WO2020198834A1 - Treatment of microbial infections using scpppq1 proteins and derived peptides - Google Patents
Treatment of microbial infections using scpppq1 proteins and derived peptides Download PDFInfo
- Publication number
- WO2020198834A1 WO2020198834A1 PCT/CA2019/051839 CA2019051839W WO2020198834A1 WO 2020198834 A1 WO2020198834 A1 WO 2020198834A1 CA 2019051839 W CA2019051839 W CA 2019051839W WO 2020198834 A1 WO2020198834 A1 WO 2020198834A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- peptide
- peptidomimetic
- salt
- polypeptide
- seq
- Prior art date
Links
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61L—METHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
- A61L2/00—Methods or apparatus for disinfecting or sterilising materials or objects other than foodstuffs or contact lenses; Accessories therefor
- A61L2/0005—Methods or apparatus for disinfecting or sterilising materials or objects other than foodstuffs or contact lenses; Accessories therefor for pharmaceuticals, biologicals or living parts
- A61L2/0082—Methods or apparatus for disinfecting or sterilising materials or objects other than foodstuffs or contact lenses; Accessories therefor for pharmaceuticals, biologicals or living parts using chemical substances
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K8/00—Cosmetics or similar toiletry preparations
- A61K8/18—Cosmetics or similar toiletry preparations characterised by the composition
- A61K8/30—Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds
- A61K8/64—Proteins; Peptides; Derivatives or degradation products thereof
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61L—METHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
- A61L2/00—Methods or apparatus for disinfecting or sterilising materials or objects other than foodstuffs or contact lenses; Accessories therefor
- A61L2/16—Methods or apparatus for disinfecting or sterilising materials or objects other than foodstuffs or contact lenses; Accessories therefor using chemical substances
- A61L2/18—Liquid substances or solutions comprising solids or dissolved gases
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61L—METHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
- A61L2/00—Methods or apparatus for disinfecting or sterilising materials or objects other than foodstuffs or contact lenses; Accessories therefor
- A61L2/16—Methods or apparatus for disinfecting or sterilising materials or objects other than foodstuffs or contact lenses; Accessories therefor using chemical substances
- A61L2/23—Solid substances, e.g. granules, powders, blocks, tablets
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P1/00—Drugs for disorders of the alimentary tract or the digestive system
- A61P1/02—Stomatological preparations, e.g. drugs for caries, aphtae, periodontitis
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/04—Antibacterial agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/10—Antimycotics
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61Q—SPECIFIC USE OF COSMETICS OR SIMILAR TOILETRY PREPARATIONS
- A61Q11/00—Preparations for care of the teeth, of the oral cavity or of dentures; Dentifrices, e.g. toothpastes; Mouth rinses
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61Q—SPECIFIC USE OF COSMETICS OR SIMILAR TOILETRY PREPARATIONS
- A61Q17/00—Barrier preparations; Preparations brought into direct contact with the skin for affording protection against external influences, e.g. sunlight, X-rays or other harmful rays, corrosive materials, bacteria or insect stings
- A61Q17/005—Antimicrobial preparations
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/46—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
- C07K14/47—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/46—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
- C07K14/47—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
- C07K14/4701—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
- C07K14/4723—Cationic antimicrobial peptides, e.g. defensins
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61L—METHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
- A61L2101/00—Chemical composition of materials used in disinfecting, sterilising or deodorising
- A61L2101/32—Organic compounds
- A61L2101/46—Macromolecular compounds
-
- Y—GENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
- Y02—TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
- Y02A—TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
- Y02A50/00—TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
- Y02A50/30—Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change
Definitions
- the present disclosure generally relates to the field of antimicrobial agents, and to the treatment of bacterial or fungal infections such as Porphyromonas gingivalis (P. gingivalis) and Candida albicans infections, and related diseases.
- bacterial or fungal infections such as Porphyromonas gingivalis (P. gingivalis) and Candida albicans infections, and related diseases.
- Antibiotics are commonly used in the treatment and/or prevention of infectious diseases caused by various microorganisms such as bacteria.
- Most antibiotics are compounds derived from plants or fungus, which are produced as a defense mechanism against bacterial infection.
- resistance of bacteria against these antibiotics often occurs.
- conventional antibiotics kill bacteria by binding to specific targets that are involved in bacterial DNA and protein synthesis. Therefore, resistance can occur when bacteria modify these targets, such that antibiotics do not bind to these proteins, or when bacteria produce specific enzymes that inactivate the antibiotics.
- Pathogenic bacteria are usually found on or surrounding the surfaces of epithelial cells forming an epithelial barrier. If the epithelial barrier is compromised, the bacteria can penetrate into deeper tissue layers where they can expand and trigger infectious diseases. For instance, periodontal diseases (PD) affect a large segment of the population and their severity increases with age and eventually lead to tooth and bone loss.
- PD periodontal diseases
- a specialized basal lamina attaches epithelial cells to mineralized surfaces rather than connective tissue, thereby creating secluded environments that are critical for mineralization and for protection of the tooth supporting tissues from the aggressive oral environment.
- the junctional epithelium is a specialized portion of the gingiva that seals off the tooth supporting tissues from the oral environment.
- the JE uniquely attaches to the tooth via the sBL, where are localized three secreted proteins namely amelotin (AMTN), odontogenic ameloblast-associated (ODAM) and secretory calcium-binding phosphoprotein- prolineglutamine-rich 1 (SCPPPQ1) that are expressed by the JE.
- Laminin-332 (Lm332)
- JE Laminin-332
- their susceptibility to bacteria and/or bacterial products e.g . , enzymes
- the human subgingival plaque harbors more than 700 bacterial species. Some of them lead to PD when the equilibrium of the oral microbiome is compromised. Among these bacterial species, Porphyromonas gingivalis (P. gingivalis) and Treponema denticola ( T . denticola) are strongly linked to PD. These bacteria secrete a variety of enzymes that are generally associated with destruction of subepithelial connective tissue. There is actually no cure and no reliable manner of controlling bacteria during PD.
- peri-implant mucositis This condition involves the presence of inflammation in the mucosa at an implant but with no signs of loss of supporting bone.
- peri-implantitis which is caused by the colonization of bacteria of the implant's surface. Inflammation in the bone surrounding the implant then causes loss of bone which ultimately may lead to failure of the implant.
- Peri-implantitis can stem from an existing periodontitis infection or patients can develop peri-implantitis without a previous history of periodontitis.
- gingivalis has been shown to induce platelet aggregation which lead to thrombus formation during cardiovascular disease.
- atherosclerotic plaques are commonly colonized with P. gingivalis, suggesting the involvement of these bacteria in atherosclerosis and myocardial infarction.
- P. gingivalis is also associated with bacterial pneumonia resulting from aspiration of oropharyngeal flora into the lower respiratory tract. More specifically, the production of gingipains by these bacteria is known to impact the innate immune response and promote chronic inflammation (Benedyk et al., Journal of Innate Immunity 2016; 8, No. 2: 185-198).
- Candida species such as Candida albicans can causes oral candidiasis (oral thrush).
- azoles such as fluconazole launched in the mid-1980s
- polyenes such as amphotericin B launched in the mid-1950s
- echinocandins such as caspofungin launched in early 2000.
- systemic antifungals such as amphotericin B tend to have relatively high toxicity and side effects
- echinocandins which also have narrow antifungal spectrum of activity.
- the present disclosure provides the following items:
- XI is phenylalanine or an analog thereof, or is absent;
- X2 is proline or an analog thereof
- X3 is leucine or an analog thereof, or is absent
- X4 is glutamine or an analog thereof, or is absent
- X5 is glutamine, proline, leucine or an analog thereof, or is absent;
- X6 is proline or an analog thereof
- X7 is glutamine or an analog thereof, or is absent
- X8 is alanine or an analog thereof, or is absent
- X9 is proline or an analog thereof
- X10 is any amino acid or amino acid analog, or is absent;
- X12 is leucine or an analog thereof, or is absent
- X13 is proline or an analog thereof, or is absent
- X14 is leucine or an analog thereof, or is absent
- X15 is proline or an analog thereof, or is absent
- X16 is isoleucine, leucine, or an analog thereof, or is absent;
- X17 is proline, glutamine, or an analog thereof, or is absent;
- X18 is isoleucine, leucine, phenylalanine, or an analog thereof, or is absent;
- X19 is proline or an analog thereof, or is absent
- the isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of items 1 to 27, which comprises a sequence of at least 7 or 8 residues from one of the following sequences: FPLPPQPP (SEQ ID NO:1), FPLPQAPLIPIP (SEQ ID NO:2), FPQPPQLQIP (SEQ ID NO:3), PLPPSPP (SEQ ID NO:4), FPFQQPPLIPFP (SEQ ID NO:5), FPPPVGLPLIPLP (SEQ ID NO:6), FPPPPQIPIP (SEQ ID NO:7), FPLPRQIPIP (SEQ ID NO:8), FPPPPQLPLIPIP (SEQ ID NO:9), FPLPQAPLIPIP (SEQ ID NO:10), or a variant thereof having at least 70% sequence identity with the at least 7 or 8 residues.
- FPLPPQPP SEQ ID NO:1
- FPLPQAPLIPIP SEQ ID NO:2
- FPLPPQPP SEQ ID NO: 1
- FPLPQAPLIPIP SEQ ID NO:2
- FPQPPQLQIP SEQ ID NO:3
- PLPPSPP SEQ ID NO:4
- polypeptide, peptide, peptidomimetic or salt thereof according to item 29, wherein the polypeptide, peptide or peptidomimetic comprises the sequence FPLPPQPP (SEQ ID NO: 1) or FPLPQAP (SEQ ID NO: 1 1), or a variant thereof having at least 70% sequence identity with these sequences.
- polypeptide, peptide or peptidomimetic comprises the sequence FPLPPQPP (SEQ ID NO: 1 ) or FPLPQAP (SEQ ID NO:1 1 ).
- the isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of items 1 to 31 , which comprises at least 10 contiguous amino acids from the amino acid sequences of any one of SEQ ID NOs: 12-29 and 62.
- the isolated polypeptide, peptide, peptidomimetic or salt thereof according to item 32 comprises an amino acid sequence having at least 70% sequence identity with the sequences of any one of SEQ ID NOs: 12-29 and 62.
- the isolated polypeptide, peptide, peptidomimetic or salt thereof according to item 33 comprises with the amino acid sequences of any one of SEQ ID NOs: 12-29 and 62.
- a vector comprising the nucleic acid of item 35.
- a host cell comprising nucleic acid of item 35 or the vector of item 36.
- composition comprising the isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of items 1 to 34, and one or more excipients.
- composition of item 38, wherein the one or more excipients comprise a biodegradable polymer.
- composition of item 38 or 39 which is in the form of a cream, a gel, a paste, a solution, a chewing gum, a patch, a film, or a strip.
- a method for treating a bacterial or fungal infection in a subject comprising administering to the subject an effective amount of the polypeptide, peptide, peptidomimetic or salt thereof according to any one of items 1 to 34, or the composition of any one of items 38 to 40.
- 50 The method of item 49, wherein the Gram+ bacteria are of the Bacillus subtilis species.
- 51 A method for treating a periodontal or gingival disease in a subject in need thereof, said method comprising contacting the gum from the subject with an effective amount of the polypeptide, peptide, peptidomimetic or salt thereof according to any one of items 1 to 34, or the composition of any one of items 38 to 40.
- a method for cleaning or disinfecting a surface or an object comprising contacting the surface or the object with an effective amount of the polypeptide, peptide, peptidomimetic or salt thereof according to any one of items 1 to 34, or the composition of any one of items 38 to 40.
- polypeptide, peptide, peptidomimetic or salt thereof according to any one of items 1 to 34, or the composition of any one of items 38 to 40 for treating a bacterial or fungal infection in a subject.
- the fungal infection is a yeast infection.
- yeast is of the Candida genus.
- yeast is of the Candida albicans species.
- Gram- bacteria are of the Porphyromonas, Prevotella, Fusobacterium, Escherichia and/or Treponema genus.
- Gram- bacteria are of the Porphyromonas gingivalis, Fusobacterium nucleatum, Prevotella intermedia, Escherichia coli and/or Treponema denticola species.
- Gram+ bacteria are of the Bacillus genus.
- polypeptide, peptide, peptidomimetic or salt thereof according to any one of items 1 to 34, or the composition of any one of items 38 to 40 for treating a periodontal or gingival disease in a subject.
- 67. The polypeptide, peptide, peptidomimetic, salt or composition for use according to item 66, wherein the composition is the composition of item 40.
- polypeptide, peptide, peptidomimetic or salt thereof according to any one of items 1 to 34, or the composition of any one of items 38 to 40 for cleaning or disinfecting a surface or an object.
- polypeptide, peptide, peptidomimetic or salt thereof according to any one of items 1 to 34, or the composition of any one of items 38 to 40 for use i) for treatment and prevention of infectious diseases (e.g., bacterial or fungal infections in the eyes, mouth, wounds, and brain); ii) for treatment and prevention of post-operation bacterial or fungal infections and systemic propagations (e.g., implants such as dental or orthopedic implants); iii) for improvement of postoperation wound healing (e.g. around implants); iv) for inhibition of P.
- infectious diseases e.g., bacterial or fungal infections in the eyes, mouth, wounds, and brain
- post-operation bacterial or fungal infections and systemic propagations e.g., implants such as dental or orthopedic implants
- post-operation wound healing e.g. around implants
- iv) for inhibition of P for inhibition of P.
- gingivalis and/or other periodontal disease-associated pathogens and treatment of its associated diseases including but not limited to Alzheimer's disease, cardiovascular disease, and bacterial pneumonia; v) treatment and prevention of periodontal disease; vi) for surface sterilization or antimicrobial/antifungal surface coating in industry (e.g., food industry) and hospital applications (e.g., surgical tools, medical devices, and areas); vii) as antibacterial/antifungal solutions (e.g.
- viii) as health products e.g., antibacterial/antifungal toothpaste, creams, drops, mouthwash
- ix as preservative in food products, beverages, cosmetics (lotions, creams, gels, soaps, shampoos, conditioners, antiperspirants) contact lens products, and food ingredients.
- FIGs. 1A-B are representative pictures of western blot analysis following in vitro digestion assay of AMTN, ODAM and SCPPPQ1 by (FIG. 1A) A. actinomycetemcomitans or F. nucleatum showing no degradation of the proteins, while digestion assay by (FIG. 1 B) more aggressive bacteria as T. denticola, P. gingivalis and P. intermedia degrade AMTN and ODAM but not SCPPPQ1.
- FIGs. 2A-C illustrate graphs showing the results of top-down mass spectrometry of AMTN (FIG. 2A, SEQ ID NO: 63), ODAM (FIG. 2B, SEQ ID NO: 64) and SCPPPQ1 (FIG.
- FIG. 3 illustrates representative pictures of scanning electron microscope (SEM) of P. gingivalis after incubation (top left) with buffer only (i.e. control) (top right) with ODAM protein and (bottom left) with the rat SCPPPQ1 protein (SEQ ID NO: 31).
- SEM scanning electron microscope
- FIG. 4 illustrates two representative examples of SEM images of P. gingivalis after incubation (left top and bottom) without protein (i.e. control), (middle top and bottom) with rat SCPPPQ1 (rSCPPPQI) protein, and (right top and bottom) with human SCPPPQ1 (hSCPPPQI) protein.
- rSCPPPQI rat SCPPPQ1
- hSCPPPQI human SCPPPQ1
- FIG. 5A illustrates representative transmission electron microscope (TEM) images of P. gingivalis, to assess the structural appearance of the bacterial membrane, after incubation with buffer only (i.e. control) (top, control), with rat SCPPPQ1 (bottom).
- TEM transmission electron microscope
- FIG. 5B illustrates representative TEM images of P. gingivalis following a two-hour incubation without protein (left, control) and with human SCPPPQ1 (right) on Porphyromonas gingivalis.
- FIG. 6A is a graph showing a comparison of the total number of bacteria following incubation with buffer only (control) or with human SCPPPQ1 (hSCPPPQI) or rat SCPPPQ1 (rSCPPPQI) after counting bacteria from SEM micrographs.
- FIG. 6B are graphs showing a comparison of the effects of the rSCPPPQI and hSCPPPQI proteins on the formation of aggregates as assessed by flow cytometry (FACS).
- FIG. 7 illustrates representative immunofluorescence microscopy images of P. gingivalis, assessing the localization of SCPPPQ1 after incubation (top) with buffer only (i.e. control), and (bottom) with the human SCPPPQ1 (hSCPPPQI) protein (SEQ ID NO: 30).
- FIG. 8 illustrates representative immunofluorescence microscopy images of P. gingivalis, assessing the localization of SCPPPQ1 , after incubation (top) without protein (i.e. control), and (bottom) with rat SCPPPQ1 (rSCPPPQI) protein.
- FIG. 9 illustrates representative TEM images of P. gingivalis, to assess the localization of rat SCPPPQ1 (rSCPPPQI) in colloidal gold immuno-labeled preparations for rSCPPPQI protein.
- FIG. 10 illustrates representative transmission (top) and scanning electron (bottom) micrographs of P. gingivalis highlighting the network of rat SCPPPQ1.
- FIG. 11 depicts representative fluorescence images of P. gingivalis incubated with increasing concentrations of rat SCPPPQ1.
- FIG. 12 depicts the sequence of (i) the predicted human SCPPPQ1 protein, (ii) the actual human SCPPPQ1 protein, (iii) the rat SCPPPQ1 protein (SEQ ID NO: 31), and (iv)-(xvii) of peptides derived therefrom.
- FIGs. 13A-B show representative SEM images of P. gingivalis to assess the structural appearance of bacteria after incubation with the different peptides depicted in FIG. 12.
- FIG. 13C is a graph showing a comparison of the effects of the peptides on the formation of aggregates visualised using SEM (FIGs. 13A-B).
- FIG. 13D-E are graphs showing a comparison of the effects of the peptides on the bacterial membranes using SEM (FIGs. 13A-B).
- FIG. 13D comparison of the percentage of bacteria that are undamaged or damaged.
- FIG. 13E percentage of undamaged membrane or the two different type of damaged membranes (holes or collapsed).
- FIG. 14A illustrate an alignment of the confirmed or predicted amino acid sequences of the SCPPPQ1 protein from several species including human, rat, mouse, chimp, gorilla, baboon, macaque, vervet, gibbon, elephant, squirrel, rabbit, Chinese hamster, bovine, sheep, cat, dog, horse, and pig.
- the sequences corresponding to the signal peptides are underlined, and the domains corresponding to residues 48-55 of the confirmed human SCPPPQ1 protein (referred to as peptide #5 in subsequent figures) are in bold.
- FIG. 14B illustrates the sequences of the domains corresponding to residues 48-55 of the confirmed human SCPPPQ1 protein depicted in FIG. 14A.
- FIG. 15 shows representative TEM images of P. gingivalis to assess the structural appearance of the bacterial membrane after incubation with buffer only (i.e. control), and with the different peptides depicted in FIG. 12.
- FIG. 16 shows representative SEM images of Fusobacterium nucleatum with buffer only (i.e. control) or in presence of rat ODAM or rat SCPPPQ1.
- FIG. 17 shows representative SEM images of Prevotella intermedia with buffer only (i.e. control) or in presence of rat ODAM or rat SCPPPQ1 .
- FIG. 18 shows representative SEM images of Treponema denticola with buffer only (i.e. control) or in presence of rat SCPPPQ1.
- FIG. 19A shows representative fluorescence images of Bacillus subtilis with buffer only (i.e. control) or in presence of rat SCPPPQ1.
- FIG. 19B shows representative SEM images of Bacillus subtilis with buffer only (i.e. control) or in presence of rat SCPPPQ1 (rSCPPPQI) or human SCPPPQ1 (hSCPPPQI).
- FIG. 20A shows representative fluorescence images of Bacillus thuringiensis with buffer only (i.e. control) or in presence of rat SCPPPQ1.
- FIG. 20B shows representative SEM images of Bacillus thuringiensis with buffer only (i.e. control) or in presence of rSCPPPQI or hSCPPPQI .
- FIG. 21 A shows representative fluorescence images of Escherichia coli with buffer only (i.e. control) or in presence of rat SCPPPQ1 (rSCPPPQI).
- FIG. 21 B shows representative SEM images of Escherichia coli with buffer only (i.e. left, control) or in presence of rat SCPPPQ1 (rSCPPPQI) or human SCPPPQ1 (hSCPPPQI).
- FIG. 22 shows representative SEM images of P. gingivalis with buffer only or water only (i.e. control) of kanamycin, rat SCPPPQ1 (rSCPPPQI) or human SCPPPQ1 (hSCPPPQI).
- FIG. 23A shows representative SEM images of Candida albicans with buffer only (i.e. control) or in the presence of SCPPPQ1 (human or rat) and of peptides #5, #8 and #10.
- FIG. 23B shows a graph of the number of Candida albicans in the presence (dark) or absence (grey) of hyphae depending of the protein or peptides used.
- FIG. 23C shows various Candida albicans strains (top) grown in culture dishes in presence or not of rat SCPPPQ1 and a summary table (bottom) of the number of Unit Forming Colony (UFC) and their appearances.
- FIG. 24 shows an analysis of the effect of rat SCPPPQ1 on LS8 cells (ameloblast-like cell line).
- FIG. 25 shows the effect of toothpaste with buffer only (i.e. control) or rat SCPPPQ1 on Porphyromonas gingivalis as assessed by SEM (top).
- FIG. 26 shows the effect of the absorption of rat SCPPPQ1 or human SCPPPQ1 on titanium surfaces on Porphyromonas gingivalis as assessed by SEM.
- the term “about” has its ordinary meaning.
- the term“about” is used to indicate that a value includes an inherent variation of error for the device or the method being employed to determine the value, or encompass values close to the recited values, for example within 10% of the recited values (or range of values).
- rat and human SCPPPQ1 proteins interfere with the membrane of bacteria and fungi, and in turn exhibit antibacterial and antifungal activities.
- the known or predicted amino acid sequences of SCPPPQ1 from various species exhibit significant sequence homology/identity with rat and human SCPPPQ1 proteins.
- the present disclosure provides an isolated SCPPPQ1 protein or a fragment thereof of at least 5, 6 or 7 amino acids, and the use of such protein or fragment as an antibacterial or antifungal agent.
- the present disclosure also provides an isolated polypeptide, peptide or peptidomimetic of 5, 6 or 7 to 100 residues comprising a sequence of formula I:
- XI is L-phenylalanine, D-phenylalanine, or an analog thereof, or is absent;
- X2 is L-proline, D-proline, or an analog thereof
- X3 is L-leucine, D-leucine, L-phenylalanine, D-phenylalanine, or an analog thereof, or is absent;
- X4 is L-glutamine, D-glutamine, or an analog thereof, or is absent;
- X5 is L-glutamine, D-glutamine, L-proline, D-proline, L-leucine, D-leucine or an analog thereof, or is absent;
- X6 is L-proline, D-proline, or an analog thereof
- X7 is L-glutamine, D-glutamine, or an analog thereof, or is absent;
- X8 is L-alanine, D-alanine, or an analog thereof, or is absent;
- X9 is L-proline, D-proline, or an analog thereof
- X10 is any amino acid or amino acid analog, preferably L-glutamine, D-glutamine, L-leucine, D-leucine, L-serine or D-serine, L-valine or D-valine, L-proline, D-proline, or an analog thereof, or is absent;
- X12 is L-leucine, D-leucine, or an analog thereof, or is absent;
- X13 is L-proline, D-proline, or an analog thereof, or is absent;
- X14 is L-leucine, D-leucine, or an analog thereof, or is absent;
- X15 is L-proline, D-proline, or an analog thereof, or is absent;
- X16 is L-isoleucine, D-isoleucine, L-leucine, D-leucine, or an analog thereof, or is absent;
- X17 is L-proline, D-proline, L-glutamine or D-glutamine, or an analog thereof, or is absent;
- X18 is L-isoleucine, D-isoleucine, L-leucine, D-leucine, L-phenylalanine, D- phenylalanine or an analog thereof, or is absent;
- X19 is L- or D-proline, or an analog thereof, or is absent;
- X1 is L-phenylalanine, D-phenylalanine, or an analog thereof, preferably L-phenylalanine.
- X2 is L-proline, D-proline, or an analog thereof, preferably L-proline.
- X3 is L-leucine, D-leucine, or an analog thereof, preferably L- leucine.
- X4 is absent.
- X5 is absent.
- X6 is L-proline or D-proline, preferably L-proline.
- X7 is L-glutamine or D-glutamine, preferably L-glutamine. In another embodiment, X7 is absent.
- X8 is L-alanine or D-alanine, preferably L-alanine. In another embodiment, X8 is absent.
- X9 is L-proline or D-proline, preferably L-proline.
- X10 is L-glutamine or D-glutamine, preferably L-glutamine. In another embodiment, X10 is absent.
- X1 1 , X12, X13, X14 and/or X15 are absent. In another embodiment, X1 1 , X12, X13, X14 and X15 are absent.
- X16 is L-isoleucine, D-isoleucine, L-leucine or D-leucine, preferably L-isoleucine or L-leucine. In another embodiment, X16 is absent.
- X17 is L-proline, D-proline, L-glutamine or D-glutamine, preferably L-proline or D-proline, more preferably L-proline.
- X18 is L-isoleucine, D-isoleucine, L-leucine, D-leucine, L- phenylalanine, or D-phenylalanine, preferably L-isoleucine, L-leucine or L-phenylalanine. In another embodiment, X18 is absent.
- X19 is L-proline or D-proline, preferably L-proline.
- polypeptide or peptide is not a naturally-occurring polypeptide or peptide.
