhere to the surface. These results were obtained for strains S14 and S39 whose MICs are very close and characterize strains as sensitive to cloxacillin.
  • the 6-well microplates have were used to obtain a sufficient biofilm formation surface for confocal microscopic observation. The concentrations corresponding to the MICs and MICs of the strains were tested. In parallel, controls in the absence of antibiotics, strains S14 and S39 were performed. After a 4h incubation at 37 ° C.
  • Example 2 Antibiofilmogram (registered trademark)
  • the microplate funds were cut and assembled with a glass slide in order to be able to observe confocal microscopy
  • the observation conditions were as follows: Objective: x40, Zoom: x2, Size: 2048 * 2048 pixels, Laser power, Zmax and Zmin: number of images of the battery, Size between each image: 0.42 ⁇ A "Live / Dead” staining was performed, SYTO9 dyes the living (and dead) bacteria, while the Propidium iodide stain specifically stains the dead bacteria, resulting in an analysis of 2880 images. Slices ").
  • FIG. 2 The results obtained are shown in FIG. 2.
  • the recovery rate of the bottom surface of the wells by the biofilm as a percentage of surface coverage is indicated on the ordinate.
  • Figure 2A the density of S14 bacteria adhered to the bottom of the wells was shown and analyzed.
  • the antibiotic at a dose CMI dose allows a decrease in the adhesion of bacteria, but does not inhibit it.
  • from 10 to more than 20% of the bottom surface of the well was covered after 4 hours of incubation which is too much to consider use in a therapeutic treatment at this dose.
  • CMIb (2 g / mL) and subCMIb (I ⁇ g / mL, lower than MIC, but greater than MIC) doses were able to completely inhibit the adhesion of S. aureus strain S14. Indeed, as shown in FIG. 2A, the incubation at a concentration equal to the CMIb and subCMIb allowed to inhibit the adhesion of bacteria, especially S. aureus strain S14 and thus the formation of biofilm. On the other hand, the doses CMI (0.25Mg / ml) and subCMI (0.125Mg / ml) do not inhibit the adhesion of the S14 strain since 10% of the surface is covered by the bacteria at the CMI dose.
  • Figure 2B shows the results obtained with S. aureus strain S39.
  • Figure 2B shows the density of S39 bacteria adhered to the polystyrene and includes the corresponding analysis.
  • the doses subCMI (1/2 MIC) and MIC (0.125 g / ml) had no effect on bacterial adhesion.
  • the results obtained are similar to that of the control condition (without antibiotics).
  • This example demonstrates and confirms in an animal model of infection that cloxacillin, used at doses in connection with the MICI, has inhibited the adhesion of bacteria to the catheter and can therefore be used to prevent infection of a patient. catheter (simulating a prosthesis).
  • MIC Minimum Inhibitory Concentration
  • the inoculum size tested was 7 log CFU / mouse (or 8.3 log CFU / mL).
  • the strains were isolated on Chapman agar medium and cultured at 37 ° C for 18h.
  • an isolated colony was resuspended in 9 ml of BHI medium with stirring for 6 hours at 37 ° C., then the broth thus obtained was flooded on MH agar medium (beef extract 2 g / L, acid hydrolyzate of casein 17 5g / L; starch 1.5g / L; agar 17g / L) placed at 37 ° C for 18h.
  • mice received (30 minutes pre-infection) a "preventive" antibiotic treatment (Cloxacillin 5mg / kg or 30mg / Kg or sterile saline) intraperitoneally. These doses were determined following an in-house PK / PD study.
  • mice received (30 minutes pre-infection) a "preventive" antibiotic treatment (Cloxacillin 7mg / kg or 45mg / Kg or sterile saline) intraperitoneally. These doses were determined following a PK PD study performed internally. Serum concentrations of cloxacillin under different conditions were measured by HPLC. All the results were integrated into a pharmacokinetic simulation software and then a PKPD software to estimate the different parameters related to the planktonic MIC (Cmax / MIC, AUC / MIC and T> CMI) and the MIC (Cmax / CMIb) , AUC / CMIb and T> CMIb). In parallel, the control animals received physiological saline.
  • a "preventive" antibiotic treatment Cloxacillin 7mg / kg or 45mg / Kg or sterile saline