- the polypeptide or peptide does not consist of the sequence of SEQ ID NO:30. In an embodiment, the polypeptide or peptide does not consist of the sequence of SEQ ID NO:31 . In an embodiment, the polypeptide or peptide does not consist of the sequence of SEQ ID NO: 1 . In an embodiment, the polypeptide or peptide does not only comprise a naturally-occurring sequence, i.e. comprises at least one amino acid deletion, addition, substitution and/or modification (e.g., amidation, conjugation to a moiety, etc.).
- polypeptide is intended to encompass a chain of more than 50 amino acids, and up to 500, 1000, 2000, 3000, 5000 or more amino acids.
- the term further includes polypeptides which have undergone post-translational modifications, for example, glycosylation, acetylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, or modification by non-naturally occurring amino acids.
- peptide refers to a chain of no more than 50 amino acids, preferably of no more than 30 amino acids.
- peptidomimetic refers to a molecule comprising a plurality of amino acid residues (naturally- and/or non-naturally-occurring amino acids, amino acid analogs) joined by a plurality of peptide and/or non-peptide bonds. Peptidomimetics typically retain the polarity, three-dimensional size and functionality (bioactivity) of their peptide equivalents, but one or more of the peptide bonds/linkages have been replaced, often by more stable linkages.
- the bond which replaces the amide bond conserves many or all of the properties of the amide bond, e.g. conformation, steric bulk, electrostatic character, potential for hydrogen bonding, etc.
- Typical peptide bond replacements include esters, polyamines and derivatives thereof as well as substituted alkanes and alkenes, such as aminomethyl and ketomethylene.
- Such peptidomimetics may have greater chemical stability, enhanced biological/pharmacological properties (e.g., half-life, absorption, potency, efficiency, etc.) and/or reduced antigenicity relative its peptide equivalent.
- in the domain of formula I represents a peptide bond.
- amino acid includes both L- and D-isomers of the naturally occurring amino acids as well as other amino acids (e.g., naturally-occurring amino acids, non- naturally-occurring amino acids, amino acids which are not encoded by nucleic acid sequences, etc.) used in peptide chemistry to prepare synthetic analogs of peptides.
- naturally- occurring amino acids are glycine, alanine, valine, leucine, isoleucine, serine, threonine, etc.
- Other amino acids include for example non-genetically encoded forms of amino acids, as well as a conservative substitution of an L-amino acid.
- Naturally-occurring non-genetically encoded amino acids include, for example, beta-alanine, 3-amino-propionic acid, 2,3-diamino propionic acid, alpha-aminoisobutyric acid (Aib), 4-amino-butyric acid, /V-methylglycine (sarcosine), hydroxyproline, ornithine (e.g., L-ornithine), citrulline, f-butylalanine, f-butylglycine, N- methylisoleucine, phenylglycine, cyclohexylalanine, norleucine (Nle), norvaline, 2- napthylalanine, pyridylalanine, 3-benzothienyl alanine, 4-chlorophenylalanine, 2- fluorophenylalanine, 3-fluorophenylalanine, 4-fluorophenylalanine, penicillamine, 1 , 2,3,
- analog when used in reference to an amino acid refers to synthetic amino acids providing similar side chain functionality (i.e. , structurally similar) as the“native” amino acid and which can be substituted for an amino acid in the formation of a peptidomimetic.
- Amino acid analogs include, without limitation, b-amino acids and amino acids, in which the amino or carboxy group is substituted by a similarly reactive group or other groups (e.g., substitution of the primary amine with a secondary or tertiary amine, or substitution of the carboxy group with an ester).
- aromatic amino acids may be replaced with D- or L-naphthylalanine, D- or L-homophenylalanine, D- or L-phenylglycine, D- or L-2-thienylalanine, D- or L-1 -, 2-, 3-, or 4- pyrenylalanine, D- or L-3-thienylalanine, D- or L-(2-pyridinyl)-alanine, D- or L-(3-pyrid inyl)- alanine, D- or L-(2-pyrazinyl)-alanine, D- or L-(4-isopropyl)-phenylglycine, D-(trifluoromethyl)- phenylglycine, D-(trifluoromethyl)-phenylalanine, D-p-fluorophenylalanine, D- or L-p- biphenylalanine D-or L-p-methoxybiphenylalanine,
- Analogs of alanine include b-alanine, aminoisobutyric acid (a or b), methylalanine and f-butylalanine.
- Analogs of phenylalanine include b-methyl-phenylalanine, b-hydroxyphenylalanine, a- methyl-3-methoxy-DL-phenylalanine, a-methyl-D-phenylalanine, a-methyl-L-phenylalanine, 2,4- dichloro-phenylalanine, 2-(trifluoromethyl)-D-phenylalanine, 2-(trifluoromethyl)-L-phenylalanine, 2-bromo-D- phenylalanine, 2-bromo-L-phenylalanine, 2-chloro-D-phenylalanine, 2-chloro-L- phenylalanine, 2-cyano-D-phenylalanine, 2-cyano-L-phenylalanine, 2-fluoro-D-phenylalanine, 2- fluoro-L-phenylalanine, 2-methyl-D-phenylalanine, 2-methyl-L-phenylalanine, 2-nitro-D
- Examples of analogs of proline include hydroxyproline, ketoproline, trans/cis-3- hydroxyproline, trans/cis-4-hydroxyproline, trans/cis-3-aminoproline, trans/cis-4-aminoproline, methylproline, 3,4-dehydroproline, L-pipecolic acid, 3-thiaproline and 4-thiaproline.
- Example of analogs of glutamine include acivicin, antipyrimidine, 6-diazo-5-oxo-L- norleucine (DON), and theanine.
- polypeptide, peptide or peptidomimetic comprises only L- or D- amino acids. In a further embodiment, the polypeptide, peptide or peptidomimetic comprises only L-amino acids.
- the polypeptide, peptide, peptidomimetic or domain disclosed herein may include substitutions of functionally equivalent amino acid residues.
- one or more amino acid residues within the sequence can be substituted by another amino acid of a similar polarity (having similar physico-chemical properties) which acts as a functional equivalent, resulting in a silent alteration.
- Substitution for an amino acid within the sequence may be selected from other members of the class to which the amino acid belongs.
- positively charged (basic) amino acids include arginine, lysine and histidine (as well as homoarginine and ornithine).
- Nonpolar (hydrophobic) amino acids include leucine, isoleucine, alanine, phenylalanine, valine, proline, tryptophan and methionine.
- Uncharged polar amino acids include serine, threonine, cysteine, tyrosine, asparagine and glutamine.
- Negatively charged (acidic) amino acids include glutamic acid and aspartic acid.
- the amino acid glycine may be included in either the nonpolar amino acid family or the uncharged (neutral) polar amino acid family. Substitutions made within a family of amino acids are generally understood to be conservative substitutions.
- the polypeptide, peptide or peptidomimetic comprises a sequence of at least 5, 6, 7 or 8 residues from one of the following sequences: FPLPPQPP (SEQ ID NO: 1 ), FPLPQAPLIPIP (SEQ ID NO:2), FPQPPQLQIP (SEQ ID NO:3), PLPPSPP (SEQ ID NO:4), FPFQQPPLIPFP (SEQ ID NO:5), FPPPVGLPLIPLP (SEQ ID NO:6), FPPPPQIPIP (SEQ ID NO:7), FPLPRQIPIP (SEQ ID NO:8), FPPPPQLPLIPIP (SEQ ID NO:9), FPLPQAPLIPIP (SEQ ID NO: 10), or a variant thereof having at least 70% sequence identity with the at least 7 or 8 residues (e.g., having 1 or 2 amino acid substitution(s)).
- FPLPPQPP SEQ ID NO: 1
- FPLPQAPLIPIP SEQ ID NO:2
- the polypeptide, peptide or peptidomimetic comprises a sequence of at least 5, 6, 7 or 8 residues from any one of SEQ ID NOs: 1 -10, or a variant thereof having at least 85% sequence identity with the at least 5, 6, 7 or 8 residues (e.g., having 1 amino acid substitution).
- the polypeptide, peptide or peptidomimetic comprises or consists of a sequence of at least 5, 6, 7 or 8 residues from any one of SEQ ID NOs: 1 -10.
- polypeptide, peptide or peptidomimetic comprises or consists of the sequences of any one of SEQ ID NOs: 1 10
- polypeptide, peptide or peptidomimetic comprises the sequence FPLPPQPP (SEQ ID NO: 1 ), FPLPQAP (SEQ ID NO: 1 1 ), or a variant thereof having at least 70% sequence identity with these sequences (e.g., having 1 or 2 amino acid substitution(s)).
- the polypeptide, peptide or peptidomimetic comprises the sequence FPLPPQPP (SEQ ID NO:1), FPLPQAP (SEQ ID NO: 1 1), FPLPPQPPRR (SEQ ID NO: 58), FPLPQAPRR (SEQ ID NO: 59), PPQVPPFFPQVLFPLPPQPP (SEQ ID NO: 60), or PPQVPPFFPQVLFPLPPQPPRR (SEQ ID NO: 61), or a variant thereof having at least 85% sequence identity with these sequences (e.g., having 1 amino acid substitution).
- the polypeptide, peptide or peptidomimetic comprises or consist the sequence FPLPPQPP (SEQ ID NO:1 ), FPLPQAP (SEQ ID NO:1 1 ), FPLPPQPPRR (SEQ ID NO: 58), FPLPQAPRR (SEQ ID NO: 59), PPQVPPFFPQVLFPLPPQPP (SEQ ID NO: 60), or PPQVPPFFPQVLFPLPPQPPRR (SEQ ID NO: 61).
- the polypeptide, peptide or peptidomimetic comprises or consist of the sequence FPLPPQPP (SEQ ID NO: 1).
- the polypeptide, peptide or peptidomimetic comprises or consist of the sequence FPLPQAP (SEQ ID NO: 1 1 ). In an embodiment, the polypeptide, peptide or peptidomimetic comprises or consist of the sequence FPLPPQPPRR (SEQ ID NO: 58). In an embodiment, the polypeptide, peptide or peptidomimetic comprises or consist of the sequence FPLPQAPRR (SEQ ID NO: 59). In an embodiment, the polypeptide, peptide or peptidomimetic comprises or consist of the sequence PPQVPPFFPQVLFPLPPQPP (SEQ ID NO: 60). In an embodiment, the polypeptide, peptide or peptidomimetic comprises or consist of the sequence PPQVPPFFPQVLFPLPPQPPRR (SEQ ID NO: 61).
- the above-mentioned polypeptide, peptide or peptidomimetic comprises, further to the sequence of formula I defined above, one more amino acids (naturally occurring or synthetic) covalently linked to the amino- and/or carboxy-termini of said domain.
- the polypeptide, peptide or peptidomimetic has 90 amino acids or less, 85 amino acid or less, 80 amino acid or less, 75 amino acids or less, or 70 amino acids or less.
- the above-mentioned polypeptide, peptide or peptidomimetic comprises at least 7 or 8 amino acids.
- the above-mentioned polypeptide, peptide or peptidomimetic comprises at least 9, 10, 1 1 , 12, 13, 14, 15, 20, 25 or 30 amino acids. In embodiments, the above-mentioned polypeptide, peptide or peptidomimetic comprises from 7 to 30, 25 or 20 amino acids.
- the polypeptide, peptide or peptidomimetic may comprise, e.g., 1 , 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40 or 50 additional amino acids at the N- and/or C-terminal end of the sequence of formula I defined above.
- the additional amino acid(s) at the N- and/or C-terminal end of the sequence of formula I defined above are/is one or more basic amino acids, e.g., arginine (Arg) or lysine (Lys).
- polypeptide, peptide or peptidomimetic comprise 1 to 10 or 1 to 5 additional basic amino acids at the N- and/or C- terminal end, preferably at the C-terminal end, of the sequence of formula I defined above.
- additional amino acid(s) at the N- and/or C-terminal end of the sequence of formula I defined above is/are one or more cysteine residues.
- the above-mentioned polypeptide, peptide or peptidomimetic comprises or consists of the sequence FPLPPQPPRR (SEQ ID NO: 58), FPLPQAPRR (SEQ ID NO: 59), PPQVPPFFPQVLFPLPPQPP (SEQ ID NO: 60), or PPQVPPFFPQVLFPLPPQPPRR (SEQ ID NO: 61 ).
- the above-mentioned polypeptide, peptide or peptidomimetic includes the above-noted sequence of formula I (e.g . , a sequence comprising at least 7 or 8 residues from the sequence of any one of SEQ ID NOs: 1-10 underlined below) and comprises at least 10, 15 or 20 contiguous amino acids from the amino acid sequences:
- the above-mentioned polypeptide, peptide or peptidomimetic includes the above-noted sequence of formula I and comprises at least 25, 30, 35, 40, 45, 50, 55, or 60 contiguous amino acids from the amino acid sequences of any one of SEQ ID NOs: 12-29.
- the contiguous amino acids are the amino acids immediately N- and/or C- terminal from the above-noted domain (underlined).
- the above-mentioned polypeptide, peptide or peptidomimetic includes the above-noted sequence of formula I and comprises an amino acid sequence having at least 70% sequence identity with the sequences of any one of SEQ ID NOs: 12-29.
- the above-mentioned polypeptide, peptide or peptidomimetic includes the above-noted sequence of formula I and comprises an amino acid sequence having at least 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with the sequences of any one of SEQ ID NOs: 12-29.
- the above-mentioned polypeptide, peptide or peptidomimetic comprises the amino acid sequence of any one of SEQ ID NOs: 12- 29. In another embodiment, the above-mentioned polypeptide, peptide or peptidomimetic does not comprises the sequences of the signal peptide depicted in FIG. 14A (underlined).
- N- and/or C-terminal amino acids of the above-mentioned polypeptide, peptide or peptidomimetic (or salt thereof) may be modified, for example by amidation, acetylation, acylation or any other modifications known in the art. Accordingly, in another aspect, the present disclosure provides a polypeptide, peptide or peptidomimetic (or salt thereof) of formula II:
- Z 1 is H (i.e. the peptide or peptidomimetic has a native NH 2 terminal) an amino-terminal modification; and Z 2 is OH (i.e. the peptide or peptidomimetic has a native COOH terminal) or a carboxy-terminal modification.
- the amino terminal residue i.e., the free amino group at the N-terminal end
- the peptide or peptidomimetic (or salt thereof) is modified ( e.g ., for protection against degradation), for example by covalent attachment of a moiety/chemical group (Z 1 ).
- the amino-terminal modification (Z 1 ) is a C1-C16 or C 3 -Ci 6 acyl group (linear or branched, saturated or unsaturated), in a further embodiment, a saturated Ci-C 6 acyl group (linear or branched) or an unsaturated C 3 -C 6 acyl group (linear or branched), in a further embodiment an acetyl group (CH 3 -CO-, Ac).
- the peptide or peptidomimetic (or salt thereof) has a native NH 2 terminal, i.e. Z 1 is H.
- the carboxy terminal residue (i.e., the free carboxy group at the C- terminal end of the peptide) of the peptide or peptidomimetic (or salt thereof) is modified (e.g. , for protection against degradation).
- the modification is an amidation (replacement of the OH group by a NH 2 group), thus in such a case Z 2 is a NH 2 group.
- polypeptide, peptide or peptidomimetic (or salt thereof) described herein may further comprise modifications that confer additional biological properties to the polypeptide, peptide or peptidomimetic such as protease resistance, plasma protein binding, increased half- life in vivo, binding to gum or teeth, intracellular penetration, etc.
- modifications include, for example, covalent attachment of fatty acids (e.g. , C 6 -Ci 8 ) to the polypeptide, peptide or peptidomimetic, attachment to proteins such as albumin (see, e.g., U.S. Patent No. 7,268, 1 13); glycosylation, biotinylation or PEGylation (see, e.g., U.S. Patent Nos.
- PEGylation may be carried out using an acylation reaction or an alkylation reaction with a reactive polyethylene glycol molecule.
- Methods for peptide PEGylation are disclosed, for example, in Roberts et al., Chemistry for peptide and protein PEGylation, Advanced Drug Delivery Reviews, Volume 64, Supplement, December 2012, Pages 1 16-127).
- the polypeptide, peptide, peptidomimetic or salt thereof is conjugated to a polyethylene glycol (PEG) chain/moiety (i.e. is PEGylated).
- PEG chain refers to polymers of ethylene glycol (represented by the general formula H(OCH 2 CH 2 ) n OH, where n is an integer of 2, 3, 4, 5, 6, 7, 8, 9, or more) which are commercially produced with different molecular weights (e.g., about 200-50,000 Da, 500-40,000 Da, 1000-30,000 Da or 2000-10,000 Da).
- PEGylated peptides/peptidomimetics may be prepared by modifying certain amino acids in the polypeptide, peptide, peptidomimetic with a suitable group-reactive reagent.
- a Cys side chain may be modified with a thiol-reactive agent, or a Lys side chain may be modified with an amine-reactive agent.
- polypeptide, peptide or peptidomimetic (or salt thereof) described herein is conjugated to a gum- or tooth-targeting agent, for example bisphosphonates (BP), diphosphoserine and pyrophosphate that have high affinity for hydroxyapatite (HAP), a major constituent of teeth.
- BP bisphosphonates
- HAP hydroxyapatite
- the above-mentioned polypeptide, peptide or peptidomimetic is in the form of a salt, e.g., a pharmaceutically acceptable salt.
- a pharmaceutically acceptable salt refers to salts of compounds that retain the biological activity of the parent compound, and which are not biologically or otherwise undesirable. Such salts can be prepared in situ during the final isolation and purification of the analog, or may be prepared separately by reacting a free base function with a suitable acid.
- Many of the peptides or peptidomimetics disclosed herein are capable of forming acid and/or base salts by virtue of the presence of amino and/or carboxyl groups or groups similar thereto.
- Acid addition salts may be prepared from inorganic and organic acids.
- Representative acid addition salts include, but are not limited to acetate, adipate, alginate, citrate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, camphorate, camphor sulfonate, decanoate, digluconate, glycerophosphate, hemisulfate, heptanoate, hexanoate, fumarate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethansulfonate (isothionate), lactate, maleate, methane sulfonate, nicotinate, 2-naphthalene sulfonate, octanoate, oxalate, palmitoate, pectinate, persulfate, 3-phenylpropionate, picrate, pivalate, propionate, succinate, tartrate, thiocyan
- Salts derived from inorganic acids include hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like.
- Salts derived from organic acids include acetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, malic acid, malonic acid, succinic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluene-sulfonic acid, salicylic acid, and the like.
- acids which can be employed to form pharmaceutically acceptable acid addition salts include, for example, an inorganic acid, e.g., hydrochloric acid, hydrobromic acid, sulphuric acid, and phosphoric acid, and an organic acid, e.g., oxalic acid, maleic acid, succinic acid, and citric acid.
- an inorganic acid e.g., hydrochloric acid, hydrobromic acid, sulphuric acid, and phosphoric acid
- organic acid e.g., oxalic acid, maleic acid, succinic acid, and citric acid.
- Basic addition salts also can be prepared by reacting a carboxylic acid-containing moiety with a suitable base such as the hydroxide, carbonate, or bicarbonate of a pharmaceutically acceptable metal cation or with ammonia or an organic primary, secondary, or tertiary amine.
- a suitable base such as the hydroxide, carbonate, or bicarbonate of a pharmaceutically acceptable metal cation or with ammonia or an organic primary, secondary, or tertiary amine.
- Pharmaceutically acceptable salts include, but are not limited to, cations based on alkali metals or alkaline earth metals such as lithium, sodium, potassium, calcium, magnesium, and aluminum salts, and the like, and nontoxic quaternary ammonia and amine cations including ammonium, tetramethylammonium, tetraethylammonium, methylammonium, dimethylammonium, trimethylammonium, triethylammonium, diethylammonium, and ethylammonium, amongst others.
- Other representative organic amines useful for the formation of base addition salts include, for example, ethylenediamine, ethanolamine, diethanolamine, piperidine, piperazine, and the like.
- Salts derived from organic bases include, but are not limited to, salts of primary, secondary and tertiary amines.
- the peptide or polypeptide described herein may be produced by expression in a host cell comprising a nucleic acid encoding the peptide or polypeptide (recombinant expression) or by chemical synthesis (e.g., solid-phase peptide synthesis).
- Polypeptides, peptides and peptidomimetics can be readily synthesized by manual and automated solid phase procedures well known in the art. Suitable syntheses can be performed for example by utilizing "t-Boc" or "Fmoc” procedures. Techniques and procedures for solid phase synthesis are described in for example Solid Phase Peptide Synthesis: A Practical Approach, by E. Atherton and R. C. Sheppard, published by IRL, Oxford University Press, 1989.
- polypeptides, peptides and peptidomimetics may be prepared by way of segment condensation, as described, for example, in Liu et al., Tetrahedron Lett. 37: 933-936, 1996; Baca et al., J. Am. Chem. Soc. 117: 1881 -1887, 1995; Tam et al., Int. J. Peptide Protein Res. 45: 209-216, 1995; Schnolzer and Kent, Science 256: 221 -225, 1992; Liu and Tam, J. Am. Chem. Soc. 116: 4149-4153, 1994; Liu and Tam, Proc. Natl. Acad. Sci.
- Polypeptides and peptides comprising naturally occurring amino acids encoded by the genetic code may also be prepared using recombinant DNA technology using standard methods. Polypeptides and peptides produced by recombinant technology may be modified (e.g., N-terminal acylation [e.g. , acetylation], C-terminal amidation), using methods well known in the art. Therefore, in embodiments, in cases where a polypeptide or peptide described herein contains naturally occurring amino acids encoded by the genetic code, the polypeptide or peptide may be produced using recombinant methods, and may in embodiments be subjected to for example the just-noted modifications (e.g., acylation, amidation). Accordingly, in another aspect, the present disclosure further provides a nucleic acid encoding the above-mentioned polypeptide or peptide. The disclosure also provides a vector or plasmid comprising the above- mentioned nucleic acid.
- the present disclosure provides a cell (e.g. , a host cell) comprising the above-mentioned nucleic acid and/or vector.
- a cell e.g. , a host cell
- the present disclosure further provides a recombinant expression system, vectors and host cells, such as those described above, for the expression/production of a polypeptide or peptide described herein, using for example culture media, production, isolation and purification methods well known in the art.
- polypeptide, peptide or peptidomimetic (or salt thereof) described herein can be purified by many techniques of polypeptide/peptide purification well known in the art, such as reverse phase chromatography, high performance liquid chromatography (HPLC), ion exchange chromatography, size exclusion chromatography, affinity chromatography, gel electrophoresis, and the like.
- HPLC high performance liquid chromatography
- ion exchange chromatography size exclusion chromatography
- affinity chromatography gel electrophoresis
- any antibody that specifically binds the peptide or peptidomimetic may for example be used.
- the present disclosure provides a composition
- a composition comprising the above-mentioned polypeptide, peptide, peptidomimetic or salt thereof and a carrier or excipient, in a further embodiment a pharmaceutically acceptable carrier or excipient.
- Supplementary active compounds can also be incorporated into the compositions.
- the carrier/excipient can be suitable, for example, for oral, mucosal, intravenous, parenteral, subcutaneous, intramuscular, intracranial, intraorbital, ophthalmic, intraventricular, intracapsular, intraspinal, intrathecal, epidural, intracisternal, intraperitoneal, intranasal or pulmonary (e.g., aerosol) administration.
- compositions may be prepared in a manner well known in the pharmaceutical art (see Remington: The Science and Practice of Pharmacy, by Loyd V Allen, Jr, 2012, 22 nd edition, Pharmaceutical Press; Handbook of Pharmaceutical Excipients, by Rowe et al., 2012, 7 th edition, Pharmaceutical Press).
- excipient has its normal meaning in the art and is any ingredient that is not an active ingredient (drug) itself. Excipients include for example binders, lubricants, diluents, fillers, thickening agents, disintegrants, plasticizers, coatings, barrier layer formulations, lubricants, stabilizing agent, release-delaying agents and other components. "Pharmaceutically acceptable excipient” as used herein refers to any excipient that does not interfere with effectiveness of the biological activity of the active ingredients and that is not toxic to the subject, i.e., is a type of excipient and/or is for use in an amount which is not toxic to the subject.
- Excipients are well known in the art, and the present system is not limited in these respects.
- the excipients including for example and without limitation, one or more binders (binding agents), buffers, thickening agents, surfactants, diluents, saline solution, release-delaying agents, colorants, flavoring agents, fillers, disintegrants/dissolution promoting agents, lubricants, plasticizers, silica flow conditioners, glidants, anti-caking agents, anti-tacking agents, stabilizing agents, anti-static agents, swelling agents and any combinations thereof.
- a single excipient can fulfill more than two functions at once, e.g., can act as both a binding agent and a thickening agent.
- these terms are not necessarily mutually exclusive.
- Useful diluents include, for example and without limitation, dicalcium phosphate, calcium diphosphate, calcium carbonate, calcium sulfate, lactose, cellulose, kaolin, sodium chloride, starches, powdered sugar, colloidal silicon dioxide, titanium oxide, alumina, talc, colloidal silica, microcrystalline cellulose, silicified micro crystalline cellulose and combinations thereof.
- Binder materials include, for example and without limitation, starches (including corn starch and pregelatinized starch), gelatin, sugars (including sucrose, glucose, dextrose and lactose), polyethylene glycol, povidone, waxes, and natural and synthetic gums, e.g.
- acacia sodium alginate polyvinylpyrrolidone, cellulosic polymers (e.g., hydroxypropyl cellulose, hydroxypropyl methylcellulose, methyl cellulose, hydroxyethyl cellulose, carboxymethylcellulose, colloidal silicon dioxide NF/EP (e.g., Cab-O-SilTM M5P), Silicified Microcrystalline Cellulose (SMCC), e.g., Silicified microcrystalline cellulose NF/EP (e.g., ProsolvTM SMCC 90), and silicon dioxide, mixtures thereof, and the like), veegum, and combinations thereof.
- colloidal silicon dioxide NF/EP e.g., Cab-O-SilTM M5P
- Silicified Microcrystalline Cellulose e.g., Silicified microcrystalline cellulose NF/EP (e.g., ProsolvTM SMCC 90)
- silicon dioxide mixtures thereof, and the like
- Useful lubricants include, for example, canola oil, glyceryl palmitostearate, hydrogenated vegetable oil (type I), magnesium oxide, magnesium stearate, mineral oil, poloxamer, polyethylene glycol, sodium lauryl sulfate, sodium stearate fumarate, stearic acid, talc and, zinc stearate, glyceryl behapate, magnesium lauryl sulfate, boric acid, sodium benzoate, sodium acetate, sodium benzoate/sodium acetate (in combination), DL-leucine, calcium stearate, sodium stearyl fumarate, mixtures thereof, and the like.
- Bulking agents include, for example: microcrystalline cellulose, for example, AVICEL® (FMC Corp.) or EMCOCEL ® (Mendell Inc.), which also has binder properties; dicalcium phosphate, for example, EMCOMPRESS ® (Mendell Inc.); calcium sulfate, for example, COMPACTROL ® (Mendell Inc.); and starches, for example, Starch 1500; and polyethylene glycols (CARBOWAX ® ).
- microcrystalline cellulose for example, AVICEL® (FMC Corp.) or EMCOCEL ® (Mendell Inc.)
- dicalcium phosphate for example, EMCOMPRESS ® (Mendell Inc.)
- calcium sulfate for example, COMPACTROL ® (Mendell Inc.)
- starches for example, Starch 1500
- CARBOWAX ® polyethylene glycols
- Disintegrating or dissolution promoting agents include: starches, clays, celluloses, alginates, gums, crosslinked polymers, colloidal silicon dioxide, osmogens, mixtures thereof, and the like, such as crosslinked sodium carboxymethyl cellulose (AC-DI-SOL ® ), sodium croscarmelose, sodium starch glycolate (EXPLOTAB ® , PRIMO JEL ® ) crosslinked polyvinylpolypyrrolidone (PLASONE-XL ® ), sodium chloride, sucrose, lactose and mannitol.
- AC-DI-SOL ® crosslinked sodium carboxymethyl cellulose
- EXPLOTAB ® sodium croscarmelose
- sodium starch glycolate EXPLOTAB ®
- PRIMO JEL ® PRIMO JEL ®
- PLASONE-XL ® crosslinked polyvinylpolypyrrolidone
- Antiadherents and glidants employable in the core and/or a coating of the solid oral dosage form may include talc, starches (e.g., cornstarch), celluloses, silicon dioxide, sodium lauryl sulfate, colloidal silica dioxide, and metallic stearates, among others.
- silica flow conditioners examples include colloidal silicon dioxide, magnesium aluminum silicate and guar gum.
- Suitable surfactants include pharmaceutically acceptable non-ionic, ionic and anionic surfactants.
- An example of a surfactant is sodium lauryl sulfate.
- the pharmaceutical composition to be administered may also contain minor amounts of nontoxic auxiliary substances such as wetting or emulsifying agents, pH-buffering agents and the like, for example, sodium acetate, sorbitan monolaurate, triethanolamine sodium acetate, triethanolamine oleate, etc.
- flavoring, coloring and/or sweetening agents may be added as well.
- stabilizing agents examples include acacia, albumin, polyvinyl alcohol, alginic acid, bentonite, dicalcium phosphate, carboxymethylcellulose (CMC), hydroxypropylcellulose (HPC), colloidal silicon dioxide, cyclodextrins, glyceryl monostearate, hydroxypropyl methylcellulose (HPMC), magnesium trisilicate, magnesium aluminum silicate, propylene glycol, propylene glycol alginate, sodium alginate, carnauba wax, xanthan gum, starch, stearate(s), stearic acid, stearic monoglyceride and stearyl alcohol.
- CMC carboxymethylcellulose
- HPC hydroxypropylcellulose
- HPMC hydroxypropyl methylcellulose
- magnesium trisilicate magnesium aluminum silicate
- propylene glycol propylene glycol alginate, sodium alginate, carnauba wax, xanthan gum, starch, stearate(s), stearic
- thickening agent can be for example talc USP/EP, a natural gum, such as guar gum or gum arabic, or a cellulose derivative such as microcrystalline cellulose NF/EP (e.g., AvicelTM PH 102), methylcellulose, ethylcellulose or hydroxyethylcellulose.
- a useful thickening agent is hydroxypropyl methylcellulose (HPMC), an adjuvant which is available in various viscosity grades.
- plasticizers include: acetylated monoglycerides; these can be used as food additives; Alkyl citrates, used in food packagings, medical products, cosmetics and children toys; Triethyl citrate (TEC); Acetyl triethyl citrate (ATEC), higher boiling point and lower volatility than TEC; Tributyl citrate (TBC); Acetyl tributyl citrate (ATBC), compatible with PVC and vinyl chloride copolymers; Trioctyl citrate (TOC), also used for gums and controlled release medicines; Acetyl trioctyl citrate (ATOC), also used for printing ink; Trihexyl citrate (THC), compatible with PVC, also used for controlled release medicines; Acetyl trihexyl citrate (ATHC), compatible with PVC; Butyryl trihexyl citrate (BTHC, trihexyl o-butyryl citrate), compatible with PVC; Trimethyl citrate (TMC), compatible with PVC;
- permeation enhancers examples include: sulphoxides (such as dimethylsulphoxide, DMSO), azones (e.g., laurocapram), pyrrolidones (for example 2-pyrrolidone, 2P), alcohols and alkanols (ethanol, or decanol), glycols (for example propylene glycol and polyethylene glycol), surfactants and terpenes.
- sulphoxides such as dimethylsulphoxide, DMSO
- azones e.g., laurocapram
- pyrrolidones for example 2-pyrrolidone, 2P
- alcohols and alkanols ethanol, or decanol
- glycols for example propylene glycol and polyethylene glycol
- surfactants examples include: terpenes.
- Formulations suitable for oral administration may include (a) liquid solutions, such as an effective amount of active agent(s)/composition(s) suspended in diluents, such as water, saline or PEG 400; (b) capsules, sachets or tablets, each containing a predetermined amount of the active ingredient, as liquids, solids, granules or gelatin; (c) suspensions in an appropriate liquid; and (d) suitable emulsions.
- liquid solutions such as an effective amount of active agent(s)/composition(s) suspended in diluents, such as water, saline or PEG 400
- capsules, sachets or tablets each containing a predetermined amount of the active ingredient, as liquids, solids, granules or gelatin
- suspensions in an appropriate liquid such as water, saline or PEG 400
- Tablet forms can include one or more of lactose, sucrose, mannitol, sorbitol, calcium phosphates, corn starch, potato starch, microcrystalline cellulose, gelatin, colloidal silicon dioxide, talc, magnesium stearate, stearic acid, and other excipients, colorants, fillers, binders, diluents, buffering agents, moistening agents, preservatives, flavoring agents, dyes, disintegrating agents, and pharmaceutically compatible carriers.
- Lozenge forms can comprise the active ingredient in a flavor, e.g., sucrose, as well as pastilles comprising the active ingredient in an inert base, such as gelatin and glycerin or sucrose and acacia emulsions, gels, and the like containing, in addition to the active ingredient, carriers known in the art.
- a flavor e.g., sucrose
- an inert base such as gelatin and glycerin or sucrose and acacia emulsions, gels, and the like containing, in addition to the active ingredient, carriers known in the art.
- Aqueous suspensions generally contain the active ingredient in finely powdered form or in the form of nano or micronized particles together with one or more suspending agents, such as sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethylcellulose, sodium alginate, polyvinyl-pyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents such as lecithin or condensation products of an alkylene oxide with fatty acids (for example polyoxethylene stearate), or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexito
- the aqueous suspensions may also contain one or more preservatives such as ethyl or propyl p-hydroxybenzoate; anti-oxidants such as ascorbic acid); coloring agents; flavoring agents; and/or sweetening agents such as sucrose, saccharine or aspartame
- Formulations for parenteral administration may, for example, contain excipients, sterile water, or saline, polyalkylene glycols such as polyethylene glycol, oils of vegetable origin, or hydrogenated napthalenes.
- Biocompatible, biodegradable lactide polymer, lactide/glycolide copolymer, or polyoxyethylene-polyoxypropylene copolymers may be used to control the release of the compounds.
- Other potentially useful parenteral delivery systems for compounds/compositions of the disclosure include ethylenevinyl acetate copolymer particles, osmotic pumps, implantable infusion systems, and liposomes.
- Formulations for inhalation may contain excipients, (e.g., lactose) or may be aqueous solutions containing, for example, polyoxyethylene-9-lauryl ether, glycocholate and deoxycholate, or may be oily solutions for administration in the form of nasal drops, or as a gel.
- excipients e.g., lactose
- aqueous solutions containing, for example, polyoxyethylene-9-lauryl ether, glycocholate and deoxycholate
- glycocholate and deoxycholate may be oily solutions for administration in the form of nasal drops, or as a gel.
- the above-mentioned polypeptide, peptide, peptidomimetic or salt thereof is formulated as a cream, a gel, a paste (e.g., toothpaste), a solution (e.g., a mouthwash), a chewing gum, a patch, a film, or a strip.
- a paste e.g., toothpaste
- a solution e.g., a mouthwash
- polypeptide, peptide, peptidomimetic or salt thereof may be incorporated into drug delivery systems for the oral cavity, such as bioadhesive formulations and nanoparticulate platforms, that typically comprise a biodegradable polymeric matrix, e.g., a mucoadhesive polymer such as gelatin, chitosan, ethylcellulose, carboxymethyl cellulose (CMC), crosslinked polyacrylic acid (PAA) polymers such as CarbopolTM (CP), poly(DL-lactide) (PLA), poly(lactic-coglycolic acid) (PLGA) (see, e.g., Sanko Nguyen & Marianne Hiorth, Therapeutic Delivery, (2015) 6(5), 595-608).
- a biodegradable polymeric matrix e.g., a mucoadhesive polymer such as gelatin, chitosan, ethylcellulose, carboxymethyl cellulose (CMC), crosslinked polyacrylic acid (PAA)
- the above-mentioned polypeptide, peptide, peptidomimetic or salt thereof is incorporated into a cleaning or detergent composition.
- cleaning or detergent composition may comprise other ingredients such as one or more surfactants (anionic, nonionic, ampholytic, zwitterionic and/or cationic surfactants), enzymes/proteases, stabilizers, conditioning polymers, cleaning polymers, surface modifying polymers, soil flocculating polymers, structurants, emollients, humectants, skin rejuvenating actives, magnesium cations, carboxylic acids, scrubbing particles, bleach and bleach activators, perfumes, malodor control agents, pigments, dyes, opacifiers, beads, pearlescent particles, microcapsules, antibacterial agents, pH adjusters, preservatives, buffering means or water or any other diluents or solvents compatible with the cleaning or detergent composition.
- surfactants anionic, nonionic, ampholytic, zwitterionic and/or
- polypeptide, peptide, peptidomimetic or salt thereof may be used for various applications including:
- infectious diseases e.g., bacterial or fungal infections in the eyes, mouth, wounds, and brain
- Treatment and prevention of post-operation bacterial or fungal infections and systemic propagations e.g., implants such as dental or orthopedic implants;
- Antibacterial/antifungal solutions e.g., hand gels, cleaning surfaces solution
- Health products e.g., antibacterial/antifungal toothpaste, creams, drops, mouthwash
- the present disclosure provides a method for preventing or treating a bacterial or fungal infection in a subject comprising administering to the subject an effective amount of the polypeptide, peptide, peptidomimetic or salt thereof (or composition comprising same) comprising a domain of formula I as defined herein.
- the present disclosure provides the use of the polypeptide, peptide, peptidomimetic or salt thereof (or composition comprising same) comprising a domain of formula I as defined herein for preventing or treating a bacterial or fungal infection in a subject.
- the present disclosure provides the use of the polypeptide, peptide, peptidomimetic or salt thereof (or composition comprising same) comprising a domain of formula I as defined herein for the manufacture of a medicament for preventing or treating a bacterial or fungal infection in a subject.
- the present disclosure provides the polypeptide, peptide, peptidomimetic or salt thereof (or composition comprising same) comprising a domain of formula I as defined herein for preventing or treating a bacterial or fungal infection in a subject.
- subject denotes any animal, preferably a mammal, and more preferably a human.
- subjects include humans, non-human primates, rodents, guinea pigs, rabbits, farm animals (e.g., sheep, pigs, goats, cows, horses) and pets (e.g., dogs and cats).
- farm animals e.g., sheep, pigs, goats, cows, horses
- pets e.g., dogs and cats.
- the polypeptide, peptide, peptidomimetic or salt thereof may be used for human health and veterinary applications.
- polypeptide, peptide, peptidomimetic or salt thereof (or composition comprising same) disclosed herein may be useful for the treatment of bacterial infection caused by Grampositive or Gram-negative species, such as Escherichia coli, Pseudomonas aeruginosa, Klebsiella pneumoniae, Acinetobacter baumannii, Enterobacter cloacae, Staphylococcus aureus, Enterococcus faecalis, Enterococcus faecium, Legionella pneumophila, Mycoplasma pneumonia, Acinetobacter haemolyticus, Acinetobacter junii, Acinetobacter Iwoffi, Burkholderia cepacia, Chlamydophila pneumoniae, Clostridium difficile, Enterobacter aerogenes, Enterobacter cloacae, Moraxella catarrhalis, Neisseria gonorrhoeae, Neisseria meningitides, Proteus mirabili
- the bacterial or fungal infection causes a periodontal disease.
- gingival disease refers to an infection caused by microorganisms that colonize the tooth or implant surface at or below the gingival margin.
- the bacteria causing the periodontal disease comprises Fusobacterium nucleatum, Prevotella intermedia, Treponema denticola and/or Porphyromonas gingivalis but are not limited to this list.
- the periodontal disease is periimplantitis, gingivitis, periodontitis or peri-implant mucositis.
- the method/use disclosed herein prevents or reduces the risk of developing a disease associated with a bacterial or fungal infection. In an embodiment, the method/use disclosed herein prevents or reduces the risk of developing a disease associated with Porphyromonas gingivalis infection, such as cardiovascular or respiratory disease, diabetes, or Alzheimer’s disease.
- the polypeptide, peptide, peptidomimetic or salt thereof may be incorporated with one or more excipients in the form of a mouthwash, dentifrice, gel, buccal tablet, oral spray, a chewing gum, a patch, a film, a strip or sublingual orally-administered formulation.
- a mouthwash may be prepared incorporating the active ingredient in the required amount in an appropriate solvent, such as a sodium borate solution (Dobell's Solution).
- the active ingredient may be incorporated into an oral solution such as one containing sodium borate, glycerin and potassium bicarbonate, or dispersed in a dentifrice, or added in a therapeutically effective amount to a composition that may include water, binders, abrasives, flavoring agents, foaming agents, and humectants.
- a composition may include water, binders, abrasives, flavoring agents, foaming agents, and humectants.
- the compositions may be fashioned into a tablet or solution form that may be placed under the tongue or otherwise dissolved in the mouth.
- the polypeptide, peptide, peptidomimetic or salt thereof is/are delivered locally to sites of periodontal disease via chemical and/or ionic attachment or adhesion to suitable substrates currently used for the localized delivery of antibiotic therapy to treat periodontal disease.
- suitable substrates currently used for the localized delivery of antibiotic therapy to treat periodontal disease.
- substrates used to deliver localized antibiotic therapy to periodontal sites of disease known in the art include, but are not limited to, biofilms, nanoparticles, suture material, microspheres, polymers, fibers, matrixes, and gels.
- substrates applied directly to pockets of periodontal disease via methods known in the art and described above include FDA-approved products such ARESTIN, ATRIDOX, ACTISITE, PERIOCHIP ® , ELYZOL, and DENTOMYSIN.
- the Porphyromonas gingivalis infection is a disseminated infection. In an embodiment, the Porphyromonas gingivalis infection is in an aortic valve.
- the above-mentioned treatment comprises the use/administration of more than one (i.e. a combination of) active/therapeutic agent, one of which being the above- mentioned polypeptide, peptide, peptidomimetic or salt thereof (or composition comprising same).
- the combination of prophylactic/therapeutic agents and/or compositions of the present disclosure may be administered or co-administered (e.g., consecutively, simultaneously, at different times) in any conventional dosage form.
- Co-administration in the context of the present disclosure refers to the administration of more than one therapeutic in the course of a coordinated treatment to achieve an improved clinical outcome. Such co-administration may also be coextensive, that is, occurring during overlapping periods of time.
- a first agent may be administered to a patient before, concomitantly, before and after, or after a second active agent is administered.
- the agents may in an embodiment be combined/formulated in a single composition and thus administered at the same time.
- the above-mentioned polypeptide, peptide, peptidomimetic or salt thereof (or composition comprising same) is used/administered in combination with one or more agent(s) currently used to prevent or treat the disorder in question.
- the above-mentioned polypeptide, peptide, peptidomimetic or salt thereof may be co-administered with an anti-bacterial or anti-fungal agent such as a quaternary ammonium compound (QAC) or a pharmaceutically acceptable salt thereof, e.g., benzalkonium chloride, or cetylpyridinium chloride; a guanidine compound or a pharmaceutically acceptable salt thereof, e.g., ampicillin, tetracycline, doxycycline, metronidazole, clindamycin, amoxicillin + Clavulinic acid (Augmentin), azithromycin, erythromycin, metronidazole + amoxicillin, spiramycin, chlorhexidine, alexidine, or polyhexamethylene biguanide (PHMB); hexetidine, triclosan, liniment, fluconazole, itraconazole, amphotericin B, vori
- QAC quaternary am
- the regimen of administration may affect what constitutes an effective amount. For example, several divided dosages, as well as staggered dosages may be administered daily or sequentially, or the dose may be continuously infused, or may be a bolus injection. Further, the dosages of the therapeutic formulations may be proportionally increased or decreased as indicated by the exigencies of the therapeutic or prophylactic situation.
- Administration of the polypeptide, peptide, peptidomimetic or salt thereof (or composition comprising same) of the present disclosure to a patient, such as a mammal, such as a human, may be carried out using known procedures, at dosages and for periods of time effective to treat a disease or disorder in the patient.
- An effective amount of the therapeutic compound necessary to achieve a therapeutic effect may vary according to factors such as the activity of the particular compound employed; the time of administration; the rate of excretion of the compound; the duration of the treatment; other drugs, compounds or materials used in combination with the compound; the state of the disease or disorder, age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well-known in the medical arts.
- Dosage regimens may be adjusted to provide the optimum therapeutic response. For example, several divided doses may be administered daily or the dose may be proportionally reduced as indicated by the exigencies of the therapeutic situation.
- a non-limiting example of an effective dose range for a therapeutic compound is from about 0.01 and 50 mg/kg of body weight/per day.
- the compound can be administered to an animal as frequently as several times daily, or it may be administered less frequently, such as once a day, once a week, once every two weeks, once a month, or even less frequently, such as once every several months or even once a year or less.
- the amount of compound dosed per day may be administered, in non-limiting examples, every day, every other day, every 2 days, every 3 days, every 4 days, or every 5 days.
- the frequency of the dose is readily apparent to the skilled artisan and depends upon any number of factors, such as, but not limited to, the type and severity of the disease being treated, and the type and age of the animal.
- Actual dosage levels of the active ingredients in the pharmaceutical compositions may be varied so as to obtain an amount of the active ingredient that is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient.
- a medical doctor e.g., physician or veterinarian, having ordinary skill in the art may readily determine and prescribe the effective amount of the pharmaceutical composition required.
- physician or veterinarian could start doses of the compounds employed in the pharmaceutical composition at levels lower than that required in order to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved.
- Dosage unit form refers to physically discrete units suited as unitary dosages for the patients to be treated; each unit containing a predetermined quantity of therapeutic compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical vehicle.
- the dosage unit forms are dictated by and directly dependent on (a) the unique characteristics of the therapeutic compound and the particular therapeutic effect to be achieved, and (b) the limitations inherent in the art of compounding/formulating such a therapeutic compound for the treatment of a disease or disorder in a patient.
- the disclosed compositions are administered to the patient in dosages that range from one to five times per day or more. In other embodiments, the compositions are administered to the patient in range of dosages that include, but are not limited to, once every day, every two, days, every three days to once a week, and once every two weeks. It is readily apparent to one skilled in the art that the frequency of administration of the various combination compositions varies from subject to subject depending on many factors including, but not limited to, age, disease or disorder to be treated, gender, overall health, and other factors. Thus, the disclosed subject matter should not be construed to be limited to any particular dosage regime and the precise dosage and composition to be administered to any patient will be determined by the attending physical taking all other factors about the patient into account.
- Compounds disclosed for administration may be in the range of from about 1 pg to about 7,500 mg, about 20 pg to about 7,000 mg, about 40 pg to about 6,500 mg, about 80 pg to about 6,000 mg, about 100 pg to about 5,500 mg, about 200 pg to about 5,000 mg, about 400 pg to about 4,000 mg, about 800 pg to about 3,000 mg, about 1 mg to about 2,500 mg, about 2 mg to about 2,000 mg, about 5 mg to about 1 ,000 mg, about 10 mg to about 750 mg, about 20 mg to about 600 mg, about 30 mg to about 500 mg, about 40 mg to about 400 mg, about 50 mg to about 300 mg, about 60 mg to about 250 mg, about 70 mg to about 200 mg, about 80 mg to about 150 mg, and any and all whole or partial increments therebetween.
- the dose of a compound is from about 0.5 pg and about 5,000 mg. In some embodiments, a dose of a compound used in compositions described herein is less than about 5,000 mg, or less than about 4,000 mg, or less than about 3,000 mg, or less than about 2,000 mg, or less than about 1 ,000 mg, or less than about 800 mg, or less than about 600 mg, or less than about 500 mg, or less than about 200 mg, or less than about 50 mg.
- a dose of a second compound as described herein is less than about 1 ,000 mg, or less than about 800 mg, or less than about 600 mg, or less than about 500 mg, or less than about 400 mg, or less than about 300 mg, or less than about 200 mg, or less than about 100 mg, or less than about 50 mg, or less than about 40 mg, or less than about 30 mg, or less than about 25 mg, or less than about 20 mg, or less than about 15 mg, or less than about 10 mg, or less than about 5 mg, or less than about 2 mg, or less than about 1 mg, or less than about 0.5 mg, and any and all whole or partial increments thereof.
- the present disclosure provides a method for cleaning or disinfecting a surface or an object comprising contacting the surface or the object with an effective amount of the polypeptide, peptide, peptidomimetic, salt or composition (e.g., cleaning or detergent composition) described herein.
- the cleaning or detergent composition may be applied for a period of 0.1 , 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1 , 2, 3, 4, 5, 10 or 15 minutes to 1 hour, such as from 10 seconds to 15 minutes, such as from 10 seconds to 1 minute, such as from 10 seconds to 5 minutes, such as from 20 seconds to 15 minutes, such as from 1 minute to 5 minutes, such as from 2 minutes to 10 minutes, such as from 20 minutes to 55 minutes such as from 20 to 30 minutes.
- Example 1 Materials and Methods Cloning procedures.
- the sister proteins ODAM and AMTN also belonging to the SCPP cluster were used as control.
- Truncated versions of SCPPPQ1, AMTN and ODAM genes lacking regions encoding the predicted N-terminal signal sequence were PCR-amplified from human cDNA sequences using primers as previously described. PCR products were cloned into the vector pHT for purification studies.
- the recombinant pHT plasmids allow to produce recombinant proteins with an in-frame N-terminal hexahistidyl-tag (His-tag) and Tobacco Etch Virus (TEV) protease cleavage site.
- Escherichia coli strain XL-1 Blue were used as hosts for cloning.
- BL21 (DE3)-star cells containing either pHT- SCPPPQ1, pHT -AMTN or pHT-ODA were grown at 37 °C and 250 rpm to an optical density at 660 nm (OD 66 o) around 0.6, and protein expression was induced with 0.1 mM isopropyl-B-D- thiogalactoside (IPTG) for ON at 30 °C and 250 rpm.
- bacterial cells were harvested, suspended in equilibration buffer (50 mM Na 2 HP0 , 150 mM NaCI, 10 mM imidazole, 6 M urea, pH 7) at 4 °C, and sonicated six times 15 s between 15 s ice incubations. Lysates were centrifuged at 13,400 g and the 6His-tagged protein in the supernatant was bound on nickel-nitriloacetic acid (Ni-NTA)-agarose affinity resin (Qiagen, Valencia, CA, USA) at room temperature.
- Ni-NTA nickel-nitriloacetic acid
- binding buffer 50 mM Na 2 HP0 4 , 300 mM NaCI, 20 mM imidazole, 6 M Urea, pH 7
- proteins were eluted with elution buffer (50 mM Na 2 HP0 , 300 mM NaCI, 300 mM imidazole, 6 M Urea, pH 7).
- Concentration of the collected fractions were assessed using a biodrop (Montreal Biotech Inc., Dorval, Canada) and fractions were then analyzed by SDS-PAGE and Coomassie blue staining. Proteins were finally dialyzed into 50 mM Na 2 HP0 , 6 M Urea (pH 7.2) and stored at 4 °C.
- ODAM and AMTN were purified in the same conditions as SCPPPQ1 but in a buffer without urea.