  • cloxacillin (Orbenine (registered trademark) 1g, France, Pfizer, France) was used. Then fixed anesthesia by intraperitoneal injection of a mixture ketamine (50mg / kg) and xylazine (10mg / kg) was administered.
  • the mixture of anesthetics was made as follows: 4ml Ketamine + 2ml of xylazine +10 ml of physiological saline. About 120 ⁇ were injected with IP into the mouse. The flanks were shaved and then disinfected (Betadine cycle).
  • a cutaneous and subcutaneous incision of 0.2 cm was performed sterilely and a 1 cm segment of polyurethane catheter cut into two longitudinal parts (Ref ES-04730 Arrow International) was inserted subcutaneously (at 2 cm distance from the incision point).
  • the deposition of the bacterial inoculum was performed at the same time.
  • the volume deposited was 50 L per mouse and the concentration tested was 10 7 log CFUs per catheter.
  • the incision was then sutured and disinfected.
  • the mice were then treated every two hours after the first preventive treatment for 10 hours with either cloxacillin at 3 mg / kg or 15 mg / kg (for strain S14) or cloxacillin at 4 mg / kg or 25 mg / kg.
  • mice were euthanized at different post-infection times (12h, 24h and 30h) by anesthesia and cervical dislocation. The catheter segments were removed. They were used for bacterial quantification after sonication.
  • Each polyurethane segment was individually and sterilely washed in an Eppendorf tube (3 successive washes with 300 L of sterile physiological saline). After the last wash, the catheter was resuspended in 1 mL of sterile physiological saline, placed in an ultrasonic bath at room temperature for 3 min (AdvantageLab) and vigorously vortexed to remove adhering bacteria from the body. catheter. Successive dilution cultures (pure, 10 "2 , 10 " 4 ) of this bacterial suspension were then performed on Chapman agar plates. The cultures were placed in an oven at 37 ° C. for 48 hours before reading.
  • Fig. 3A shows the enumeration of the attached strain S14 bacteria on the implanted catheter based on the absence of cloxacillin (control), a concentration equal to the MIC or concentration equal to the IJCB.
  • concentration of cloxacillin under the control and MIC conditions shows the inefficacy of cloxacillin at the CMI dose to inhibit bacterial adhesion, at the time of measurement at 12h, 24h and 30h.
  • ANOVA One Way analysis of variance complemented by a Bonferroni post-hoc test was used.
  • FIG. 3B shows the enumeration of the bacteria of strain S39 attached to the implanted catheter as a function of the absence of cloxacillin (control), a concentration equal to the MIC or at a concentration equal to the MIC.
  • control a concentration equal to the MIC
  • concentration of cloxacillin under the control and MIC conditions shows the inefficacy of cloxacillin at the CMI dose to inhibit the adhesion of the bacteria, at the time of measurement 12h, 24h and 30h.
  • cloxacillin makes it possible to inhibit the adhesion of bacteria to a surface, for example a prosthesis.
  • this example clearly demonstrates that the usual concentrations of cloxacillin as an antibiotic do not prevent / inhibit the adhesion of bacteria.
  • the objective of these trials was to test the efficacy of cloxacillin, in selected doses, in an in vivo model of wound infection in mouse skin by Staphylococcus aureus strains.
  • the in vivo efficacy of particular doses of cloxacillin, deduced from the MIC and MIC doses in vitro was determined by measuring the bacterial load (established as a biofilm) at 12h, Day 5, and D-Day. infection, and by measurement of healing on Day 5 and Day 1 1 post-infection (in the form of a scarring score).
  • mice used were 7 week old C57BL / 6 females, weighing between 20 and 22 g, from Janvier Labs, Genest St Isle, 53941 St Berthevin, France. The number of mice used was 5 per condition tested.
  • the cloxacillin used came from a commercial solution of cloxacillin (Orbenine (registered trademark) 1g, France, Pfizer, France).
  • the cutaneous wound model was performed as follows.
  • the concentration of S. aureus was adjusted in PBS to give a concentration of 10 6 CFU in 10 ⁇ (10 8 CFU / ml).