- Porphyromonas gingivalis ATCC 33277, Fusobacterium nucleatum ATCC 25586, Prevotella intermedia ATCC 2561 1 and Aggregatibacter actinomycetemcomitans ATCC 29522 were grown anaerobically for 48 h (80% N 2 , 10% C0 2 , 10% H 2 ) at 37 °C in Todd- Hewitt broth (THB; Becton Dickinson, Canada) supplemented with 0.001 % hemin and 0.0001 % vitamin K.
- Treponema denticola ATCC 35404 was grown anaerobically for 24 h in liquid medium containing 12.5 mg of brain heart infusion, 10 mg of trypticase, 2.5 mg of yeast extract, 0.5 mg of sodium thioglycolate, 1 mg of L-cysteine, 0.25 mg of L-asparagine, 2 mg of glucose, 6 pg of thiamine pyrophosphate, and 2 mg of sodium bicarbonate per ml; 2% rabbit serum; and 0.2% volatile fatty acids (19).
- the volatile fatty acids solution consisted of 0.5 ml each of isobutyric, DL-2-methylbutyric, isovaleric, and valeric acids dissolved in 100 ml of 0.1 N KOH.
- Escherichia coli, Bacillus subtilis and Bacillus thuringiensis was grown for 24 h in LB broth at 37 °C and 250 rpm, then the solution was diluted and grown around 6 hours to reach OD 66 o around 1 .
- Western blot analysis For western blot analysis, 40 pi was sampled every 15 min for western blot analysis using antibodies for rat ODAM (1 :5000), rat AMTN (1 :2000), or rat SCPPPQ1 (1 :2000). Protein alone in the bacteria media was used as a control. Western blot gels were acquired using a Bio-Rad ChemiDoc imager and the software Image Lab (Bio-Rad, Hercules, CA, USA). Exposure time for the acquisition were between 1 and 2 s.
- Flow cytometry (FACS) analysis Various concentration of proteins or peptides were exposed in a test tube to 500 pi of a suspension of bacteria at OD 66 o around 1 for up to 2 h at 37 °C before fixation using 1 % Glutaraldehyde. The suspension was then stained using FM4-64 (Thermo Scientific, Waltham, MA, USA) before analysis using the analyzer FacsAria III SORP (Becton Dickinson, NJ, USA). The collected data were then analyzed using the FlowJo program (Becton Dickinson, NJ, USA).
- Porphyromonas gingivalis was incubated 30 min at 37 °C on titanium surfaces before being mounted of a coverslip which was covered with toothpaste mixed with buffer (control) or rat SCPPPQ1 .
- an O-ring was positioned between the coverslip and the titanium disk and the space between was filled with PB 0.1 M (pH 7.2). After a one-hour incubation at 37 °C, the samples on the disks were fixed and prepared for scanning electron microscopy as previously described.
- Rat SCPPPQ1 , human SCPPPQ1 and buffer (control) were applied 30 min on separate titanium surface for absorption. The excess of liquid was then removed by airflow and the surfaces was dried 30 min at room temperature. Porphyromonas gingivalis was incubated for 1 h at 37 °C before being fixed and prepared for scanning electron microscopy as previously described.
- Embedding procedures Following bacterial incubations, some bacteria were centrifuged and prepared for embedding in Epon (Electron Microscopy Sciences, Hatfield, PA, USA). The bacteria were fixed as previously described and post-fixed with potassium ferrocyanide-reduced osmium tetroxide and then processed for embedding in Epon resin. For some samples, ultrathin sections of 80-100 nm were cut with a diamond knife and transferred onto Formvar®-coated (polyvinyl formate) 200-mesh nickel grids for TEM imaging. Grids were examined in a FEI Tecnai 12 (Eindhoven, The Netherlands) transmission electron microscope operating at 80 kV. Some embedded samples were used for colloidal gold immunolabelling.
- the grids were blocked for 15 min using 0.1 % ovalbumin, then incubated for 1 h with rabbit antibodies anti-SCPPPQ1 at RT, followed by a second blocking step and a final incubation of 30 min with 10 nm gold beads coupled to protein A at RT before to be air dried for observation on the TEM.
- Yeast analysis Candida albicans were grown in Lee and Sabouraud liquid media at 37 °C for 24 h.
- Various proteins or peptides at a final concentration of 20 mM were incubated with the yeast for 2 h in a test tube before application on titanium surfaces for 30 min.
- Ameloblast-like cells LS8 were cultured in DMEM supplemented with 10% heat inactivated fetal bovine serum (FBS) at 37 °C in a 5% C0 2 atmosphere. 250,000 cells were placed on 18x18mm coverslips and grown overnight. After 24 hours, rat SCPPPQ1 were added to the media cell culture to obtain a final concentration of 20 pM and the same volume of buffer were added to different coverslips as control. After 24 hours of incubation, cells were stained with rhodamine phalloidin (1 : 500, Thermo Scientific, Waltham, MA, USA) to highlight the actin and with Hoechst (1 :2000, Thermo Scientific, Waltham, MA, USA) for the nucleus. Fluorescent samples were acquired on an Axio-lmager (Carl Zeiss, Oberkochen, Germany) using the Zen Blue edition software and images were then analyzed and quantified using ImageJ.
- FBS heat inactivated fetal bovine serum
- the HPLC system was coupled to Orbitrap Fusion mass spectrometer (Thermo Scientific) through an Electrospray Ion Source.
- the spray and S-lens voltages were set to 3.6 kV and 50 V, respectively.
- Capillary temperature was set to 225°C.
- Full scan MS survey spectra m/z 500-1700 in profile mode were acquired in the Orbitrap with a resolution of 120,000 with a target value at 5e5.
- the 3 most intense protein/peptide ions were fragmented in the HCD collision cell and analyzed in the Orbitrap with a target value at 5e5 and a normalized collision energy at 36 V.
- Target ions selected for fragmentation were dynamically excluded for 25 sec.
- protein database searching was performed with ProSightPC 4.0.2.1 (Thermo Scientific) against the protein sequences of interest.
- the mass tolerances for precursor and fragment ions were set to 1000 Da and 20 ppm, respectively.
- the minimum number of matching fragments was set to 4 and the only modification used was acetylation of protein N-terminal.
- the number of matching fragments were quantified and classed by kDa to evaluate the effect of P. gingivalis incubation on each protein.
- Example 3 Determination of the antibacterial activity of recombinant human and rat
- SCPPPQ1 Having provided evidence that SCPPPQ1 exhibits antibacterial activity, the extent of the antimicrobial potential of SCPPPQ1 and its mechanism of action was assessed.
- FIG. 6B top panel, total volume of aggregates (A.U)
- rSCPPPQI rat SCPPPQ1
- hSCPPPQI human SCPPPQ1
- Decomposition of the total volumes of aggregates to determine the percentage of various aggregates formed show that rat SCPPPQ1 and human SCPPPQ1 form bigger aggregates than the control (Buffer) (FIG. 6B, lower panel).
- FIG. 7 The localization of the human (FIG. 7) and rat (FIG. 8) SCPPPQ1 proteins following treatment of P. gingivalis was assessed by immunofluorescence microscopy. Fluorescence images demonstrate that both proteins were localized at the surface of bacteria following the incubation period. A further characterization of the localization of the rat SCPPPQ1 protein was performed by colloidal gold immunolabelling of the protein and visualization by TEM. The results of FIG. 9 demonstrate that the protein surrounds the bacteria (left) and enters the bacteria after disruption of the membrane (right). This double effect is a distinctive feature that ensures destruction of bacteria, while reducing the possibility of bacteria developing resistance to this protein.
- FIG. 10 illustrates representative transmission and scanning electron micrograph of P. gingivalis incubated with rat SCPPPQ1 .
- the colloidal gold immunolabelling of rat SCPPPQ1 reveal the localization of the protein on the cells, which creates also a network between the bacteria and scanning electron microscopy of the bacteria confirms the network only in the presence of the protein.
- Fluorescent images of P. gingivalis show that increasing the concentration of rat SCPPPQ1 decreases the quantity of bacteria, and also that the bacteria create more clusters when the concentration of protein is increased (FIG. 11) .
- the sequences of human and rat SCPPPQ1 and derived peptides are depicted in FIG. 12.
- the predicted human SCPPPQ1 protein sequence corresponds to the putative sequence that was previously identified based on homology with the rat sequence, and the sequence human SCPPPQ1 protein truly expressed by human cells was identified and reported thereafter (Genbank accession No. MK322956.1).
- the first 56 amino-terminal residues of these sequences are identical, but the remaining C-terminal residues have no or very little sequence identity.
- FIG. 13 shows a comparison of the effects of the peptides on the formation of aggregates using SEM (FIGs. 13A-B). Results of the counting show that the percentage of aggregates is more important for peptides #1 1 , #12, #13 and #14. The results depicted in FIGs. 13D-E show that all peptides except peptide #9 induce damages to the bacterial membrane relative to control (buffer).
- results depicted in FIG. 16 show that incubation of Fusobacterium nucleatum, a commensal oral bacterium that plays a role in periodontal disease, with rat SCPPPQ1 , reduces the interaction between the bacteria, induces damages at their surface, and decreases the bacterial population by about 75% relative to the control.
- FIG. 17 shows that incubation of Prevotella intermedia, a gram-negative bacterium involved in periodontal infections (including gingivitis and periodontitis) with rat SCPPPQ1 decreases the bacterial population by about 70% relative to the control, and affects the interaction between the bacteria. No significant effect was observed following incubation with rat ODAM, his sister protein. Rat SCPPPQ1 was shown to have similar effects on Treponema denticola, another Gram-negative bacterium present in the oral cavity and associated with the incidence and severity of human periodontal disease (FIG. 18).
- Rat and human SCPPPQ1 were also shown to induce defects on the bacterial membrane (presence of cellular debris and signs of membrane damages) of a Gram+ bacterium, Bacillus subtilis, that is found in the gastrointestinal tract of humans (FIGs. 19A-B). A similar effect was obtained following incubation of another Gram+ Bacillus, Bacillus thuringiensis (FIGs. 20A-B).
- FIGs. 21A-B show that rat SCPPPQ1 has an inhibitory effect on the growth of Escherichia coli, as evidenced by an important decrease in the number of bacteria in the presence of rat SCPPPQ1 relative to control (FIG. 21A).
- Rat and human SCPPPQ1 were also demonstrated to induce defects in the Gram- bacterial membrane of Escherichia coli, as shown in the SEM images of FIG. 21 B.
- FIG. 23C shows representative agar plates of five different Candida albicans strains incubated in the presence or not of rat SCPPPQ1 . All the strains are“hairy” (indicative of the presence of long hyphae) and in healthy conditions in the absence of SCPPPQ1 , but become “smooth” in the presence of 6pm of rat SCPPPQ1 in the agar, indicative of a reduction in the number of and/or length of hyphae. Furthermore, a notable decrease of unit forming colony (UFC) was also detected for strains BL152 and ATCC 36802.
- Example 7 Assessment of the antimicrobial activity of SCPPPQ1 against P. gingivalis on titanium surface
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- General Health & Medical Sciences (AREA)
- Veterinary Medicine (AREA)
- Public Health (AREA)
- Animal Behavior & Ethology (AREA)
- Organic Chemistry (AREA)
- Medicinal Chemistry (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Epidemiology (AREA)
- Molecular Biology (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Pharmacology & Pharmacy (AREA)
- Oncology (AREA)
- Biochemistry (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Engineering & Computer Science (AREA)
- Genetics & Genomics (AREA)
- Communicable Diseases (AREA)
- Biophysics (AREA)
- Toxicology (AREA)
- Gastroenterology & Hepatology (AREA)
- Zoology (AREA)
- Dermatology (AREA)
- Birds (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Biomedical Technology (AREA)
- Oral & Maxillofacial Surgery (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
Abstract
Polypeptides, peptides, peptidomimetics, and salts thereof derived from secretory calcium-binding phosphoprotein-proline glutamine-rich 1 (SCPPPQ1) are described. The use of these polypeptides, peptides, peptidomimetics, and salts thereof, and compositions comprising same, as antibacterial or antifungal agents is also described. Methods for treating bacterial or fungal infections and related diseases such as periodontal or gingival diseases, or for cleaning or disinfecting a surface, are also described.
Description
TREATMENT OF MICROBIAL INFECTIONS USING SCPPPQ1 PROTEINS AND
DERIVED PEPTIDES
CROSS REFERENCE TO RELATED APPLICATIONS
This application claims the benefit of U.S. provisional application serial No. 62/829,732, filed on April 5, 2019, which is incorporated herein by reference in its entirety.
TECHNICAL FIELD
The present disclosure generally relates to the field of antimicrobial agents, and to the treatment of bacterial or fungal infections such as Porphyromonas gingivalis (P. gingivalis) and Candida albicans infections, and related diseases.
BACKGROUND ART
Antibiotics are commonly used in the treatment and/or prevention of infectious diseases caused by various microorganisms such as bacteria. Most antibiotics are compounds derived from plants or fungus, which are produced as a defense mechanism against bacterial infection. However, resistance of bacteria against these antibiotics often occurs. In fact, conventional antibiotics kill bacteria by binding to specific targets that are involved in bacterial DNA and protein synthesis. Therefore, resistance can occur when bacteria modify these targets, such that antibiotics do not bind to these proteins, or when bacteria produce specific enzymes that inactivate the antibiotics.
Pathogenic bacteria are usually found on or surrounding the surfaces of epithelial cells forming an epithelial barrier. If the epithelial barrier is compromised, the bacteria can penetrate into deeper tissue layers where they can expand and trigger infectious diseases. For instance, periodontal diseases (PD) affect a large segment of the population and their severity increases with age and eventually lead to tooth and bone loss.
In the tooth, a specialized basal lamina (sBL) attaches epithelial cells to mineralized surfaces rather than connective tissue, thereby creating secluded environments that are critical for mineralization and for protection of the tooth supporting tissues from the aggressive oral environment. The junctional epithelium (JE) is a specialized portion of the gingiva that seals off the tooth supporting tissues from the oral environment. The JE uniquely attaches to the tooth via the sBL, where are localized three secreted proteins namely amelotin (AMTN), odontogenic ameloblast-associated (ODAM) and secretory calcium-binding phosphoprotein- prolineglutamine-rich 1 (SCPPPQ1) that are expressed by the JE. These proteins are believed to participate with Laminin-332 (Lm332), that is also expressed by the JE, in structuring the supramolecular organization of the sBL and, hence, represent solid candidates in maintaining the integrity and function of the JE. However, their susceptibility to bacteria and/or bacterial
products ( e.g . , enzymes) remains unknown, which constitutes an important aspect for maintaining the gingival seal and health.
The human subgingival plaque harbors more than 700 bacterial species. Some of them lead to PD when the equilibrium of the oral microbiome is compromised. Among these bacterial species, Porphyromonas gingivalis (P. gingivalis) and Treponema denticola ( T . denticola) are strongly linked to PD. These bacteria secrete a variety of enzymes that are generally associated with destruction of subepithelial connective tissue. There is actually no cure and no reliable manner of controlling bacteria during PD.
Patients with dental implants are susceptible to developing conditions similar to the above described PD but which instead attack the tissues surrounding the implant. One such disease is peri-implant mucositis. This condition involves the presence of inflammation in the mucosa at an implant but with no signs of loss of supporting bone.
Patients with implants can also suffer from a condition called peri-implantitis, which is caused by the colonization of bacteria of the implant's surface. Inflammation in the bone surrounding the implant then causes loss of bone which ultimately may lead to failure of the implant. Peri-implantitis can stem from an existing periodontitis infection or patients can develop peri-implantitis without a previous history of periodontitis.
Moreover, there is growing evidence that PD are linked to systemic complications such as cardiovascular and respiratory diseases, diabetes, Alzheimer’s disease, thus causing serious health concerns that goes beyond the mouth. Recently, P. gingivalis, and toxic proteases produced by this bacterium, called gingipains, were detected in the brain of patients suffering from Alzheimer's disease (AD). Levels of gingipains in the brain correlated with AD diagnosis, and markers involved in AD pathology. Small-molecule inhibitors targeting gingipains were found to reduce the bacterial load of an established P. gingivalis brain infection and rescued the neurotoxic effects of gingipains (Dominy et al., Science Advances 23 Jan 2019: Vol. 5, no. 1). P. gingivalis has been shown to induce platelet aggregation which lead to thrombus formation during cardiovascular disease. In addition, reports indicate that atherosclerotic plaques are commonly colonized with P. gingivalis, suggesting the involvement of these bacteria in atherosclerosis and myocardial infarction. Furthermore, P. gingivalis is also associated with bacterial pneumonia resulting from aspiration of oropharyngeal flora into the lower respiratory tract. More specifically, the production of gingipains by these bacteria is known to impact the innate immune response and promote chronic inflammation (Benedyk et al., Journal of Innate Immunity 2016; 8, No. 2: 185-198).
Globally, over 300 million people are afflicted by a serious fungal infection and 25 million are at risk of dying or losing their sight (Fungal Infection Trust 201 1). Among fungal infections, invasive fungal infections such as cryptococcosis, candidiasis, aspergillosis and pneumocystosis are the most common and the most life-threatening. Candida species such as
Candida albicans can causes oral candidiasis (oral thrush). While there are about 30 branded prescription antifungal drugs on the market, three classes of antifungals are mainly used to manage invasive fungal infections: 1) azoles, such as fluconazole launched in the mid-1980s, 2) polyenes, such as amphotericin B launched in the mid-1950s and 3) echinocandins, such as caspofungin launched in early 2000. However, the increased use of current azoles has led to an increase in drug resistance, systemic antifungals such as amphotericin B tend to have relatively high toxicity and side effects, and there are no oral formulations for echinocandins, which also have narrow antifungal spectrum of activity.
There is thus a need for novel preventive and therapeutic treatments of infections and related diseases such as PD, while limiting microbial resistance or causing undesirable side effects.
The present description refers to a number of documents, the content of which is herein incorporated by reference in their entirety.
SUMMARY OF THE INVENTION
In a first aspect, the present disclosure provides the following items:
1 . An isolated polypeptide, peptide or peptidomimetic of 7 to 100 residues comprising a sequence of formula I:
X1 -X2-X3-X4-X5-X6-X7 -X8-X9-X10-X1 1 -X12-X13-X14-X15-X16-X17-X18-X19 (I) wherein
represents a bond;
XI is phenylalanine or an analog thereof, or is absent;
X2 is proline or an analog thereof;
X3 is leucine or an analog thereof, or is absent;
X4 is glutamine or an analog thereof, or is absent;
X5 is glutamine, proline, leucine or an analog thereof, or is absent;
X6 is proline or an analog thereof;
X7 is glutamine or an analog thereof, or is absent;
X8 is alanine or an analog thereof, or is absent;
X9 is proline or an analog thereof;
X10 is any amino acid or amino acid analog, or is absent;
XI I glutamine, glycine, or an analog thereof, or is absent;
X12 is leucine or an analog thereof, or is absent;
X13 is proline or an analog thereof, or is absent;
X14 is leucine or an analog thereof, or is absent;
X15 is proline or an analog thereof, or is absent;
X16 is isoleucine, leucine, or an analog thereof, or is absent;
X17 is proline, glutamine, or an analog thereof, or is absent;
X18 is isoleucine, leucine, phenylalanine, or an analog thereof, or is absent;
X19 is proline or an analog thereof, or is absent;
or a salt thereof.
2. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to item 1 , wherein X1 is L-phenylalanine.
3. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to item 1 or 2, wherein X2 is L-proline.
4. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of items 1 to 3, wherein X3 is L-leucine.
5. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of items 1 to 4, wherein X4 is absent.
6. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of items 1 to 4, wherein X4 is L-glutamine.
7. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of items 1 to 6, wherein X5 is absent.
8. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of items 1 to 6, wherein X5 is L-glutamine, L-proline or L-leucine.
9. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of items 1 to 8, wherein X6 is L-proline.
10. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of items 1 to 9, wherein X7 is L-glutamine.
1 1 . The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of items 1 to 10, wherein X8 is L-alanine.
12. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of items 1 to 1 1 , wherein X9 is L-proline.
13. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of items 1 to 12, wherein X10 is L-glutamine, L-leucine, L-serine, L-valine or L-proline, D-proline.
14. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of items 1 to 13, wherein X1 1 , X12, X13, X14 and/or X15 are absent.
15. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of items 1 to 13, wherein X1 1 , X12, X13, X14 and X15 are absent.
16. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of items 1 to 13, wherein X1 1 is L-glutamine.
17. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of items 1 to 14 and 16, wherein X12 is L-leucine.
18. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of items 1 to 14, 16 and 17, wherein X13 is L-proline.
19. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of items 1 to 14 and 16 to 18, wherein X14 is L-leucine.
20. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of items 1 to 14 and 16 to 19, wherein X15 is L-proline.
21 . The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of items 1 to 20, wherein X16 is L-isoleucine or L-leucine.
22. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of items 1 to 20, wherein X16 is absent.
23. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of items 1 to 22, wherein X17 is L-proline.
24. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of items 1 to 23, wherein X18 is L-isoleucine, L-leucine or L-phenylalanine.
25. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of items 1 to 24, wherein X18 is L-isoleucine, L-leucine or L-phenylalanine.
26. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of items 1 to 25, wherein X19 is L-proline.
27. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of items 1 to 26, wherein represents a peptide bond.
28. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of items 1 to 27, which comprises a sequence of at least 7 or 8 residues from one of the following sequences: FPLPPQPP (SEQ ID NO:1), FPLPQAPLIPIP (SEQ ID NO:2), FPQPPQLQIP (SEQ ID NO:3), PLPPSPP (SEQ ID NO:4), FPFQQPPLIPFP (SEQ ID NO:5), FPPPVGLPLIPLP (SEQ ID NO:6), FPPPPQIPIP (SEQ ID NO:7), FPLPRQIPIP (SEQ ID NO:8), FPPPPQLPLIPIP (SEQ ID NO:9), FPLPQAPLIPIP (SEQ ID NO:10), or a variant thereof having at least 70% sequence identity with the at least 7 or 8 residues.
29. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to item 28, which comprises a sequence of at least 7 or 8 residues from one of the following sequences: FPLPPQPP (SEQ ID NO: 1 ), FPLPQAPLIPIP (SEQ ID NO:2), FPQPPQLQIP (SEQ ID NO:3), PLPPSPP (SEQ ID NO:4), FPFQQPPLIPFP (SEQ ID NO:5), FPPPVGLPLIPLP (SEQ ID NO:6), FPPPPQIPIP (SEQ ID NO:7), FPLPRQIPIP (SEQ ID NO:8), FPPPPQLPLIPIP (SEQ ID NO:9), or FPLPQAPLIPIP (SEQ ID NO: 10).
30. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to item 29, wherein the polypeptide, peptide or peptidomimetic comprises the sequence FPLPPQPP (SEQ ID NO: 1) or FPLPQAP (SEQ ID NO: 1 1), or a variant thereof having at least 70% sequence identity with these sequences.
31 . The isolated polypeptide, peptide, peptidomimetic or salt thereof according to item 30, wherein the polypeptide, peptide or peptidomimetic comprises the sequence FPLPPQPP (SEQ ID NO: 1 ) or FPLPQAP (SEQ ID NO:1 1 ).
32. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of items 1 to 31 , which comprises at least 10 contiguous amino acids from the amino acid sequences of any one of SEQ ID NOs: 12-29 and 62.
33. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to item 32, comprises an amino acid sequence having at least 70% sequence identity with the sequences of any one of SEQ ID NOs: 12-29 and 62.
34. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to item 33, comprises with the amino acid sequences of any one of SEQ ID NOs: 12-29 and 62.
35. An isolated nucleic acid comprising a nucleotide sequence encoding the polypeptide or peptide of any one of items 1 to 34.
36. A vector comprising the nucleic acid of item 35.
37. A host cell comprising nucleic acid of item 35 or the vector of item 36.
38. A composition comprising the isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of items 1 to 34, and one or more excipients.
39. The composition of item 38, wherein the one or more excipients comprise a biodegradable polymer.
40. The composition of item 38 or 39, which is in the form of a cream, a gel, a paste, a solution, a chewing gum, a patch, a film, or a strip.
41 . A method for treating a bacterial or fungal infection in a subject comprising administering to the subject an effective amount of the polypeptide, peptide, peptidomimetic or salt thereof according to any one of items 1 to 34, or the composition of any one of items 38 to 40.
42. The method of item 41 , wherein the fungal infection is a yeast infection.
43. The method of item 42, wherein the yeast is of the Candida genus.
44. The method of item 43, wherein the yeast is of the Candida albicans species.
45. The method of item 41 , wherein the bacterial infection is by Gram- bacteria.
46. The method of item 45, wherein the Gram- bacteria are of the Porphyromonas, Prevotella, Fusobacterium, Escherichia and/or Treponema genus.
47. The method of item 46, wherein the Gram- bacteria are of the Porphyromonas gingivalis, Fusobacterium nucleatum, Prevotella intermedia, Escherichia coli and/or Treponema denticola species.
48. The method of item 41 , wherein the bacterial infection is by Gram+ bacteria.
49. The method of item 48, wherein the Gram+ bacteria are of the Bacillus genus.
50. The method of item 49, wherein the Gram+ bacteria are of the Bacillus subtilis species.
51 . A method for treating a periodontal or gingival disease in a subject in need thereof, said method comprising contacting the gum from the subject with an effective amount of the polypeptide, peptide, peptidomimetic or salt thereof according to any one of items 1 to 34, or the composition of any one of items 38 to 40.
52. The method of item 51 , wherein the composition is the composition of item 40.
53. The method of item 51 or 52, wherein the periodontal or gingival disease is peri- implantitis, gingivitis, periodontitis or peri-implant mucositis.
54. A method for cleaning or disinfecting a surface or an object comprising contacting the surface or the object with an effective amount of the polypeptide, peptide, peptidomimetic or salt thereof according to any one of items 1 to 34, or the composition of any one of items 38 to 40.
55. The method of item 54, wherein the object is a dental or orthopedic implant.
56. The polypeptide, peptide, peptidomimetic or salt thereof according to any one of items 1 to 34, or the composition of any one of items 38 to 40 for treating a bacterial or fungal infection in a subject.
57. The polypeptide, peptide, peptidomimetic, salt or composition for use according to item
56, wherein the fungal infection is a yeast infection.
58. The polypeptide, peptide, peptidomimetic, salt or composition for use according to item
57, wherein the yeast is of the Candida genus.
59. The polypeptide, peptide, peptidomimetic, salt or composition for use according to item
58, wherein the yeast is of the Candida albicans species.
60. The polypeptide, peptide, peptidomimetic, salt or composition for use according to item 56, wherein the bacterial infection is by Gram- bacteria.
61 . The polypeptide, peptide, peptidomimetic, salt or composition for use according to item
60, wherein the Gram- bacteria are of the Porphyromonas, Prevotella, Fusobacterium, Escherichia and/or Treponema genus.
62. The polypeptide, peptide, peptidomimetic, salt or composition for use according to item
61 , wherein the Gram- bacteria are of the Porphyromonas gingivalis, Fusobacterium nucleatum, Prevotella intermedia, Escherichia coli and/or Treponema denticola species.
63. The polypeptide, peptide, peptidomimetic, salt or composition for use according to item 56, wherein the bacterial infection is by Gram+ bacteria.
64. The polypeptide, peptide, peptidomimetic, salt or composition for use according to item
63, wherein the Gram+ bacteria are of the Bacillus genus.
65. The polypeptide, peptide, peptidomimetic, salt or composition for use according to item
64, wherein the Gram+ bacteria are of the Bacillus subtilis species.
66. The polypeptide, peptide, peptidomimetic or salt thereof according to any one of items 1 to 34, or the composition of any one of items 38 to 40 for treating a periodontal or gingival disease in a subject.
67. The polypeptide, peptide, peptidomimetic, salt or composition for use according to item 66, wherein the composition is the composition of item 40.
68. The polypeptide, peptide, peptidomimetic, salt or composition for use according to item 66 or 67, wherein the periodontal or gingival disease is peri-implantitis, gingivitis, periodontitis or peri-implant mucositis.
69. The polypeptide, peptide, peptidomimetic or salt thereof according to any one of items 1 to 34, or the composition of any one of items 38 to 40 for cleaning or disinfecting a surface or an object.
70. The polypeptide, peptide, peptidomimetic, salt or composition for use according to item 69, wherein the object is a dental or orthopedic implant.
71. The polypeptide, peptide, peptidomimetic or salt thereof according to any one of items 1 to 34, or the composition of any one of items 38 to 40 for use i) for treatment and prevention of infectious diseases (e.g., bacterial or fungal infections in the eyes, mouth, wounds, and brain); ii) for treatment and prevention of post-operation bacterial or fungal infections and systemic propagations (e.g., implants such as dental or orthopedic implants); iii) for improvement of postoperation wound healing (e.g. around implants); iv) for inhibition of P. gingivalis and/or other periodontal disease-associated pathogens and treatment of its associated diseases including but not limited to Alzheimer's disease, cardiovascular disease, and bacterial pneumonia; v) treatment and prevention of periodontal disease; vi) for surface sterilization or antimicrobial/antifungal surface coating in industry (e.g., food industry) and hospital applications (e.g., surgical tools, medical devices, and areas); vii) as antibacterial/antifungal solutions (e.g. , hand gels, cleaning surfaces solution); viii) as health products (e.g., antibacterial/antifungal toothpaste, creams, drops, mouthwash); ix) as preservative in food products, beverages, cosmetics (lotions, creams, gels, soaps, shampoos, conditioners, antiperspirants) contact lens products, and food ingredients.
Other objects, advantages and features of the present invention will become more apparent upon reading of the following non-restrictive description of specific embodiments thereof, given by way of example only with reference to the accompanying drawings.
BRIEF DESCRIPTION OF DRAWINGS
In the appended drawings:
FIGs. 1A-B are representative pictures of western blot analysis following in vitro digestion assay of AMTN, ODAM and SCPPPQ1 by (FIG. 1A) A. actinomycetemcomitans or F. nucleatum showing no degradation of the proteins, while digestion assay by (FIG. 1 B) more aggressive bacteria as T. denticola, P. gingivalis and P. intermedia degrade AMTN and ODAM but not SCPPPQ1.
FIGs. 2A-C illustrate graphs showing the results of top-down mass spectrometry of AMTN (FIG. 2A, SEQ ID NO: 63), ODAM (FIG. 2B, SEQ ID NO: 64) and SCPPPQ1 (FIG. 2C, SEQ ID NO: 65) proteins before and after incubation with P. gingivalis, confirming the degradation of AMTN and ODAM and the non-degradation of SCPPPQ1 as previously observed by western blot analysis. The right panels highlight the cleavage sites (arrows) for the proteins.
FIG. 3 illustrates representative pictures of scanning electron microscope (SEM) of P. gingivalis after incubation (top left) with buffer only (i.e. control) (top right) with ODAM protein and (bottom left) with the rat SCPPPQ1 protein (SEQ ID NO: 31).
FIG. 4 illustrates two representative examples of SEM images of P. gingivalis after incubation (left top and bottom) without protein (i.e. control), (middle top and bottom) with rat SCPPPQ1 (rSCPPPQI) protein, and (right top and bottom) with human SCPPPQ1 (hSCPPPQI) protein.
FIG. 5A illustrates representative transmission electron microscope (TEM) images of P. gingivalis, to assess the structural appearance of the bacterial membrane, after incubation with buffer only (i.e. control) (top, control), with rat SCPPPQ1 (bottom).
FIG. 5B illustrates representative TEM images of P. gingivalis following a two-hour incubation without protein (left, control) and with human SCPPPQ1 (right) on Porphyromonas gingivalis.
FIG. 6A is a graph showing a comparison of the total number of bacteria following incubation with buffer only (control) or with human SCPPPQ1 (hSCPPPQI) or rat SCPPPQ1 (rSCPPPQI) after counting bacteria from SEM micrographs.
FIG. 6B are graphs showing a comparison of the effects of the rSCPPPQI and hSCPPPQI proteins on the formation of aggregates as assessed by flow cytometry (FACS).
FIG. 7 illustrates representative immunofluorescence microscopy images of P. gingivalis, assessing the localization of SCPPPQ1 after incubation (top) with buffer only (i.e. control), and (bottom) with the human SCPPPQ1 (hSCPPPQI) protein (SEQ ID NO: 30).
FIG. 8 illustrates representative immunofluorescence microscopy images of P. gingivalis, assessing the localization of SCPPPQ1 , after incubation (top) without protein (i.e. control), and (bottom) with rat SCPPPQ1 (rSCPPPQI) protein.
FIG. 9 illustrates representative TEM images of P. gingivalis, to assess the localization of rat SCPPPQ1 (rSCPPPQI) in colloidal gold immuno-labeled preparations for rSCPPPQI protein.
FIG. 10 illustrates representative transmission (top) and scanning electron (bottom) micrographs of P. gingivalis highlighting the network of rat SCPPPQ1.
FIG. 11 depicts representative fluorescence images of P. gingivalis incubated with increasing concentrations of rat SCPPPQ1.
FIG. 12 depicts the sequence of (i) the predicted human SCPPPQ1 protein, (ii) the actual human SCPPPQ1 protein, (iii) the rat SCPPPQ1 protein (SEQ ID NO: 31), and (iv)-(xvii) of peptides derived therefrom.
FIGs. 13A-B show representative SEM images of P. gingivalis to assess the structural appearance of bacteria after incubation with the different peptides depicted in FIG. 12.
FIG. 13C is a graph showing a comparison of the effects of the peptides on the formation of aggregates visualised using SEM (FIGs. 13A-B).
FIG. 13D-E are graphs showing a comparison of the effects of the peptides on the bacterial membranes using SEM (FIGs. 13A-B). FIG. 13D: comparison of the percentage of bacteria that are undamaged or damaged. FIG. 13E: percentage of undamaged membrane or the two different type of damaged membranes (holes or collapsed).
FIG. 14A illustrate an alignment of the confirmed or predicted amino acid sequences of the SCPPPQ1 protein from several species including human, rat, mouse, chimp, gorilla, baboon, macaque, vervet, gibbon, elephant, squirrel, rabbit, Chinese hamster, bovine, sheep, cat, dog, horse, and pig. The sequences corresponding to the signal peptides are underlined, and the domains corresponding to residues 48-55 of the confirmed human SCPPPQ1 protein (referred to as peptide #5 in subsequent figures) are in bold.
FIG. 14B illustrates the sequences of the domains corresponding to residues 48-55 of the confirmed human SCPPPQ1 protein depicted in FIG. 14A.
FIG. 15 shows representative TEM images of P. gingivalis to assess the structural appearance of the bacterial membrane after incubation with buffer only (i.e. control), and with the different peptides depicted in FIG. 12.
FIG. 16 shows representative SEM images of Fusobacterium nucleatum with buffer only (i.e. control) or in presence of rat ODAM or rat SCPPPQ1.
FIG. 17 shows representative SEM images of Prevotella intermedia with buffer only (i.e. control) or in presence of rat ODAM or rat SCPPPQ1 .
FIG. 18 shows representative SEM images of Treponema denticola with buffer only (i.e. control) or in presence of rat SCPPPQ1.
FIG. 19A shows representative fluorescence images of Bacillus subtilis with buffer only (i.e. control) or in presence of rat SCPPPQ1.
FIG. 19B shows representative SEM images of Bacillus subtilis with buffer only (i.e. control) or in presence of rat SCPPPQ1 (rSCPPPQI) or human SCPPPQ1 (hSCPPPQI).
FIG. 20A shows representative fluorescence images of Bacillus thuringiensis with buffer only (i.e. control) or in presence of rat SCPPPQ1.
FIG. 20B shows representative SEM images of Bacillus thuringiensis with buffer only (i.e. control) or in presence of rSCPPPQI or hSCPPPQI .
FIG. 21 A shows representative fluorescence images of Escherichia coli with buffer only (i.e. control) or in presence of rat SCPPPQ1 (rSCPPPQI).
FIG. 21 B shows representative SEM images of Escherichia coli with buffer only (i.e. left, control) or in presence of rat SCPPPQ1 (rSCPPPQI) or human SCPPPQ1 (hSCPPPQI).
FIG. 22 shows representative SEM images of P. gingivalis with buffer only or water only (i.e. control) of kanamycin, rat SCPPPQ1 (rSCPPPQI) or human SCPPPQ1 (hSCPPPQI).
FIG. 23A shows representative SEM images of Candida albicans with buffer only (i.e. control) or in the presence of SCPPPQ1 (human or rat) and of peptides #5, #8 and #10.
FIG. 23B shows a graph of the number of Candida albicans in the presence (dark) or absence (grey) of hyphae depending of the protein or peptides used.
FIG. 23C shows various Candida albicans strains (top) grown in culture dishes in presence or not of rat SCPPPQ1 and a summary table (bottom) of the number of Unit Forming Colony (UFC) and their appearances.
FIG. 24 shows an analysis of the effect of rat SCPPPQ1 on LS8 cells (ameloblast-like cell line).
FIG. 25 shows the effect of toothpaste with buffer only (i.e. control) or rat SCPPPQ1 on Porphyromonas gingivalis as assessed by SEM (top).
FIG. 26 shows the effect of the absorption of rat SCPPPQ1 or human SCPPPQ1 on titanium surfaces on Porphyromonas gingivalis as assessed by SEM.
DISCLOSURE OF INVENTION
The use of the terms "a" and "an" and "the" and similar referents in the context of describing the technology (especially in the context of the following claims) are to be construed to cover both the singular and the plural, unless otherwise indicated herein or clearly contradicted by context.
The terms "comprising", "having", "including", and "containing" are to be construed as open-ended terms (i.e., meaning "including, but not limited to") unless otherwise noted.
All methods described herein can be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context.
The use of any and all examples, or exemplary language (“e.g.”, "such as") provided herein, is intended merely to better illustrate embodiments of the claimed technology and does not pose a limitation on the scope unless otherwise claimed.
No language in the specification should be construed as indicating any non-claimed element as essential to the practice of embodiments of the claimed technology.
Herein, the term "about" has its ordinary meaning. The term“about” is used to indicate that a value includes an inherent variation of error for the device or the method being employed
to determine the value, or encompass values close to the recited values, for example within 10% of the recited values (or range of values).
Recitation of ranges of values herein are merely intended to serve as a shorthand method of referring individually to each separate value falling within the range, unless otherwise indicated herein, and each separate value is incorporated into the specification as if it were individually recited herein. All subsets of values within the ranges are also incorporated into the specification as if they were individually recited herein.
Where features or aspects of the disclosure are described in terms of Markush groups or list of alternatives, those skilled in the art will recognize that the disclosure is also thereby described in terms of any individual member, or subgroup of members, of the Markush group or list of alternatives.
Unless specifically defined otherwise, all technical and scientific terms used herein shall be taken to have the same meaning as commonly understood by one of ordinary skill in the art (e.g., in stem cell biology, cell culture, molecular genetics, immunology, immunohistochemistry, protein chemistry, and biochemistry).
Unless otherwise indicated, the recombinant protein, cell culture, and immunological techniques utilized in the present disclosure are standard procedures, well known to those skilled in the art. Such techniques are described and explained throughout the literature in sources such as, J. Perbal, A Practical Guide to Molecular Cloning, John Wiley and Sons (1984), J. Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbour Laboratory Press (1989), T. A. Brown (editor), Essential Molecular Biology: A Practical Approach, Volumes 1 and 2, IRL Press (1991), D. M. Glover and B. D. Hames (editors), DNA Cloning: A Practical Approach, Volumes 1-4, IRL Press (1995 and 1996), and F. M. Ausubel et al. (editors), Current Protocols in Molecular Biology, Greene Pub. Associates and Wiley- Interscience (1988, including all updates until present), Ed Harlow and David Lane (editors) Antibodies: A Laboratory Manual, Cold Spring Harbour Laboratory, (1988), and J. E. Coligan et al. (editors) Current Protocols in Immunology, John Wiley & Sons (including all updates until present).
In the studies described herein, the present inventors have shown that rat and human SCPPPQ1 proteins, and peptides derived from these proteins, interfere with the membrane of bacteria and fungi, and in turn exhibit antibacterial and antifungal activities. As shown in FIG. 15, the known or predicted amino acid sequences of SCPPPQ1 from various species exhibit significant sequence homology/identity with rat and human SCPPPQ1 proteins.
The present disclosure provides an isolated SCPPPQ1 protein or a fragment thereof of at least 5, 6 or 7 amino acids, and the use of such protein or fragment as an antibacterial or antifungal agent.
The present disclosure also provides an isolated polypeptide, peptide or peptidomimetic of 5, 6 or 7 to 100 residues comprising a sequence of formula I:
X1 -X2-X3-X4-X5-X6-X7 -X8-X9-X10-X1 1 -X12-X13-X14-X15-X16-X17-X18-X19 (I) wherein
represents a bond;
XI is L-phenylalanine, D-phenylalanine, or an analog thereof, or is absent;
X2 is L-proline, D-proline, or an analog thereof;
X3 is L-leucine, D-leucine, L-phenylalanine, D-phenylalanine, or an analog thereof, or is absent;
X4 is L-glutamine, D-glutamine, or an analog thereof, or is absent;
X5 is L-glutamine, D-glutamine, L-proline, D-proline, L-leucine, D-leucine or an analog thereof, or is absent;
X6 is L-proline, D-proline, or an analog thereof;
X7 is L-glutamine, D-glutamine, or an analog thereof, or is absent;
X8 is L-alanine, D-alanine, or an analog thereof, or is absent;
X9 is L-proline, D-proline, or an analog thereof;
X10 is any amino acid or amino acid analog, preferably L-glutamine, D-glutamine, L-leucine, D-leucine, L-serine or D-serine, L-valine or D-valine, L-proline, D-proline, or an analog thereof, or is absent;
XI I L-glutamine or D-glutamine, glycine, or an analog thereof, or is absent;
X12 is L-leucine, D-leucine, or an analog thereof, or is absent;
X13 is L-proline, D-proline, or an analog thereof, or is absent;
X14 is L-leucine, D-leucine, or an analog thereof, or is absent;
X15 is L-proline, D-proline, or an analog thereof, or is absent;
X16 is L-isoleucine, D-isoleucine, L-leucine, D-leucine, or an analog thereof, or is absent;
X17 is L-proline, D-proline, L-glutamine or D-glutamine, or an analog thereof, or is absent;
X18 is L-isoleucine, D-isoleucine, L-leucine, D-leucine, L-phenylalanine, D- phenylalanine or an analog thereof, or is absent;
X19 is L- or D-proline, or an analog thereof, or is absent;
or a pharmaceutically acceptable salt thereof.
In an embodiment, X1 is L-phenylalanine, D-phenylalanine, or an analog thereof, preferably L-phenylalanine.
In an embodiment, X2 is L-proline, D-proline, or an analog thereof, preferably L-proline. In an embodiment, X3 is L-leucine, D-leucine, or an analog thereof, preferably L- leucine.
In an embodiment, X4 is absent.
In an embodiment, X5 is absent.
In an embodiment, X6 is L-proline or D-proline, preferably L-proline.
In an embodiment, X7 is L-glutamine or D-glutamine, preferably L-glutamine. In another embodiment, X7 is absent.
In an embodiment, X8 is L-alanine or D-alanine, preferably L-alanine. In another embodiment, X8 is absent.
In an embodiment, X9 is L-proline or D-proline, preferably L-proline.
In an embodiment, X10 is L-glutamine or D-glutamine, preferably L-glutamine. In another embodiment, X10 is absent.
In an embodiment, X1 1 , X12, X13, X14 and/or X15 are absent. In another embodiment, X1 1 , X12, X13, X14 and X15 are absent.
In an embodiment, X16 is L-isoleucine, D-isoleucine, L-leucine or D-leucine, preferably L-isoleucine or L-leucine. In another embodiment, X16 is absent.
In an embodiment, X17 is L-proline, D-proline, L-glutamine or D-glutamine, preferably L-proline or D-proline, more preferably L-proline.
In an embodiment, X18 is L-isoleucine, D-isoleucine, L-leucine, D-leucine, L- phenylalanine, or D-phenylalanine, preferably L-isoleucine, L-leucine or L-phenylalanine. In another embodiment, X18 is absent.
In an embodiment, X19 is L-proline or D-proline, preferably L-proline.
In an embodiment, the polypeptide or peptide is not a naturally-occurring polypeptide or peptide.
In an embodiment, the polypeptide or peptide does not consist of the sequence of SEQ ID NO:30. In an embodiment, the polypeptide or peptide does not consist of the sequence of SEQ ID NO:31 . In an embodiment, the polypeptide or peptide does not consist of the sequence of SEQ ID NO: 1 . In an embodiment, the polypeptide or peptide does not only comprise a naturally-occurring sequence, i.e. comprises at least one amino acid deletion, addition, substitution and/or modification (e.g., amidation, conjugation to a moiety, etc.).
As used herein, the term“polypeptide” is intended to encompass a chain of more than 50 amino acids, and up to 500, 1000, 2000, 3000, 5000 or more amino acids. The term further includes polypeptides which have undergone post-translational modifications, for example, glycosylation, acetylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, or modification by non-naturally occurring amino acids.
The term“peptide” refers to a chain of no more than 50 amino acids, preferably of no more than 30 amino acids.
As used herein, the term "peptidomimetic" refers to a molecule comprising a plurality of amino acid residues (naturally- and/or non-naturally-occurring amino acids, amino acid analogs) joined by a plurality of peptide and/or non-peptide bonds. Peptidomimetics typically retain the polarity, three-dimensional size and functionality (bioactivity) of their peptide equivalents, but one or more of the peptide bonds/linkages have been replaced, often by more stable linkages. Generally, the bond which replaces the amide bond (amide bond surrogate) conserves many or all of the properties of the amide bond, e.g. conformation, steric bulk, electrostatic character, potential for hydrogen bonding, etc. Typical peptide bond replacements include esters, polyamines and derivatives thereof as well as substituted alkanes and alkenes, such as aminomethyl and ketomethylene. For example, domain or polypeptide may have one or more peptide bonds replaced by bonds such as -CH2NH-, -CH2S-, -CH2-CH2-, -CH=CH- (cis or trans), -CH2SO-, -CH(OH)CH2-, or -COCH2-. Such peptidomimetics may have greater chemical stability, enhanced biological/pharmacological properties (e.g., half-life, absorption, potency, efficiency, etc.) and/or reduced antigenicity relative its peptide equivalent. In an embodiment, in the domain of formula I, represents a peptide bond.
The term "amino acid" as used herein includes both L- and D-isomers of the naturally occurring amino acids as well as other amino acids (e.g., naturally-occurring amino acids, non- naturally-occurring amino acids, amino acids which are not encoded by nucleic acid sequences, etc.) used in peptide chemistry to prepare synthetic analogs of peptides. Examples of naturally- occurring amino acids are glycine, alanine, valine, leucine, isoleucine, serine, threonine, etc. Other amino acids include for example non-genetically encoded forms of amino acids, as well as a conservative substitution of an L-amino acid. Naturally-occurring non-genetically encoded amino acids include, for example, beta-alanine, 3-amino-propionic acid, 2,3-diamino propionic acid, alpha-aminoisobutyric acid (Aib), 4-amino-butyric acid, /V-methylglycine (sarcosine), hydroxyproline, ornithine (e.g., L-ornithine), citrulline, f-butylalanine, f-butylglycine, N- methylisoleucine, phenylglycine, cyclohexylalanine, norleucine (Nle), norvaline, 2- napthylalanine, pyridylalanine, 3-benzothienyl alanine, 4-chlorophenylalanine, 2- fluorophenylalanine, 3-fluorophenylalanine, 4-fluorophenylalanine, penicillamine, 1 , 2,3,4- tetrahydro-isoquinoline-3-carboxylic acid, beta-2-thienylalanine, methionine sulfoxide, L- homoarginine (Hoarg), N-acetyl lysine, 2-amino butyric acid, 2-amino butyric acid, 2,4,- diaminobutyric acid (D- or L-), p-aminophenylalanine, /V-methylvaline, homocysteine, homoserine (HoSer), cysteic acid, epsilon-amino hexanoic acid, delta-amino valeric acid, or 2,3- diaminobutyric acid (D- or L-), etc. These amino acids are well known in the art of biochemistry/peptide chemistry.
The term“analog” when used in reference to an amino acid refers to synthetic amino acids providing similar side chain functionality (i.e. , structurally similar) as the“native” amino acid and which can be substituted for an amino acid in the formation of a peptidomimetic. Amino
acid analogs include, without limitation, b-amino acids and amino acids, in which the amino or carboxy group is substituted by a similarly reactive group or other groups (e.g., substitution of the primary amine with a secondary or tertiary amine, or substitution of the carboxy group with an ester).
For example, aromatic amino acids may be replaced with D- or L-naphthylalanine, D- or L-homophenylalanine, D- or L-phenylglycine, D- or L-2-thienylalanine, D- or L-1 -, 2-, 3-, or 4- pyrenylalanine, D- or L-3-thienylalanine, D- or L-(2-pyridinyl)-alanine, D- or L-(3-pyrid inyl)- alanine, D- or L-(2-pyrazinyl)-alanine, D- or L-(4-isopropyl)-phenylglycine, D-(trifluoromethyl)- phenylglycine, D-(trifluoromethyl)-phenylalanine, D-p-fluorophenylalanine, D- or L-p- biphenylalanine D-or L-p-methoxybiphenylalanine, D- or L-2-indole(alkyl)alanines, and D- or L- alkylalanines wherein the alkyl group is substituted or unsubstituted methyl, ethyl, propyl, hexyl, butyl, pentyl, isopropyl, iso-butyl, or iso-pentyl.
Analogs of alanine include b-alanine, aminoisobutyric acid (a or b), methylalanine and f-butylalanine.
Analogs of phenylalanine include b-methyl-phenylalanine, b-hydroxyphenylalanine, a- methyl-3-methoxy-DL-phenylalanine, a-methyl-D-phenylalanine, a-methyl-L-phenylalanine, 2,4- dichloro-phenylalanine, 2-(trifluoromethyl)-D-phenylalanine, 2-(trifluoromethyl)-L-phenylalanine, 2-bromo-D- phenylalanine, 2-bromo-L-phenylalanine, 2-chloro-D-phenylalanine, 2-chloro-L- phenylalanine, 2-cyano-D-phenylalanine, 2-cyano-L-phenylalanine, 2-fluoro-D-phenylalanine, 2- fluoro-L-phenylalanine, 2-methyl-D-phenylalanine, 2-methyl-L-phenylalanine, 2-nitro-D- phenylalanine, 2-nitro-L-phenylalanine, 2,4,5-trihydroxy-phenylalanine, 3,4,5-trifluoro-D- phenylalanine, 3,4,5-trifluoro-L-phenylalanine, 3,4-dichloro-D-phenylalanine, 3,4-dichloro-L- phenylalanine, 3,4-difluoro-D-phenylalanine, 3,4-difluoro-L-phenylalanine, 3,4-dihydroxy-L- phenylalanine, 3,4-dimethoxy-L-phenylalanine, 3-(trifluoromethyl)-D-phenylalanine, 3-
(trifluoromethyl)-L-phenylalanine, 3-amino-L-tyrosine, 3-bromo-D-phenylalanine, 3-bromo-L- phenylalanine, 3-chloro-D-phenylalanine, 3-chloro-L-phenylalanine, 3-cyano-D-phenylalanine, 3 cyano-L-phenylalanine, 3-fluoro-D-phenylalanine, 3-fluoro-L-phenylalanine, 3-iodo-D- phenylalanine, 3-iodo-L-phenylalanine, 3-methyl-D-phenylalanine, 3-methyl-L-phenylalanine, 3- nitro-D-phenylalanine, 3-nitro-L-phenylalanine, 4-(trifluoromethyl)-D-phenylalanine, 4-
(trifluoromethyl)-L-phenylalanine, 4-amino-D- phenylalanine, 4-amino-L-phenylalanine, 4- benzoyl-D-phenylalanine, 4-benzoyl-L-phenylalanine, 4-bis(2-chloroethyl)amino-L- phenylalanine, 4-bromo-D-phenylalanine, 4-bromo-L-phenylalanine, 4-chloro-D-phenylalanine, 4-chloro-L-phenylalanine, 4-cyano-D-phenylalanine, 4-cyano-L-phenylalanine, 4-fluoro-D- phenylalanine, 4-fluoro-L-phenylalanine, 4-iodo-D-phenylalanine, 4-iodo-L-phenylalanine, homophenylalanine, 3,3-diphenylalanine.