  • mice received, 30 minutes pre-infection, a "preventive" treatment of 5 mg / kg of cloxacillin (for mice then receiving cloxacillin at 3 mg / kg) or 30 mg / kg. of Cloxacillin (for mice then receiving cloxacillin at 15 mg / kg) or sterile saline intraperitoneally. These doses were determined following an in-house PK / PD study.
  • the serum concentrations of cloxacillin according to the different conditions were measured by HPLC according to a conventionally described protocol (see for example Jonsson et al, "Cloxacillin concentrations in serum, subcutaneous fat, and muscle in patients with chronic critical limb ischemia” Eur J Clin Pharmacol (2014) 70 (8): 957-63). All the results were integrated in a pharmacokinetic simulation software, namely then a PKPD software to estimate the various parameters related to the planktonic MIC (Cmax / MIC, AUC / MIC and T> CMI) and the CMIb (Cmax / MICb, AUC / MICb and T> MICb).
  • control animals namely 5 mice, received physiological saline.
  • mice were then treated every two hours after the first preventive treatment for 10 hours with cloxacillin at 3 mg / kg or 15 mg / kg or physiological saline. These doses were determined beforehand so as to simulate blood concentrations between the planktonic MIC and the MIC biofilm of strain S14. Three groups of mice were used, a first group being sacrificed at t + 12h after infection to measure the bacterial load. A second group is treated identically and sacrificed 5 days after infection and a third group treated identically is sacrificed 11 days after infection. Each day, a scarring score is established on the animals, and on the sacrificed animals, a count of the bacteria installed is made (on D5 and J1 1). The score is set as follows, assigning a value between 0 and 3.
  • the skin at the level of the wounds is cut, rinsed in PBS to remove the non-adhered bacteria and then the tissue is homogenized in the Precellys tubes using glass beads.
  • the resulting homogenate was serially diluted, and different dilutions were plated on TSA plates and grown overnight at 37 ° C. The colonies (CFU) were quantified for each dilution and the bacterial load at the site of the wound was thus determined.
  • FIG. 4A shows the measurement of the scarring score in infected mice, treated either with cloxacillin corresponding to the MIC of strain S14 (3 mg / kg), or with cloxacillin corresponding to the CMIb dose.
  • FIG. 4B shows the measurement of the scarring score in uninfected mice (control), treated either with cloxacillin corresponding to the MIC of strain S14 (lnfected / 3 mg / kg), or with cloxacillin corresponding to the CMIb dose ( lnfected / 15mg / Kg), either with physiological saline (Infected / control).
  • Figure 4C shows the count of bacteria installed in the wound. Their number is conventionally expressed in CFU (Colony-Forming Units) corresponding to the number of colonies counted on a petri dish.
  • CFU Coldy-Forming Units
  • Figure 4B shows that cloxacillin does not have a per se effect on healing in the absence of infection.
  • the count of bacteria installed on the wound after 12h, 5 days and 11 days shows that the administration of cloxacillin at the dose CMIb of the strain (15mg / kg) can significantly reduce the bacterial load at the site of injury (Iogio6.1 vs Iogio8.82 for control at 12h), almost 3logio of decrease, then the bacterial burden continues to decrease faster than control conditions without cloxacillin and cloxacillin at the MIC dose while the dose of cloxacillin CMIb leads, on day 5, to a measurement of Iogio4.9 for the dose CMIb against Iogio6.86 for the condition CMI and Iogio6.84 for the control.
  • the parallel evolution, starting from 5 days, of the control conditions without cloxacillin and cloxacillin at the MIC dose suggests that it is the immune system that is responsible for the decrease of the
  • This example shows the inhibitory effect of installation of Staphylococcus aureus on the site of a wound (formation of a biofilm) by cloxacillin at the dose corresponding to the MICI, which is correlated with better healing of the injured site, and allows to conclude to the effect promoting cicatrization of cloxacillin at the CMIb dose of the strain by inhibition of the installation of the strain.
  • the administration of cloxacillin at an effective dose as an antibiotic (MIC dose) has no effect.