Examples of analogs of proline include hydroxyproline, ketoproline, trans/cis-3- hydroxyproline, trans/cis-4-hydroxyproline, trans/cis-3-aminoproline, trans/cis-4-aminoproline, methylproline, 3,4-dehydroproline, L-pipecolic acid, 3-thiaproline and 4-thiaproline.
Example of analogs of glutamine include acivicin, antipyrimidine, 6-diazo-5-oxo-L- norleucine (DON), and theanine.
In an embodiment, the polypeptide, peptide or peptidomimetic comprises only L- or D- amino acids. In a further embodiment, the polypeptide, peptide or peptidomimetic comprises only L-amino acids.
In embodiments, the polypeptide, peptide, peptidomimetic or domain disclosed herein may include substitutions of functionally equivalent amino acid residues. For example, one or more amino acid residues within the sequence can be substituted by another amino acid of a similar polarity (having similar physico-chemical properties) which acts as a functional equivalent, resulting in a silent alteration. Substitution for an amino acid within the sequence may be selected from other members of the class to which the amino acid belongs. For example, positively charged (basic) amino acids include arginine, lysine and histidine (as well as homoarginine and ornithine). Nonpolar (hydrophobic) amino acids include leucine, isoleucine, alanine, phenylalanine, valine, proline, tryptophan and methionine. Uncharged polar amino acids include serine, threonine, cysteine, tyrosine, asparagine and glutamine. Negatively charged (acidic) amino acids include glutamic acid and aspartic acid. The amino acid glycine may be included in either the nonpolar amino acid family or the uncharged (neutral) polar amino acid family. Substitutions made within a family of amino acids are generally understood to be conservative substitutions.
In an embodiment, the polypeptide, peptide or peptidomimetic comprises a sequence of at least 5, 6, 7 or 8 residues from one of the following sequences: FPLPPQPP (SEQ ID NO: 1 ), FPLPQAPLIPIP (SEQ ID NO:2), FPQPPQLQIP (SEQ ID NO:3), PLPPSPP (SEQ ID NO:4), FPFQQPPLIPFP (SEQ ID NO:5), FPPPVGLPLIPLP (SEQ ID NO:6), FPPPPQIPIP (SEQ ID NO:7), FPLPRQIPIP (SEQ ID NO:8), FPPPPQLPLIPIP (SEQ ID NO:9), FPLPQAPLIPIP (SEQ ID NO: 10), or a variant thereof having at least 70% sequence identity with the at least 7 or 8 residues (e.g., having 1 or 2 amino acid substitution(s)). In an embodiment, the polypeptide, peptide or peptidomimetic comprises a sequence of at least 5, 6, 7 or 8 residues from any one of SEQ ID NOs: 1 -10, or a variant thereof having at least 85% sequence identity with the at least 5, 6, 7 or 8 residues (e.g., having 1 amino acid substitution). In a further embodiment, the polypeptide, peptide or peptidomimetic comprises or consists of a sequence of at least 5, 6, 7 or 8 residues from any one of SEQ ID NOs: 1 -10. In a further embodiment, the polypeptide, peptide or peptidomimetic comprises or consists of the sequences of any one of SEQ ID NOs: 1 10
In an embodiment, the polypeptide, peptide or peptidomimetic comprises the sequence FPLPPQPP (SEQ ID NO: 1 ), FPLPQAP (SEQ ID NO: 1 1 ), or a variant thereof having at least 70% sequence identity with these sequences (e.g., having 1 or 2 amino acid substitution(s)). In another embodiment, the polypeptide, peptide or peptidomimetic comprises the sequence FPLPPQPP (SEQ ID NO:1), FPLPQAP (SEQ ID NO: 1 1), FPLPPQPPRR (SEQ ID NO: 58), FPLPQAPRR (SEQ ID NO: 59), PPQVPPFFPQVLFPLPPQPP (SEQ ID NO: 60), or PPQVPPFFPQVLFPLPPQPPRR (SEQ ID NO: 61), or a variant thereof having at least 85% sequence identity with these sequences (e.g., having 1 amino acid substitution).
In an embodiment, the polypeptide, peptide or peptidomimetic comprises or consist the sequence FPLPPQPP (SEQ ID NO:1 ), FPLPQAP (SEQ ID NO:1 1 ), FPLPPQPPRR (SEQ ID NO: 58), FPLPQAPRR (SEQ ID NO: 59), PPQVPPFFPQVLFPLPPQPP (SEQ ID NO: 60), or PPQVPPFFPQVLFPLPPQPPRR (SEQ ID NO: 61). In an embodiment, the polypeptide, peptide or peptidomimetic comprises or consist of the sequence FPLPPQPP (SEQ ID NO: 1). In an embodiment, the polypeptide, peptide or peptidomimetic comprises or consist of the sequence FPLPQAP (SEQ ID NO: 1 1 ). In an embodiment, the polypeptide, peptide or peptidomimetic comprises or consist of the sequence FPLPPQPPRR (SEQ ID NO: 58). In an embodiment, the polypeptide, peptide or peptidomimetic comprises or consist of the sequence FPLPQAPRR (SEQ ID NO: 59). In an embodiment, the polypeptide, peptide or peptidomimetic comprises or consist of the sequence PPQVPPFFPQVLFPLPPQPP (SEQ ID NO: 60). In an embodiment, the polypeptide, peptide or peptidomimetic comprises or consist of the sequence PPQVPPFFPQVLFPLPPQPPRR (SEQ ID NO: 61).
In embodiments, the above-mentioned polypeptide, peptide or peptidomimetic comprises, further to the sequence of formula I defined above, one more amino acids (naturally occurring or synthetic) covalently linked to the amino- and/or carboxy-termini of said domain. In an embodiment, the polypeptide, peptide or peptidomimetic has 90 amino acids or less, 85 amino acid or less, 80 amino acid or less, 75 amino acids or less, or 70 amino acids or less. In an embodiment, the above-mentioned polypeptide, peptide or peptidomimetic comprises at least 7 or 8 amino acids. In embodiments, the above-mentioned polypeptide, peptide or peptidomimetic comprises at least 9, 10, 1 1 , 12, 13, 14, 15, 20, 25 or 30 amino acids. In embodiments, the above-mentioned polypeptide, peptide or peptidomimetic comprises from 7 to 30, 25 or 20 amino acids.
Thus, the polypeptide, peptide or peptidomimetic may comprise, e.g., 1 , 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40 or 50 additional amino acids at the N- and/or C-terminal end of the sequence of formula I defined above. In an embodiment, the additional amino acid(s) at the N- and/or C-terminal end of the sequence of formula I defined above are/is one or more basic amino acids, e.g., arginine (Arg) or lysine (Lys). In an embodiment, the polypeptide, peptide or peptidomimetic comprise 1 to 10 or 1 to 5 additional basic amino acids at the N- and/or C-
terminal end, preferably at the C-terminal end, of the sequence of formula I defined above. In another embodiment, the additional amino acid(s) at the N- and/or C-terminal end of the sequence of formula I defined above is/are one or more cysteine residues.
In embodiments, the above-mentioned polypeptide, peptide or peptidomimetic comprises or consists of the sequence FPLPPQPPRR (SEQ ID NO: 58), FPLPQAPRR (SEQ ID NO: 59), PPQVPPFFPQVLFPLPPQPP (SEQ ID NO: 60), or PPQVPPFFPQVLFPLPPQPPRR (SEQ ID NO: 61 ).
In embodiments, the above-mentioned polypeptide, peptide or peptidomimetic includes the above-noted sequence of formula I ( e.g . , a sequence comprising at least 7 or 8 residues from the sequence of any one of SEQ ID NOs: 1-10 underlined below) and comprises at least 10, 15 or 20 contiguous amino acids from the amino acid sequences:
LPIPLEQYAE SSSEQRFIFY PPQVPPFFPQ VLFPLPPQPP LVLTPNDLIA LLIAILNQLG ILFP
(SEQ ID NO:12);
LPIPLGQSGG SSSEQRFNLY PPQILPFFPQ FPLPQAPLIP IPFPFPFDPN QVLTPNQLLA
LITSILNQLQ GFLGR (SEQ ID NO:13);
LPIPLEQYAE SSSEQRFIFY PPQVPPFFPQ VLFPLPPQPP LISIPFPYNP NQVLTPNDLI
ALLIAILNQL GNFSGT (SEQ ID NO: 14);
LPIPLEQYAE SSSEQRFIFY PPQVPPFFPQ VLFPLPPQPP LISIPFPYNP NQVLTPNDLI
ALLIAILNQL G (SEQ ID NO: 15);
LPVPLEQYAE SSSEQRFIFY PPQVPPFFPQ VPFPLPPQPP LTSIPFPYNP NQVLTPNDLI ALLIAILNQL GNFFGT (SEQ ID NO:16);
LPIPLEQYAE SSSEQRFIFY PPQVPPFFPQ VPFPLPPQPP LTSIPFPYNP NQVLTPNDLI ALLIAILNQL GNFFGT (SEQ ID NO:17);
LPIPLEQYAE SSSEQRFIFY PPQVPPFFPQ VPFPLPPQPP LISIPFPYNP NQVLTPNDLI
ALLIAILNQL GNFFGT (SEQ ID NO:18);
LPIPLEQYAE SSSEQRFIFY PPQVPPFFPQ VPFPLPPQPP LISIPFPFPYNP YQVLTPNDLI ALLIAILNQL GNFFGT (SEQ ID NO:19);
LLIPFEQSRG RSSEQGLNFY PLQVPPFFPQ IPFPLPPLPL IPIPFPSNPN QVLTPNDIIA LITTILNQLG (SEQ ID NO:20);
LPIPLEQSGG SSSAQRFNFF PLQVSPFFPQ ILFPQPPQLQ IPILFPYNPN QVLTPNEFIM LITSVLNQLG GFLG (SEQ ID NO:21 );
PPIPVLPAST SSSEQRFTFY PPQVPPLFPQ IPLPPSPPLI PIPLPFPYDP SQALTPNDFI ALITSILNQL GGYLV (SEQ ID NO:22);
LPIPLGQSAG SSSEQRFNFF PPQILPFFPQ FPFQQPPLIP FPFPFPFDPN QVLTPNQLIA LITSILNQLQ GFLGK (SEQ ID NO:23);
LPVPLEQSGG SSSAQRFNFY PPRGAPFFPQ IPFPPPVGLP LIPLPFPFPN NPNQVLTLND PIALI ISILN QLG (SEQ ID NO:24);
LPIPLEPSGG SSSGQRFGFY PPQGPSFFPQ ILFPPPPQI PIPFPFPYYP NQVLTLNDLI TLITSILNE LEGFFGI (SEQ ID NO:25);
LPIPLEQYGG GSSEQRFGFY PPQGPPFFPQ ILFPPPPQL PLPIPIPLPF PFDPNQVLTL NDLITLITSI LNQLGGFFGI (SEQ ID NO:26);
LPIRLEQSGG SSSGQRFNFY PPQGPPFFPP IPFPLPRQIP IPLPFPYDPN QVLTPNNLIL LITAILNQLG GFFG (SEQ ID NO:27);
LPSPFEQSGG HSSGQRFNFH PPQGPLFFPQ IPFPPPPQLP LIPIPFPIPN NPNQVPTLND LIALIISVLN QLGGFFGIYP SKPQT (SEQ ID NO:28);
LPIPLGQSGG SSSEQRFNLY APQILPFFPP FPLPQAPLIP IPFPFPFDPN QVLTPNQLLE LITSILNQLQ GFLGR (SEQ ID NO:29) or
LPIPLEQYAE SSSEQRFIFY PPQVPPFFPQ VLFPLPPQPP LISIPFPFPY NPNQVLTPND LIALLIAILN QLGNFSGT (SEQ ID NO:62).
In embodiments, the above-mentioned polypeptide, peptide or peptidomimetic includes the above-noted sequence of formula I and comprises at least 25, 30, 35, 40, 45, 50, 55, or 60 contiguous amino acids from the amino acid sequences of any one of SEQ ID NOs: 12-29. In an embodiment, the contiguous amino acids are the amino acids immediately N- and/or C- terminal from the above-noted domain (underlined).
In another embodiment, the above-mentioned polypeptide, peptide or peptidomimetic includes the above-noted sequence of formula I and comprises an amino acid sequence having at least 70% sequence identity with the sequences of any one of SEQ ID NOs: 12-29. In another embodiment, the above-mentioned polypeptide, peptide or peptidomimetic includes the above-noted sequence of formula I and comprises an amino acid sequence having at least 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with the sequences of any one of SEQ ID NOs: 12-29. In another embodiment, the above-mentioned polypeptide, peptide or peptidomimetic comprises the amino acid sequence of any one of SEQ ID NOs: 12- 29. In another embodiment, the above-mentioned polypeptide, peptide or peptidomimetic does not comprises the sequences of the signal peptide depicted in FIG. 14A (underlined).
In embodiments, the N- and/or C-terminal amino acids of the above-mentioned polypeptide, peptide or peptidomimetic (or salt thereof) may be modified, for example by amidation, acetylation, acylation or any other modifications known in the art. Accordingly, in another aspect, the present disclosure provides a polypeptide, peptide or peptidomimetic (or salt thereof) of formula II:
Z1-[polypeptide, peptide or peptidomimetic as defined above]-Z2 (II);
wherein Z1 is H (i.e. the peptide or peptidomimetic has a native NH2 terminal) an amino-terminal modification; and Z2 is OH (i.e. the peptide or peptidomimetic has a native COOH terminal) or a carboxy-terminal modification. In an embodiment, the amino terminal residue (i.e., the free amino group at the N-terminal end) of the peptide or peptidomimetic (or
salt thereof) is modified ( e.g ., for protection against degradation), for example by covalent attachment of a moiety/chemical group (Z1). In an embodiment, the amino-terminal modification (Z1) is a C1-C16 or C3-Ci6 acyl group (linear or branched, saturated or unsaturated), in a further embodiment, a saturated Ci-C6 acyl group (linear or branched) or an unsaturated C3-C6 acyl group (linear or branched), in a further embodiment an acetyl group (CH3-CO-, Ac). In another embodiment, the peptide or peptidomimetic (or salt thereof) has a native NH2 terminal, i.e. Z1 is H. In an embodiment, the carboxy terminal residue (i.e., the free carboxy group at the C- terminal end of the peptide) of the peptide or peptidomimetic (or salt thereof) is modified (e.g. , for protection against degradation). In an embodiment, the modification is an amidation (replacement of the OH group by a NH2 group), thus in such a case Z2 is a NH2 group.
The polypeptide, peptide or peptidomimetic (or salt thereof) described herein may further comprise modifications that confer additional biological properties to the polypeptide, peptide or peptidomimetic such as protease resistance, plasma protein binding, increased half- life in vivo, binding to gum or teeth, intracellular penetration, etc. Such modifications include, for example, covalent attachment of fatty acids (e.g. , C6-Ci8) to the polypeptide, peptide or peptidomimetic, attachment to proteins such as albumin (see, e.g., U.S. Patent No. 7,268, 1 13); glycosylation, biotinylation or PEGylation (see, e.g., U.S. Patent Nos. 7,256,258 and 6,528,485). PEGylation may be carried out using an acylation reaction or an alkylation reaction with a reactive polyethylene glycol molecule. Methods for peptide PEGylation are disclosed, for example, in Roberts et al., Chemistry for peptide and protein PEGylation, Advanced Drug Delivery Reviews, Volume 64, Supplement, December 2012, Pages 1 16-127). In an embodiment, the polypeptide, peptide, peptidomimetic or salt thereof is conjugated to a polyethylene glycol (PEG) chain/moiety (i.e. is PEGylated). The term “PEG chain” refers to polymers of ethylene glycol (represented by the general formula H(OCH2CH2)nOH, where n is an integer of 2, 3, 4, 5, 6, 7, 8, 9, or more) which are commercially produced with different molecular weights (e.g., about 200-50,000 Da, 500-40,000 Da, 1000-30,000 Da or 2000-10,000 Da). PEGylated peptides/peptidomimetics may be prepared by modifying certain amino acids in the polypeptide, peptide, peptidomimetic with a suitable group-reactive reagent. For example, a Cys side chain may be modified with a thiol-reactive agent, or a Lys side chain may be modified with an amine-reactive agent. In another embodiment, the polypeptide, peptide or peptidomimetic (or salt thereof) described herein is conjugated to a gum- or tooth-targeting agent, for example bisphosphonates (BP), diphosphoserine and pyrophosphate that have high affinity for hydroxyapatite (HAP), a major constituent of teeth.
In embodiments, the above-mentioned polypeptide, peptide or peptidomimetic is in the form of a salt, e.g., a pharmaceutically acceptable salt. As used herein the term "pharmaceutically acceptable salt" refers to salts of compounds that retain the biological activity of the parent compound, and which are not biologically or otherwise undesirable. Such salts can
be prepared in situ during the final isolation and purification of the analog, or may be prepared separately by reacting a free base function with a suitable acid. Many of the peptides or peptidomimetics disclosed herein are capable of forming acid and/or base salts by virtue of the presence of amino and/or carboxyl groups or groups similar thereto.
Pharmaceutically acceptable acid addition salts may be prepared from inorganic and organic acids. Representative acid addition salts include, but are not limited to acetate, adipate, alginate, citrate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, camphorate, camphor sulfonate, decanoate, digluconate, glycerophosphate, hemisulfate, heptanoate, hexanoate, fumarate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethansulfonate (isothionate), lactate, maleate, methane sulfonate, nicotinate, 2-naphthalene sulfonate, octanoate, oxalate, palmitoate, pectinate, persulfate, 3-phenylpropionate, picrate, pivalate, propionate, succinate, tartrate, thiocyanate, phosphate, glutamate, bicarbonate, p- toluenesulfonate, and undecanoate. Salts derived from inorganic acids include hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like. Salts derived from organic acids include acetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, malic acid, malonic acid, succinic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluene-sulfonic acid, salicylic acid, and the like. Examples of acids which can be employed to form pharmaceutically acceptable acid addition salts include, for example, an inorganic acid, e.g., hydrochloric acid, hydrobromic acid, sulphuric acid, and phosphoric acid, and an organic acid, e.g., oxalic acid, maleic acid, succinic acid, and citric acid.
Basic addition salts also can be prepared by reacting a carboxylic acid-containing moiety with a suitable base such as the hydroxide, carbonate, or bicarbonate of a pharmaceutically acceptable metal cation or with ammonia or an organic primary, secondary, or tertiary amine. Pharmaceutically acceptable salts include, but are not limited to, cations based on alkali metals or alkaline earth metals such as lithium, sodium, potassium, calcium, magnesium, and aluminum salts, and the like, and nontoxic quaternary ammonia and amine cations including ammonium, tetramethylammonium, tetraethylammonium, methylammonium, dimethylammonium, trimethylammonium, triethylammonium, diethylammonium, and ethylammonium, amongst others. Other representative organic amines useful for the formation of base addition salts include, for example, ethylenediamine, ethanolamine, diethanolamine, piperidine, piperazine, and the like. Salts derived from organic bases include, but are not limited to, salts of primary, secondary and tertiary amines.
The peptide or polypeptide described herein may be produced by expression in a host cell comprising a nucleic acid encoding the peptide or polypeptide (recombinant expression) or by chemical synthesis (e.g., solid-phase peptide synthesis). Polypeptides, peptides and peptidomimetics can be readily synthesized by manual and automated solid phase procedures
well known in the art. Suitable syntheses can be performed for example by utilizing "t-Boc" or "Fmoc" procedures. Techniques and procedures for solid phase synthesis are described in for example Solid Phase Peptide Synthesis: A Practical Approach, by E. Atherton and R. C. Sheppard, published by IRL, Oxford University Press, 1989. Alternatively, the polypeptides, peptides and peptidomimetics may be prepared by way of segment condensation, as described, for example, in Liu et al., Tetrahedron Lett. 37: 933-936, 1996; Baca et al., J. Am. Chem. Soc. 117: 1881 -1887, 1995; Tam et al., Int. J. Peptide Protein Res. 45: 209-216, 1995; Schnolzer and Kent, Science 256: 221 -225, 1992; Liu and Tam, J. Am. Chem. Soc. 116: 4149-4153, 1994; Liu and Tam, Proc. Natl. Acad. Sci. USA 91 : 6584-6588, 1994; and Yamashiro and Li, Int. J. Peptide Protein Res. 31 : 322-334, 1988). Other methods useful for synthesizing the polypeptides, peptides and peptidomimetics are described in Nakagawa et al., J. Am. Chem. Soc. 107: 7087-7092, 1985.
Polypeptides and peptides comprising naturally occurring amino acids encoded by the genetic code may also be prepared using recombinant DNA technology using standard methods. Polypeptides and peptides produced by recombinant technology may be modified (e.g., N-terminal acylation [e.g. , acetylation], C-terminal amidation), using methods well known in the art. Therefore, in embodiments, in cases where a polypeptide or peptide described herein contains naturally occurring amino acids encoded by the genetic code, the polypeptide or peptide may be produced using recombinant methods, and may in embodiments be subjected to for example the just-noted modifications (e.g., acylation, amidation). Accordingly, in another aspect, the present disclosure further provides a nucleic acid encoding the above-mentioned polypeptide or peptide. The disclosure also provides a vector or plasmid comprising the above- mentioned nucleic acid.
In yet another aspect, the present disclosure provides a cell (e.g. , a host cell) comprising the above-mentioned nucleic acid and/or vector. The present disclosure further provides a recombinant expression system, vectors and host cells, such as those described above, for the expression/production of a polypeptide or peptide described herein, using for example culture media, production, isolation and purification methods well known in the art.
The polypeptide, peptide or peptidomimetic (or salt thereof) described herein can be purified by many techniques of polypeptide/peptide purification well known in the art, such as reverse phase chromatography, high performance liquid chromatography (HPLC), ion exchange chromatography, size exclusion chromatography, affinity chromatography, gel electrophoresis, and the like. The actual conditions used to purify a particular peptide or peptide analog will depend, in part, on synthesis strategy and on factors such as net charge, hydrophobicity, hydrophilicity, and the like, and will be apparent to those of ordinary skill in the art. For affinity chromatography purification, any antibody that specifically binds the peptide or peptidomimetic may for example be used.
Compositions
In another aspect, the present disclosure provides a composition comprising the above-mentioned polypeptide, peptide, peptidomimetic or salt thereof and a carrier or excipient, in a further embodiment a pharmaceutically acceptable carrier or excipient. Supplementary active compounds can also be incorporated into the compositions. The carrier/excipient can be suitable, for example, for oral, mucosal, intravenous, parenteral, subcutaneous, intramuscular, intracranial, intraorbital, ophthalmic, intraventricular, intracapsular, intraspinal, intrathecal, epidural, intracisternal, intraperitoneal, intranasal or pulmonary (e.g., aerosol) administration. Such compositions may be prepared in a manner well known in the pharmaceutical art (see Remington: The Science and Practice of Pharmacy, by Loyd V Allen, Jr, 2012, 22nd edition, Pharmaceutical Press; Handbook of Pharmaceutical Excipients, by Rowe et al., 2012, 7th edition, Pharmaceutical Press).
An "excipient," as used herein, has its normal meaning in the art and is any ingredient that is not an active ingredient (drug) itself. Excipients include for example binders, lubricants, diluents, fillers, thickening agents, disintegrants, plasticizers, coatings, barrier layer formulations, lubricants, stabilizing agent, release-delaying agents and other components. "Pharmaceutically acceptable excipient" as used herein refers to any excipient that does not interfere with effectiveness of the biological activity of the active ingredients and that is not toxic to the subject, i.e., is a type of excipient and/or is for use in an amount which is not toxic to the subject. Excipients are well known in the art, and the present system is not limited in these respects. In certain embodiments, the excipients, including for example and without limitation, one or more binders (binding agents), buffers, thickening agents, surfactants, diluents, saline solution, release-delaying agents, colorants, flavoring agents, fillers, disintegrants/dissolution promoting agents, lubricants, plasticizers, silica flow conditioners, glidants, anti-caking agents, anti-tacking agents, stabilizing agents, anti-static agents, swelling agents and any combinations thereof. As those of skill would recognize, a single excipient can fulfill more than two functions at once, e.g., can act as both a binding agent and a thickening agent. As those of skill will also recognize, these terms are not necessarily mutually exclusive.
Useful diluents, e.g., fillers, include, for example and without limitation, dicalcium phosphate, calcium diphosphate, calcium carbonate, calcium sulfate, lactose, cellulose, kaolin, sodium chloride, starches, powdered sugar, colloidal silicon dioxide, titanium oxide, alumina, talc, colloidal silica, microcrystalline cellulose, silicified micro crystalline cellulose and combinations thereof. Fillers that can add bulk to tablets with minimal drug dosage to produce tablets of adequate size and weight include croscarmellose sodium NF/EP (e.g., Ac-Di-Sol); anhydrous lactose NF/EP (e.g., Pharmatose™ DCL 21); and/or povidone USP/EP.
Binder materials include, for example and without limitation, starches (including corn starch and pregelatinized starch), gelatin, sugars (including sucrose, glucose, dextrose and lactose), polyethylene glycol, povidone, waxes, and natural and synthetic gums, e.g. , acacia sodium alginate, polyvinylpyrrolidone, cellulosic polymers (e.g., hydroxypropyl cellulose, hydroxypropyl methylcellulose, methyl cellulose, hydroxyethyl cellulose, carboxymethylcellulose, colloidal silicon dioxide NF/EP (e.g., Cab-O-Sil™ M5P), Silicified Microcrystalline Cellulose (SMCC), e.g., Silicified microcrystalline cellulose NF/EP (e.g., Prosolv™ SMCC 90), and silicon dioxide, mixtures thereof, and the like), veegum, and combinations thereof.
Useful lubricants include, for example, canola oil, glyceryl palmitostearate, hydrogenated vegetable oil (type I), magnesium oxide, magnesium stearate, mineral oil, poloxamer, polyethylene glycol, sodium lauryl sulfate, sodium stearate fumarate, stearic acid, talc and, zinc stearate, glyceryl behapate, magnesium lauryl sulfate, boric acid, sodium benzoate, sodium acetate, sodium benzoate/sodium acetate (in combination), DL-leucine, calcium stearate, sodium stearyl fumarate, mixtures thereof, and the like.
Bulking agents include, for example: microcrystalline cellulose, for example, AVICEL® (FMC Corp.) or EMCOCEL® (Mendell Inc.), which also has binder properties; dicalcium phosphate, for example, EMCOMPRESS® (Mendell Inc.); calcium sulfate, for example, COMPACTROL® (Mendell Inc.); and starches, for example, Starch 1500; and polyethylene glycols (CARBOWAX®).
Disintegrating or dissolution promoting agents include: starches, clays, celluloses, alginates, gums, crosslinked polymers, colloidal silicon dioxide, osmogens, mixtures thereof, and the like, such as crosslinked sodium carboxymethyl cellulose (AC-DI-SOL®), sodium croscarmelose, sodium starch glycolate (EXPLOTAB®, PRIMO JEL®) crosslinked polyvinylpolypyrrolidone (PLASONE-XL®), sodium chloride, sucrose, lactose and mannitol.
Antiadherents and glidants employable in the core and/or a coating of the solid oral dosage form may include talc, starches (e.g., cornstarch), celluloses, silicon dioxide, sodium lauryl sulfate, colloidal silica dioxide, and metallic stearates, among others.
Examples of silica flow conditioners include colloidal silicon dioxide, magnesium aluminum silicate and guar gum.
Suitable surfactants include pharmaceutically acceptable non-ionic, ionic and anionic surfactants. An example of a surfactant is sodium lauryl sulfate. If desired, the pharmaceutical composition to be administered may also contain minor amounts of nontoxic auxiliary substances such as wetting or emulsifying agents, pH-buffering agents and the like, for example, sodium acetate, sorbitan monolaurate, triethanolamine sodium acetate, triethanolamine oleate, etc. If desired, flavoring, coloring and/or sweetening agents may be added as well.
Examples of stabilizing agents include acacia, albumin, polyvinyl alcohol, alginic acid, bentonite, dicalcium phosphate, carboxymethylcellulose (CMC), hydroxypropylcellulose (HPC), colloidal silicon dioxide, cyclodextrins, glyceryl monostearate, hydroxypropyl methylcellulose (HPMC), magnesium trisilicate, magnesium aluminum silicate, propylene glycol, propylene glycol alginate, sodium alginate, carnauba wax, xanthan gum, starch, stearate(s), stearic acid, stearic monoglyceride and stearyl alcohol.
Examples of thickening agent can be for example talc USP/EP, a natural gum, such as guar gum or gum arabic, or a cellulose derivative such as microcrystalline cellulose NF/EP (e.g., Avicel™ PH 102), methylcellulose, ethylcellulose or hydroxyethylcellulose. A useful thickening agent is hydroxypropyl methylcellulose (HPMC), an adjuvant which is available in various viscosity grades.
Examples of plasticizers include: acetylated monoglycerides; these can be used as food additives; Alkyl citrates, used in food packagings, medical products, cosmetics and children toys; Triethyl citrate (TEC); Acetyl triethyl citrate (ATEC), higher boiling point and lower volatility than TEC; Tributyl citrate (TBC); Acetyl tributyl citrate (ATBC), compatible with PVC and vinyl chloride copolymers; Trioctyl citrate (TOC), also used for gums and controlled release medicines; Acetyl trioctyl citrate (ATOC), also used for printing ink; Trihexyl citrate (THC), compatible with PVC, also used for controlled release medicines; Acetyl trihexyl citrate (ATHC), compatible with PVC; Butyryl trihexyl citrate (BTHC, trihexyl o-butyryl citrate), compatible with PVC; Trimethyl citrate (TMC), compatible with PVC; alkyl sulphonic acid phenyl ester, polyethylene glycol (PEG) or any combination thereof. Optionally, the plasticizer can comprise triethyl citrate NF/EP.
Examples of permeation enhancers include: sulphoxides (such as dimethylsulphoxide, DMSO), azones (e.g., laurocapram), pyrrolidones (for example 2-pyrrolidone, 2P), alcohols and alkanols (ethanol, or decanol), glycols (for example propylene glycol and polyethylene glycol), surfactants and terpenes.
Formulations suitable for oral administration may include (a) liquid solutions, such as an effective amount of active agent(s)/composition(s) suspended in diluents, such as water, saline or PEG 400; (b) capsules, sachets or tablets, each containing a predetermined amount of the active ingredient, as liquids, solids, granules or gelatin; (c) suspensions in an appropriate liquid; and (d) suitable emulsions. Tablet forms can include one or more of lactose, sucrose, mannitol, sorbitol, calcium phosphates, corn starch, potato starch, microcrystalline cellulose, gelatin, colloidal silicon dioxide, talc, magnesium stearate, stearic acid, and other excipients, colorants, fillers, binders, diluents, buffering agents, moistening agents, preservatives, flavoring agents, dyes, disintegrating agents, and pharmaceutically compatible carriers. Lozenge forms can comprise the active ingredient in a flavor, e.g., sucrose, as well as pastilles comprising the active ingredient in an inert base, such as gelatin and glycerin or sucrose and acacia emulsions,
gels, and the like containing, in addition to the active ingredient, carriers known in the art. Aqueous suspensions generally contain the active ingredient in finely powdered form or in the form of nano or micronized particles together with one or more suspending agents, such as sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethylcellulose, sodium alginate, polyvinyl-pyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents such as lecithin or condensation products of an alkylene oxide with fatty acids (for example polyoxethylene stearate), or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example polyethylene sorbitan monooleate. The aqueous suspensions may also contain one or more preservatives such as ethyl or propyl p-hydroxybenzoate; anti-oxidants such as ascorbic acid); coloring agents; flavoring agents; and/or sweetening agents such as sucrose, saccharine or aspartame
Formulations for parenteral administration may, for example, contain excipients, sterile water, or saline, polyalkylene glycols such as polyethylene glycol, oils of vegetable origin, or hydrogenated napthalenes. Biocompatible, biodegradable lactide polymer, lactide/glycolide copolymer, or polyoxyethylene-polyoxypropylene copolymers may be used to control the release of the compounds. Other potentially useful parenteral delivery systems for compounds/compositions of the disclosure include ethylenevinyl acetate copolymer particles, osmotic pumps, implantable infusion systems, and liposomes. Formulations for inhalation may contain excipients, (e.g., lactose) or may be aqueous solutions containing, for example, polyoxyethylene-9-lauryl ether, glycocholate and deoxycholate, or may be oily solutions for administration in the form of nasal drops, or as a gel.
In an embodiment, the above-mentioned polypeptide, peptide, peptidomimetic or salt thereof is formulated as a cream, a gel, a paste (e.g., toothpaste), a solution (e.g., a mouthwash), a chewing gum, a patch, a film, or a strip. The above-mentioned polypeptide, peptide, peptidomimetic or salt thereof may be incorporated into drug delivery systems for the oral cavity, such as bioadhesive formulations and nanoparticulate platforms, that typically comprise a biodegradable polymeric matrix, e.g., a mucoadhesive polymer such as gelatin, chitosan, ethylcellulose, carboxymethyl cellulose (CMC), crosslinked polyacrylic acid (PAA) polymers such as Carbopol™ (CP), poly(DL-lactide) (PLA), poly(lactic-coglycolic acid) (PLGA) (see, e.g., Sanko Nguyen & Marianne Hiorth, Therapeutic Delivery, (2015) 6(5), 595-608).
In another embodiment, the above-mentioned polypeptide, peptide, peptidomimetic or salt thereof is incorporated into a cleaning or detergent composition. Such cleaning or detergent composition may comprise other ingredients such as one or more surfactants (anionic, nonionic, ampholytic, zwitterionic and/or cationic surfactants), enzymes/proteases, stabilizers, conditioning polymers, cleaning polymers, surface modifying polymers, soil flocculating polymers, structurants, emollients, humectants, skin rejuvenating actives, magnesium cations, carboxylic acids, scrubbing particles, bleach and bleach activators, perfumes, malodor control agents, pigments, dyes, opacifiers, beads, pearlescent particles, microcapsules, antibacterial agents, pH adjusters, preservatives, buffering means or water or any other diluents or solvents compatible with the cleaning or detergent composition.
Uses/applications
The above-mentioned polypeptide, peptide, peptidomimetic or salt thereof (or composition comprising same) may be used for various applications including:
• Treatment and prevention of infectious diseases (e.g., bacterial or fungal infections in the eyes, mouth, wounds, and brain),
• Treatment and prevention of post-operation bacterial or fungal infections and systemic propagations (e.g., implants such as dental or orthopedic implants);
• Improvement of post-operation wound healing (e.g. around implants);
• Inhibition of P. gingivalis and/or other periodontal disease-associated pathogens and treatment of its associated diseases including but not limited to Alzheimer's disease, cardiovascular disease, and bacterial pneumonia;
• Treatment and prevention of periodontal disease;
• Surface sterilization or antimicrobial/antifungal surface coating in industry (e.g. , food industry) and hospital applications (e.g., surgical tools, medical devices, and areas);
• Antibacterial/antifungal solutions (e.g., hand gels, cleaning surfaces solution);
• Health products (e.g., antibacterial/antifungal toothpaste, creams, drops, mouthwash);
• Preservative in food products, beverages, cosmetics (lotions, creams, gels, soaps, shampoos, conditioners, antiperspirants) contact lens products, and food ingredients.
In another aspect, the present disclosure provides a method for preventing or treating a bacterial or fungal infection in a subject comprising administering to the subject an effective amount of the polypeptide, peptide, peptidomimetic or salt thereof (or composition comprising same) comprising a domain of formula I as defined herein.
In another aspect, the present disclosure provides the use of the polypeptide, peptide, peptidomimetic or salt thereof (or composition comprising same) comprising a domain of formula I as defined herein for preventing or treating a bacterial or fungal infection in a subject.
In another aspect, the present disclosure provides the use of the polypeptide, peptide, peptidomimetic or salt thereof (or composition comprising same) comprising a domain of formula I as defined herein for the manufacture of a medicament for preventing or treating a bacterial or fungal infection in a subject.
In another aspect, the present disclosure provides the polypeptide, peptide, peptidomimetic or salt thereof (or composition comprising same) comprising a domain of formula I as defined herein for preventing or treating a bacterial or fungal infection in a subject.
The term“subject” as used herein denotes any animal, preferably a mammal, and more preferably a human. Examples of subjects include humans, non-human primates, rodents, guinea pigs, rabbits, farm animals (e.g., sheep, pigs, goats, cows, horses) and pets (e.g., dogs and cats). Thus, the polypeptide, peptide, peptidomimetic or salt thereof (or composition comprising same) may be used for human health and veterinary applications.
The polypeptide, peptide, peptidomimetic or salt thereof (or composition comprising same) disclosed herein may be useful for the treatment of bacterial infection caused by Grampositive or Gram-negative species, such as Escherichia coli, Pseudomonas aeruginosa, Klebsiella pneumoniae, Acinetobacter baumannii, Enterobacter cloacae, Staphylococcus aureus, Enterococcus faecalis, Enterococcus faecium, Legionella pneumophila, Mycoplasma pneumonia, Acinetobacter haemolyticus, Acinetobacter junii, Acinetobacter Iwoffi, Burkholderia cepacia, Chlamydophila pneumoniae, Clostridium difficile, Enterobacter aerogenes, Enterobacter cloacae, Moraxella catarrhalis, Neisseria gonorrhoeae, Neisseria meningitides, Proteus mirabilis, Proteus housed, Citrobacter freundii, Citrobacter kosari, Citrobacter barakii, Seratia marcescens, Klebsiella oxytoca, Morganella morganii, Helicobacter pylori, Mycobacterium tuberculosis, Fusobacterium nucleatum, Prevotella intermedia, Treponema denticola, Porphyromonas gingivalis, Bacillus subtilis, Bacillus thuringiensis. In an embodiment, the bacteria are pathogenic, Gram-negative anaerobic bacteria.
In an embodiment, the bacterial or fungal infection causes a periodontal disease. The term“periodontal disease” refers to an infection caused by microorganisms that colonize the tooth or implant surface at or below the gingival margin. In an embodiment, the bacteria causing the periodontal disease comprises Fusobacterium nucleatum, Prevotella intermedia, Treponema denticola and/or Porphyromonas gingivalis but are not limited to this list.
In an embodiment, the periodontal disease is periimplantitis, gingivitis, periodontitis or peri-implant mucositis.
In an embodiment, the method/use disclosed herein prevents or reduces the risk of developing a disease associated with a bacterial or fungal infection. In an embodiment, the
method/use disclosed herein prevents or reduces the risk of developing a disease associated with Porphyromonas gingivalis infection, such as cardiovascular or respiratory disease, diabetes, or Alzheimer’s disease.
For the treatment of oral/mouth infections, the polypeptide, peptide, peptidomimetic or salt thereof may be incorporated with one or more excipients in the form of a mouthwash, dentifrice, gel, buccal tablet, oral spray, a chewing gum, a patch, a film, a strip or sublingual orally-administered formulation. For example, a mouthwash may be prepared incorporating the active ingredient in the required amount in an appropriate solvent, such as a sodium borate solution (Dobell's Solution). Alternatively, the active ingredient may be incorporated into an oral solution such as one containing sodium borate, glycerin and potassium bicarbonate, or dispersed in a dentifrice, or added in a therapeutically effective amount to a composition that may include water, binders, abrasives, flavoring agents, foaming agents, and humectants. Alternatively, the compositions may be fashioned into a tablet or solution form that may be placed under the tongue or otherwise dissolved in the mouth.
In certain embodiments, the polypeptide, peptide, peptidomimetic or salt thereof is/are delivered locally to sites of periodontal disease via chemical and/or ionic attachment or adhesion to suitable substrates currently used for the localized delivery of antibiotic therapy to treat periodontal disease. Examples of substrates used to deliver localized antibiotic therapy to periodontal sites of disease known in the art include, but are not limited to, biofilms, nanoparticles, suture material, microspheres, polymers, fibers, matrixes, and gels. Commercial examples of substrates applied directly to pockets of periodontal disease via methods known in the art and described above include FDA-approved products such ARESTIN, ATRIDOX, ACTISITE, PERIOCHIP®, ELYZOL, and DENTOMYSIN.
In an embodiment, the Porphyromonas gingivalis infection is a disseminated infection. In an embodiment, the Porphyromonas gingivalis infection is in an aortic valve.
In an embodiment, the above-mentioned treatment comprises the use/administration of more than one (i.e. a combination of) active/therapeutic agent, one of which being the above- mentioned polypeptide, peptide, peptidomimetic or salt thereof (or composition comprising same). The combination of prophylactic/therapeutic agents and/or compositions of the present disclosure may be administered or co-administered (e.g., consecutively, simultaneously, at different times) in any conventional dosage form. Co-administration in the context of the present disclosure refers to the administration of more than one therapeutic in the course of a coordinated treatment to achieve an improved clinical outcome. Such co-administration may also be coextensive, that is, occurring during overlapping periods of time. For example, a first agent may be administered to a patient before, concomitantly, before and after, or after a second active agent is administered. The agents may in an embodiment be combined/formulated in a single composition and thus administered at the same time. In an
embodiment, the above-mentioned polypeptide, peptide, peptidomimetic or salt thereof (or composition comprising same) is used/administered in combination with one or more agent(s) currently used to prevent or treat the disorder in question. For instance, the above-mentioned polypeptide, peptide, peptidomimetic or salt thereof (or composition comprising same) may be co-administered with an anti-bacterial or anti-fungal agent such as a quaternary ammonium compound (QAC) or a pharmaceutically acceptable salt thereof, e.g., benzalkonium chloride, or cetylpyridinium chloride; a guanidine compound or a pharmaceutically acceptable salt thereof, e.g., ampicillin, tetracycline, doxycycline, metronidazole, clindamycin, amoxicillin + Clavulinic acid (Augmentin), azithromycin, erythromycin, metronidazole + amoxicillin, spiramycin, chlorhexidine, alexidine, or polyhexamethylene biguanide (PHMB); hexetidine, triclosan, liniment, fluconazole, itraconazole, amphotericin B, voriconazole, nystatin, clotrimazole, econazole nitrate, miconazole, terbinafine, ketoconazole, enilconazole, boric acid, or miconazole, an anti-inflammatory agent, e.g., a nonsteroidal anti-inflammatory drug such as celecoxib, rofecoxib, valdecoxib, parecoxib, etoricoxib, lumiracoxib, etodolac, aspirin, naproxen, ibuprofen, indomethacin, piroxicam or nabumetone; an anti-pain agent such as a local anesthetic, lidocaine, benzocaine, dibucaine, tetracaine, or proparacaine.
The regimen of administration may affect what constitutes an effective amount. For example, several divided dosages, as well as staggered dosages may be administered daily or sequentially, or the dose may be continuously infused, or may be a bolus injection. Further, the dosages of the therapeutic formulations may be proportionally increased or decreased as indicated by the exigencies of the therapeutic or prophylactic situation.
Administration of the polypeptide, peptide, peptidomimetic or salt thereof (or composition comprising same) of the present disclosure to a patient, such as a mammal, such as a human, may be carried out using known procedures, at dosages and for periods of time effective to treat a disease or disorder in the patient. An effective amount of the therapeutic compound necessary to achieve a therapeutic effect may vary according to factors such as the activity of the particular compound employed; the time of administration; the rate of excretion of the compound; the duration of the treatment; other drugs, compounds or materials used in combination with the compound; the state of the disease or disorder, age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well-known in the medical arts. Dosage regimens may be adjusted to provide the optimum therapeutic response. For example, several divided doses may be administered daily or the dose may be proportionally reduced as indicated by the exigencies of the therapeutic situation. A non-limiting example of an effective dose range for a therapeutic compound is from about 0.01 and 50 mg/kg of body weight/per day. One of ordinary skill in the art would be able to study the relevant factors and make the determination regarding the effective amount of the therapeutic compound without undue experimentation.
The compound can be administered to an animal as frequently as several times daily, or it may be administered less frequently, such as once a day, once a week, once every two weeks, once a month, or even less frequently, such as once every several months or even once a year or less. It is understood that the amount of compound dosed per day may be administered, in non-limiting examples, every day, every other day, every 2 days, every 3 days, every 4 days, or every 5 days. The frequency of the dose is readily apparent to the skilled artisan and depends upon any number of factors, such as, but not limited to, the type and severity of the disease being treated, and the type and age of the animal.
Actual dosage levels of the active ingredients in the pharmaceutical compositions may be varied so as to obtain an amount of the active ingredient that is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient.
A medical doctor, e.g., physician or veterinarian, having ordinary skill in the art may readily determine and prescribe the effective amount of the pharmaceutical composition required. For example, the physician or veterinarian could start doses of the compounds employed in the pharmaceutical composition at levels lower than that required in order to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved.
In particular embodiments, it is especially advantageous to formulate the polypeptide, peptide, peptidomimetic or salt thereof (or composition comprising same) in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the patients to be treated; each unit containing a predetermined quantity of therapeutic compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical vehicle.
The dosage unit forms are dictated by and directly dependent on (a) the unique characteristics of the therapeutic compound and the particular therapeutic effect to be achieved, and (b) the limitations inherent in the art of compounding/formulating such a therapeutic compound for the treatment of a disease or disorder in a patient.
In certain embodiments, the disclosed compositions are administered to the patient in dosages that range from one to five times per day or more. In other embodiments, the compositions are administered to the patient in range of dosages that include, but are not limited to, once every day, every two, days, every three days to once a week, and once every two weeks. It is readily apparent to one skilled in the art that the frequency of administration of the various combination compositions varies from subject to subject depending on many factors including, but not limited to, age, disease or disorder to be treated, gender, overall health, and other factors. Thus, the disclosed subject matter should not be construed to be limited to any particular dosage regime and the precise dosage and composition to be administered to any
patient will be determined by the attending physical taking all other factors about the patient into account.
Compounds disclosed for administration may be in the range of from about 1 pg to about 7,500 mg, about 20 pg to about 7,000 mg, about 40 pg to about 6,500 mg, about 80 pg to about 6,000 mg, about 100 pg to about 5,500 mg, about 200 pg to about 5,000 mg, about 400 pg to about 4,000 mg, about 800 pg to about 3,000 mg, about 1 mg to about 2,500 mg, about 2 mg to about 2,000 mg, about 5 mg to about 1 ,000 mg, about 10 mg to about 750 mg, about 20 mg to about 600 mg, about 30 mg to about 500 mg, about 40 mg to about 400 mg, about 50 mg to about 300 mg, about 60 mg to about 250 mg, about 70 mg to about 200 mg, about 80 mg to about 150 mg, and any and all whole or partial increments therebetween.
In some embodiments, the dose of a compound is from about 0.5 pg and about 5,000 mg. In some embodiments, a dose of a compound used in compositions described herein is less than about 5,000 mg, or less than about 4,000 mg, or less than about 3,000 mg, or less than about 2,000 mg, or less than about 1 ,000 mg, or less than about 800 mg, or less than about 600 mg, or less than about 500 mg, or less than about 200 mg, or less than about 50 mg. Similarly, in some embodiments, a dose of a second compound as described herein is less than about 1 ,000 mg, or less than about 800 mg, or less than about 600 mg, or less than about 500 mg, or less than about 400 mg, or less than about 300 mg, or less than about 200 mg, or less than about 100 mg, or less than about 50 mg, or less than about 40 mg, or less than about 30 mg, or less than about 25 mg, or less than about 20 mg, or less than about 15 mg, or less than about 10 mg, or less than about 5 mg, or less than about 2 mg, or less than about 1 mg, or less than about 0.5 mg, and any and all whole or partial increments thereof.
In another aspect, the present disclosure provides a method for cleaning or disinfecting a surface or an object comprising contacting the surface or the object with an effective amount of the polypeptide, peptide, peptidomimetic, salt or composition (e.g., cleaning or detergent composition) described herein. The cleaning or detergent composition may be applied for a period of 0.1 , 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1 , 2, 3, 4, 5, 10 or 15 minutes to 1 hour, such as from 10 seconds to 15 minutes, such as from 10 seconds to 1 minute, such as from 10 seconds to 5 minutes, such as from 20 seconds to 15 minutes, such as from 1 minute to 5 minutes, such as from 2 minutes to 10 minutes, such as from 20 minutes to 55 minutes such as from 20 to 30 minutes.
MODE(S) FOR CARRYING OUT THE INVENTION
The present invention is illustrated in further details by the following non-limiting examples.
Example 1 : Materials and Methods
Cloning procedures. The sister proteins ODAM and AMTN also belonging to the SCPP cluster were used as control. Truncated versions of SCPPPQ1, AMTN and ODAM genes lacking regions encoding the predicted N-terminal signal sequence were PCR-amplified from human cDNA sequences using primers as previously described. PCR products were cloned into the vector pHT for purification studies. The recombinant pHT plasmids allow to produce recombinant proteins with an in-frame N-terminal hexahistidyl-tag (His-tag) and Tobacco Etch Virus (TEV) protease cleavage site. Escherichia coli strain XL-1 Blue were used as hosts for cloning.