Landscapes

  • Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Steroid Compounds (AREA)
EP18762363.2A 2017-10-18 2018-07-23 Verwendung von cloxacillin zur verhinderung/hemmung von biofilmbildung Withdrawn EP3697414A1 (de)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
FR1759783A FR3072279B1 (fr) 2017-10-18 2017-10-18 Utilisation de la cloxacilline pour inhiber/ empecher la formation de biofilm
PCT/FR2018/051880 WO2019077211A1 (fr) 2017-10-18 2018-07-23 Utilisation de la cloxacilline pour inhiber/ empecher la formation de biofilm

Publications (1)

Publication Number Publication Date
EP3697414A1 true EP3697414A1 (de) 2020-08-26

Family

ID=61223995

Family Applications (1)

Application Number Title Priority Date Filing Date
EP18762363.2A Withdrawn EP3697414A1 (de) 2017-10-18 2018-07-23 Verwendung von cloxacillin zur verhinderung/hemmung von biofilmbildung

Country Status (6)

Country Link
US (1) US20200237731A1 (de)
EP (1) EP3697414A1 (de)
CN (1) CN111479569A (de)
CA (1) CA3079023A1 (de)
FR (1) FR3072279B1 (de)
WO (1) WO2019077211A1 (de)

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013032964A1 (en) * 2011-08-26 2013-03-07 Board Of Regents Of The University Of Nebraska Compositions and methods for preventing and treating biofilms
WO2014155020A1 (fr) * 2013-03-28 2014-10-02 Biofilm Control Antibiogramme rapide

Also Published As

Publication number Publication date
FR3072279B1 (fr) 2020-11-13
FR3072279A1 (fr) 2019-04-19
WO2019077211A1 (fr) 2019-04-25
CA3079023A1 (fr) 2019-04-25
US20200237731A1 (en) 2020-07-30
CN111479569A (zh) 2020-07-31

Similar Documents

Publication Publication Date Title
EP3419676B1 (de) Antimikrobielle zusammensetzungen und verwendungen davon
DK1879655T3 (en) Pharmaceutical composition comprising an antibacterial agent and an active substance selected from carveol, carvacrol, alpha-ionone, beta-ionone, and thymol
AU2018282257A1 (en) A composition comprising an antibiotic and a dispersant or an anti-adhesive agent
US20170044197A1 (en) Antimicrobial preparation and uses thereof
US9034918B2 (en) Composition including at least one trans-cinnamaldehyde and the use thereof in the treatment of bacterial infections, specifically in the treatment of nosocomial infections
WO2009020455A1 (en) Compositions and methods for microbe storage and delivery
JP2019529398A (ja) 細菌感染の予防および治療のためのトリアゾロ(4,5−d)ピリミジン誘導体の新規な使用
EP3986394A1 (de) Biofilmunterbrechung
WO2020081247A1 (en) Potentiated antibiotic compositions and methods of use for treating bacterial infections and biofilms
CA3168188A1 (en) Compositions for disrupting biofilm formation and for treating biofilm-related disorders
WO2019077211A1 (fr) Utilisation de la cloxacilline pour inhiber/ empecher la formation de biofilm
US20210338637A1 (en) Indole derivatives for biofilm disruption and inhibition
US10905691B2 (en) Use of triazolo(4,5-d)pyrimidine derivatives for prevention and treatment of bacterial infection
WO2020193409A1 (fr) Utilisation de la gentamicine pour inhiber/ empecher la formation de biofilm bactérien
US20210369675A1 (en) Antimicrobial drug methods of use & therapeutic compositions
EP3989990A1 (de) Bakterium der familie christellaceae und diese enthaltende zusammensetzung zur vorbeugung und/oder behandlung von pathologischem muskelschwund oder einer krankheit, die durch pathologischen muskelschwund gekennzeichnet ist
AU2019201479A1 (en) Use of peptides in antibiotic resistance
WO2023038522A1 (en) Compositions and methods for treating biofilm disorders and infection
EP4398891A1 (de) Zusammensetzungen und verfahren zur behandlung von biofilmerkrankungen und -infektionen
Andersen et al. Pivmecillinam in the treatment of therapy resistant urinary tract infections: A comparison with pivmecillinam, pivampicillin and their combination
WO2003045390A1 (en) A METHOD OF TREATING BACTERIAL INFECTIONS USING GEMIFLOXACIN OR A SALT THEREOF AND A β -LACTAM ANTIBIOTIC
FR2812549A1 (fr) Associations dalfopristine/quinupristine avec le cefpirome

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: UNKNOWN

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20200403

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20230201