Protein overexpression and purification. BL21 (DE3)-star cells containing either pHT- SCPPPQ1, pHT -AMTN or pHT-ODA were grown at 37 °C and 250 rpm to an optical density at 660 nm (OD66o) around 0.6, and protein expression was induced with 0.1 mM isopropyl-B-D- thiogalactoside (IPTG) for ON at 30 °C and 250 rpm. For SCPPPQ1 , bacterial cells were harvested, suspended in equilibration buffer (50 mM Na2HP0 , 150 mM NaCI, 10 mM imidazole, 6 M urea, pH 7) at 4 °C, and sonicated six times 15 s between 15 s ice incubations. Lysates were centrifuged at 13,400 g and the 6His-tagged protein in the supernatant was bound on nickel-nitriloacetic acid (Ni-NTA)-agarose affinity resin (Qiagen, Valencia, CA, USA) at room temperature. After washing the resin with 10 volumes of binding buffer (50 mM Na2HP04, 300 mM NaCI, 20 mM imidazole, 6 M Urea, pH 7), proteins were eluted with elution buffer (50 mM Na2HP0 , 300 mM NaCI, 300 mM imidazole, 6 M Urea, pH 7). Concentration of the collected fractions were assessed using a biodrop (Montreal Biotech Inc., Dorval, Canada) and fractions were then analyzed by SDS-PAGE and Coomassie blue staining. Proteins were finally dialyzed into 50 mM Na2HP0 , 6 M Urea (pH 7.2) and stored at 4 °C. ODAM and AMTN were purified in the same conditions as SCPPPQ1 but in a buffer without urea.
Western blot gels were acquired using a Bio-Rad ChemiDoc imager and the software Image Lab (Bio-Rad, Hercules, CA, USA). Exposure time for the acquisition were between 1 and 2 s.
Protein and peptide production. All peptides, human SCPPPQ1 and rat SCPPPQ1 were synthesized by LifeTein Inc. The synthetic peptides were resolubilized in 50 mM Na2HP0 buffer, 6 M urea, pH 7 for a final concentration of 1 mg/ml. All tests were performed using the synthetic peptides and proteins.
Bacterial culture. Porphyromonas gingivalis ATCC 33277, Fusobacterium nucleatum ATCC 25586, Prevotella intermedia ATCC 2561 1 and Aggregatibacter actinomycetemcomitans ATCC 29522 were grown anaerobically for 48 h (80% N2, 10% C02, 10% H2) at 37 °C in Todd- Hewitt broth (THB; Becton Dickinson, Canada) supplemented with 0.001 % hemin and 0.0001 % vitamin K. Treponema denticola ATCC 35404 was grown anaerobically for 24 h in liquid medium containing 12.5 mg of brain heart infusion, 10 mg of trypticase, 2.5 mg of yeast extract, 0.5 mg of sodium thioglycolate, 1 mg of L-cysteine, 0.25 mg of L-asparagine, 2 mg of glucose, 6 pg of
thiamine pyrophosphate, and 2 mg of sodium bicarbonate per ml; 2% rabbit serum; and 0.2% volatile fatty acids (19). The volatile fatty acids solution consisted of 0.5 ml each of isobutyric, DL-2-methylbutyric, isovaleric, and valeric acids dissolved in 100 ml of 0.1 N KOH. Escherichia coli, Bacillus subtilis and Bacillus thuringiensis was grown for 24 h in LB broth at 37 °C and 250 rpm, then the solution was diluted and grown around 6 hours to reach OD66o around 1 .
Bacterial incubation with purified proteins. Various concentration of proteins or peptides was exposed in a test tube to 800 pi of a suspension of bacteria at OD66o around 1 for up to 2 h at 37 °C or exposed directly to the bacteria on titanium surfaces for up to 2 h at 37 °C. Various volumes were taken for the different assays.
Western blot analysis. For western blot analysis, 40 pi was sampled every 15 min for western blot analysis using antibodies for rat ODAM (1 :5000), rat AMTN (1 :2000), or rat SCPPPQ1 (1 :2000). Protein alone in the bacteria media was used as a control. Western blot gels were acquired using a Bio-Rad ChemiDoc imager and the software Image Lab (Bio-Rad, Hercules, CA, USA). Exposure time for the acquisition were between 1 and 2 s.
Scanning electron microscopy. Following bacterial incubations in a 1 .5 ml tube and then applied to SEM support stubbs or the bacterial incubations directly applied to titanium surfaces, both bacterial samples were fixed for 1 h at 4°C in 4% Paraformaldehyde and 0.1 % glutaraldehyde in 0.1 M phosphate buffer (PB), pH 7.3, and subsequently rinsed three times with PB. Following fixation, samples were incubated for 1 hour in 1 % osmium tetroxide at RT and then dehydrated through an ethanol series (30%, 50%, 70%, 90%, 95% and two times 100%) followed by drying using a Critical Point Drier CPD300 (Leica Biosystems, Concord, ON, Canada). A FE-SEM Regulus 8230 (Hitachi, Tokyo, Japan) operated at 1 kV was used for high resolution images to observe the effect of the protein to the bacteria.
Fluorescence studies. Some bacteria were stained using the Live/Dead BacLight kit (Thermo Scientific, Waltham, MA, USA) before incubation with SCPPPQ1 . Briefly, 1 .5 mI of Syto9 and 1.5 mI of propidium were added to 800 mI of bacteria at OD66o of 1 . A quantity of proteins (mI) were added to the mixture in order to obtain a final concentration of protein of up to 40 mM. As a control, only the buffer was added to the bacterial suspension. Depending the assay, the suspensions were directly observed or sampled at 15 min, 30 min, 60 min and 120 min. After each sampling, the mixture was fixed using glutaraldehyde to obtain a final concentration of 2.5%. Ten mI of each suspension were finally examined under structured illumination microscopy using an Elyra PS1 microscope (Carl Zeiss, Oberkochen, Germany). Z- stack volumes were acquired using the Structural Illumination Method (SIM) and reconstructed using the Zen Black edition software. For some conditions, counting and intensity analysis of at least 20 representative images were analyzed using ImageJ (NIH, USA).
Immunofluorescence studies. Following bacterial incubations on coverslips with or without proteins, samples were fixed for 30 min at 4°C in 4% Paraformaldehyde and 0, 1 %
glutaraldehyde in 0.1 M phosphate buffer (PB), pH 7.3, and subsequently rinsed three times with PB before to be blocked for 1 h with 5% milk in PB 0.1 M. Samples were then incubated for 1 h with some rabbit antibodies anti-SCPPPQ1 at RT and then incubated 1 h with mouse antibodies Alexa-488 anti-rabbit at RT before to be mounted using prolong for observation. The samples were finally examined under structured illumination microscopy using an Elyra PS1 microscope (Carl Zeiss, Oberkochen, Germany). Z-stack volumes were acquired using the Structural Illumination Method (SIM) and reconstructed using the Zen Black edition software.
Flow cytometry (FACS) analysis. Various concentration of proteins or peptides were exposed in a test tube to 500 pi of a suspension of bacteria at OD66o around 1 for up to 2 h at 37 °C before fixation using 1 % Glutaraldehyde. The suspension was then stained using FM4-64 (Thermo Scientific, Waltham, MA, USA) before analysis using the analyzer FacsAria III SORP (Becton Dickinson, NJ, USA). The collected data were then analyzed using the FlowJo program (Becton Dickinson, NJ, USA).
Toothpaste analysis. Porphyromonas gingivalis was incubated 30 min at 37 °C on titanium surfaces before being mounted of a coverslip which was covered with toothpaste mixed with buffer (control) or rat SCPPPQ1 . To avoid direct contact between the toothpaste and the bacteria, an O-ring was positioned between the coverslip and the titanium disk and the space between was filled with PB 0.1 M (pH 7.2). After a one-hour incubation at 37 °C, the samples on the disks were fixed and prepared for scanning electron microscopy as previously described.
Surfaces absorbed with proteins. Rat SCPPPQ1 , human SCPPPQ1 and buffer (control) were applied 30 min on separate titanium surface for absorption. The excess of liquid was then removed by airflow and the surfaces was dried 30 min at room temperature. Porphyromonas gingivalis was incubated for 1 h at 37 °C before being fixed and prepared for scanning electron microscopy as previously described.
Embedding procedures. Following bacterial incubations, some bacteria were centrifuged and prepared for embedding in Epon (Electron Microscopy Sciences, Hatfield, PA, USA). The bacteria were fixed as previously described and post-fixed with potassium ferrocyanide-reduced osmium tetroxide and then processed for embedding in Epon resin. For some samples, ultrathin sections of 80-100 nm were cut with a diamond knife and transferred onto Formvar®-coated (polyvinyl formate) 200-mesh nickel grids for TEM imaging. Grids were examined in a FEI Tecnai 12 (Eindhoven, The Netherlands) transmission electron microscope operating at 80 kV. Some embedded samples were used for colloidal gold immunolabelling. Briefly, the grids were blocked for 15 min using 0.1 % ovalbumin, then incubated for 1 h with rabbit antibodies anti-SCPPPQ1 at RT, followed by a second blocking step and a final incubation of 30 min with 10 nm gold beads coupled to protein A at RT before to be air dried for observation on the TEM.
Yeast analysis. Candida albicans were grown in Lee and Sabouraud liquid media at 37 °C for 24 h. Various proteins or peptides at a final concentration of 20 mM were incubated with the yeast for 2 h in a test tube before application on titanium surfaces for 30 min. Samples were then fixed for 1 h at 4 °C in 4% paraformaldehyde and 0, 1 % glutaraldehyde in 0.1 M phosphate buffer (PB), pH 7.3 followed by the preparation of the samples for SEM as previously described. The samples were then observed by a FE-SEM Regulus 8230 (Hitachi, Tokyo, Japan) operated at 1 kV. The rat SCPPPQ1 proteins were also prepared in the agar plates at a final concentration of 6 mM. The Candida albicans were then applied to the agar plates for 72 hours. The Unit Forming colony (UFC) formed were then evaluated for their appearances and number, and some UFC were observed under a variable pressure JEOL SEM.
Cell analysis. Ameloblast-like cells LS8 were cultured in DMEM supplemented with 10% heat inactivated fetal bovine serum (FBS) at 37 °C in a 5% C02 atmosphere. 250,000 cells were placed on 18x18mm coverslips and grown overnight. After 24 hours, rat SCPPPQ1 were added to the media cell culture to obtain a final concentration of 20 pM and the same volume of buffer were added to different coverslips as control. After 24 hours of incubation, cells were stained with rhodamine phalloidin (1 : 500, Thermo Scientific, Waltham, MA, USA) to highlight the actin and with Hoechst (1 :2000, Thermo Scientific, Waltham, MA, USA) for the nucleus. Fluorescent samples were acquired on an Axio-lmager (Carl Zeiss, Oberkochen, Germany) using the Zen Blue edition software and images were then analyzed and quantified using ImageJ.
Top-down LC-MS/MS analysis. Samples before and after incubation with P. gingivalis were diluted in 25% ACN 0.3% TFA and loaded onto a 50x4.6 mm PLRP-S 300A column (Agilent Technologies) connected to an Accela pump (Thermo Scientific) and an RTC autosampler (Pal systems). The buffers used for chromatography were 0.1 % formic acid (buffer A) and 100% acetonitrile/0.1 % formic acid (buffer B). Proteins and peptides were eluted with a two-slope gradient at a flow rate of 120 ml/min. Solvent B first increased from 20 to 40% in 70 min and then from 40 to 70% in 5 min. The HPLC system was coupled to Orbitrap Fusion mass spectrometer (Thermo Scientific) through an Electrospray Ion Source. The spray and S-lens voltages were set to 3.6 kV and 50 V, respectively. Capillary temperature was set to 225°C. Full scan MS survey spectra (m/z 500-1700) in profile mode were acquired in the Orbitrap with a resolution of 120,000 with a target value at 5e5. The 3 most intense protein/peptide ions were fragmented in the HCD collision cell and analyzed in the Orbitrap with a target value at 5e5 and a normalized collision energy at 36 V. Target ions selected for fragmentation were dynamically excluded for 25 sec.
For data processing, protein database searching was performed with ProSightPC 4.0.2.1 (Thermo Scientific) against the protein sequences of interest. The mass tolerances for precursor and fragment ions were set to 1000 Da and 20 ppm, respectively. The minimum
number of matching fragments was set to 4 and the only modification used was acetylation of protein N-terminal. The number of matching fragments were quantified and classed by kDa to evaluate the effect of P. gingivalis incubation on each protein.
Example 2: Characterization of the recombinant sBL proteins
In vitro susceptibility of sBL proteins to oral bacteria was assessed. Recombinant sBL proteins were incubated during two hours with various oral bacteria and western immunoblotting analysis revealed no degradation in the presence of F. nucleatum and A. actinomycetemcomitans (FIG. 1A). In contrast, incubation with P. gingivalis, T denticola and P. intermedia resulted in complete degradation of AMTN and ODAM, but not of SCPPPQ1 (FIG. 1 B).
To confirm the degradation of AMTN, ODAM but not of SCPPPQ1 proteins and to determine their respective cleavage sites, mass spectrometry was performed using the top- down assay after incubation of the sBL proteins with P. gingivalis. The bacteria extensively cleaved AMTN into multiple fragments of 1 to 14 kDa (FIG. 2A) and ODAM of 1 to 9 kDa (FIG. 2B). The predominant fragments were from 2 to 5 kDa for both proteins. SCPPPPQ1 protein was not degraded after incubation with P. gingivalis (FIG. 2C), confirming the previously presented SDS-PAGE and western blot analysis observations. Under control condition, at time 0 and when no bacteria were added to the samples for two hours, no degradation was observed.
To verify whether the lack of degradation of SCPPPQ1 was associated with an antibacterial activity, it was next assessed whether one or more of these proteins have anti-bacterial properties. To do so, the number of bacteria (P. gingivalis) was assessed by scanning electron microscopy (SEM) after the incubation. A sample from bacterial suspensions was taken at 120 min, and then deposited on a titanium support for visualization by SEM. The pictures were analyzed by ImageJ. The results demonstrate that whereas there was no change in the number of bacteria in the control (i.e. bacteria incubated with buffer only, FIG. 3A) or following incubation with ODAM (FIG. 3B), a significant reduction of about 75% in the bacteria number was measured after incubation with the rat SCPPPQ1 protein (SEQ ID NO: 31) (FIGs. 3C-D). The bacteria were in very low numbers, separated and were not showing any sign of bacterial division (bacteria attached to each other with a continuous membrane). Similar results were obtained with P. intermedia and T. denticola, providing evidence that the SCPPPQ1 protein has anti-bacterial activity.
Example 3: Determination of the antibacterial activity of recombinant human and rat
SCPPPQ1
Having provided evidence that SCPPPQ1 exhibits antibacterial activity, the extent of the antimicrobial potential of SCPPPQ1 and its mechanism of action was assessed.
Full-length recombinant mature human SCPPPQ1 (SEQ ID NO: 30) and rat SCPPPQ1 (SEQ ID NO: 31) (but without the signal peptide underlined in FIG. 14A) were acquired by synthetizing the protein.
The appearance of bacteria (P. gingivalis) following incubation with the rat and human SCPPPQ1 proteins was assessed by SEM. The results depicted in FIG. 4 show that rat and human SCPPPQ1 protein lead to major disruption of the outer membrane of the bacteria, further evidencing the antibacterial activity of these proteins.
Transmission electron microscopy (TEM) experiments were performed to better elucidate the mechanism of action of SCPPPQ1 against bacteria such P. gingivalis. The results depicted in FIGs. 5A and 5B show that the membrane of the bacteria was affected (notably by swelling and disruption of the outer bacterial membrane) by incubation with rat and human SCPPPQ1 , relative to the control.
A comparison of the total number of bacteria between the control and with human SCPPPQ1 (hSCPPPQI) or rat SCPPPQ1 (rSCPPPQI) was next performed by SEM using bacteria previously affixed to titanium surfaces. Results of the counting in FIG. 6A show that hSCPPPQI has a more pronounced effect on the diminution of the bacterial population. The total number of aggregates in suspension under the different conditions was also assessed by flow cytometry. The results depicted in FIG. 6B (top panel, total volume of aggregates (A.U)) show that rat SCPPPQ1 (rSCPPPQI) creates important aggregates under 20 min that diminish thereafter, whereas human SCPPPQ1 (hSCPPPQI) forms important aggregates that are stable for up to 2 hours. Decomposition of the total volumes of aggregates to determine the percentage of various aggregates formed show that rat SCPPPQ1 and human SCPPPQ1 form bigger aggregates than the control (Buffer) (FIG. 6B, lower panel).
The localization of the human (FIG. 7) and rat (FIG. 8) SCPPPQ1 proteins following treatment of P. gingivalis was assessed by immunofluorescence microscopy. Fluorescence images demonstrate that both proteins were localized at the surface of bacteria following the incubation period. A further characterization of the localization of the rat SCPPPQ1 protein was performed by colloidal gold immunolabelling of the protein and visualization by TEM. The results of FIG. 9 demonstrate that the protein surrounds the bacteria (left) and enters the bacteria after disruption of the membrane (right). This double effect is a distinctive feature that ensures destruction of bacteria, while reducing the possibility of bacteria developing resistance to this protein.
These results indicate that both human and rat SCPPPQ1 can directly attack P. gingivalis via the outer membrane, resulting in the death of the bacteria by a distinctive mechanism, which reduces the possibility of developing bacterial resistance.
Example 4: Further characterization of the activity of SCPPPQ1 and derived peptides against P. gingivalis
Additional experiments were performed to further characterize the activity of SCPPPQ1 and peptides derived therefrom. FIG. 10 illustrates representative transmission and scanning electron micrograph of P. gingivalis incubated with rat SCPPPQ1 . The colloidal gold immunolabelling of rat SCPPPQ1 reveal the localization of the protein on the cells, which creates also a network between the bacteria and scanning electron microscopy of the bacteria confirms the network only in the presence of the protein. Fluorescent images of P. gingivalis show that increasing the concentration of rat SCPPPQ1 decreases the quantity of bacteria, and also that the bacteria create more clusters when the concentration of protein is increased (FIG. 11) .
The antibacterial activity and potential therapeutic capacity of peptides derived from SCPPPQ1 , peptides derived from the predicted (SEQ ID NO: 57) and reported (SEQ ID NO: 30, Genbank accession No. MK322956.1) human SCPPPQ1 protein as well as rat SCPPPQ1 (SEQ ID NO: 31), was tested. The sequences of human and rat SCPPPQ1 and derived peptides are depicted in FIG. 12. The predicted human SCPPPQ1 protein sequence corresponds to the putative sequence that was previously identified based on homology with the rat sequence, and the sequence human SCPPPQ1 protein truly expressed by human cells was identified and reported thereafter (Genbank accession No. MK322956.1). The first 56 amino-terminal residues of these sequences are identical, but the remaining C-terminal residues have no or very little sequence identity.
The effect of a two-hour incubation of various peptides depicted in FIG. 13 on the structural appearance of P. gingivalis was assessed by SEM. The results depicted in FIGs. 13A-B demonstrate a disruption of the membrane and some blebbing for the bacteria incubated with the peptides relative to the control (buffer). FIG. 13C shows a comparison of the effects of the peptides on the formation of aggregates using SEM (FIGs. 13A-B). Results of the counting show that the percentage of aggregates is more important for peptides #1 1 , #12, #13 and #14. The results depicted in FIGs. 13D-E show that all peptides except peptide #9 induce damages to the bacterial membrane relative to control (buffer).
These results suggest that the antibacterial activity of the SCPPPQ1 protein is at least partly conferred by the domain corresponding to peptide #5, and that the antibacterial activity of peptides derived from SCPPPQ1 can potentially exceed that of the full-length protein. The amino acid sequences (confirmed or predicted) of the SCPPPQ1 protein from various species are depicted in FIG. 14A, and the sequences of the domain corresponding to peptide #5 in the different SCPPPQ1 orthologs are shown in FIG. 14B.
TEM analyses also reveal disruption of the bacterial membranes and intracellular problems for the bacteria incubated with the peptides relative to the control (FIG. 15). Also, debris between the cells were only seen in the presence of the peptides.
Example 5: Assessment of the antimicrobial activity of SCPPPQ1 against other microorganisms
A series of experiments was performed to determine whether SCPPPQ1 exhibits inhibitory activity against microorganisms other than P. gingivalis.
The results depicted in FIG. 16 show that incubation of Fusobacterium nucleatum, a commensal oral bacterium that plays a role in periodontal disease, with rat SCPPPQ1 , reduces the interaction between the bacteria, induces damages at their surface, and decreases the bacterial population by about 75% relative to the control.
FIG. 17 shows that incubation of Prevotella intermedia, a gram-negative bacterium involved in periodontal infections (including gingivitis and periodontitis) with rat SCPPPQ1 decreases the bacterial population by about 70% relative to the control, and affects the interaction between the bacteria. No significant effect was observed following incubation with rat ODAM, his sister protein. Rat SCPPPQ1 was shown to have similar effects on Treponema denticola, another Gram-negative bacterium present in the oral cavity and associated with the incidence and severity of human periodontal disease (FIG. 18).
Rat and human SCPPPQ1 were also shown to induce defects on the bacterial membrane (presence of cellular debris and signs of membrane damages) of a Gram+ bacterium, Bacillus subtilis, that is found in the gastrointestinal tract of humans (FIGs. 19A-B). A similar effect was obtained following incubation of another Gram+ Bacillus, Bacillus thuringiensis (FIGs. 20A-B).
The results presented in FIGs. 21A-B show that rat SCPPPQ1 has an inhibitory effect on the growth of Escherichia coli, as evidenced by an important decrease in the number of bacteria in the presence of rat SCPPPQ1 relative to control (FIG. 21A). Rat and human SCPPPQ1 were also demonstrated to induce defects in the Gram- bacterial membrane of Escherichia coli, as shown in the SEM images of FIG. 21 B.
The effect of rat or human SCPPPQ1 on P. gingivalis was compared to that of kanamycin, which is known to kill the bacteria. The results presented in FIG. 22 show that kanamycin, similar to rat or human SCPPPQ1 , reduces the number of bacteria and induces the formation of aggregates, further validating the antibacterial activity of SCPPPQ1 .
The results presented above provide compelling evidence that the SCPPPQ1 protein and derived peptides have broad anti-bacterial activities (i.e. against various bacterial strains including Gram- and Gram+ bacteria). It was next assessed whether these molecules have inhibitory activities against other microorganisms such as yeasts (fungi). To do so, SCPPPQ1
(human and rat) as well as peptides #5, #8 and #10 (FIG. 12) were incubated with a representative opportunistic pathogenic yeast, Candida albicans. The results depicted in FIGs. 23A-B show that in the presence of rat SCPPPQ1 , human SCPPPQ1 and peptide #8, an important decrease of the number of hyphae was observed. In the presence of peptides #5 and #10, the number of hyphae did not decrease but were significantly reduced in size. It is well known that the ability of Candida albicans to grow as long filamentous hyphae is important for its pathogenic potential (Desai, J Fungi (Basel). 2018 Jan 1 1 ;4(1)). Thus, the reduction in the number and/or length of Candida albicans hyphae by the SCPPPQ1 proteins and peptides indicates that these molecules have ability to decrease the pathogenicity of Candida albicans.
FIG. 23C shows representative agar plates of five different Candida albicans strains incubated in the presence or not of rat SCPPPQ1 . All the strains are“hairy” (indicative of the presence of long hyphae) and in healthy conditions in the absence of SCPPPQ1 , but become “smooth” in the presence of 6pm of rat SCPPPQ1 in the agar, indicative of a reduction in the number of and/or length of hyphae. Furthermore, a notable decrease of unit forming colony (UFC) was also detected for strains BL152 and ATCC 36802.
The results presented above provide compelling evidence that the SCPPPQ1 protein and derived peptides have anti-bacterial and anti-fungal activities. It was next tested whether SCPPPQ1 could be toxic for normal cells. As shown in FIG. 25, no significant difference in cell number or appearance was observed following incubation of LS8 cells (ameloblast-like cell line) with rat SCPPPQ1 , confirming that SCPPPQ1 is not toxic for normal mammalian cells.
Example 6: Assessment of the antimicrobial activity of SCPPPQ1 added to toothpaste against P. gingivalis
It was next tested whether a toothpaste comprising SCPPPQ1 would have an inhibitory effect against P. gingivalis. The results depicted in FIG. 26 that the toothpaste comprising rat SCPPPQ1 reduces the bacterial population relative to the control (toothpaste + buffer). This result provides evidence that incorporating the peptides or proteins described herein into a toothpaste is a suitable approach to treat P. gingivalis infection and related disease.
Example 7: Assessment of the antimicrobial activity of SCPPPQ1 against P. gingivalis on titanium surface
The effect of the absorption of rat SCPPPQ1 or human SCPPPQ1 on titanium surfaces on Porphyromonas gingivalis was assessed by SEM. Absorbing the proteins to the surface led to a decrease of about 30% of the bacterial population (FIG. 27). It can also be noticed that the bacterial population in the control sample is uniformly dispersed, whereas the bacterial population in the samples with the SCPPPQ1 proteins shows a more patchy distribution with important spaces with no bacteria.
Although the present invention has been described hereinabove by way of specific embodiments thereof, it can be modified, without departing from the spirit and nature of the subject invention as defined in the appended claims. In the claims, the word "comprising" is used as an open-ended term, substantially equivalent to the phrase "including, but not limited to". The singular forms "a", "an" and "the" include corresponding plural references unless the context clearly dictates otherwise.
Claims
1 . An isolated polypeptide, peptide or peptidomimetic of 7 to 100 residues comprising a sequence of formula I:
X1 -X2-X3-X4-X5-X6-X7 -X8-X9-X10-X1 1 -X12-X13-X14-X15-X16-X17-X18-X19 (I) wherein
represents a bond;
XI is phenylalanine or an analog thereof, or is absent;
X2 is proline or an analog thereof;
X3 is leucine or an analog thereof, or is absent;
X4 is glutamine or an analog thereof, or is absent;
X5 is glutamine, proline, leucine or an analog thereof, or is absent;
X6 is proline or an analog thereof;
X7 is glutamine or an analog thereof, or is absent;
X8 is alanine or an analog thereof, or is absent;
X9 is proline or an analog thereof;
X10 is any amino acid or amino acid analog, or is absent;
XI I glutamine, glycine, or an analog thereof, or is absent;
X12 is leucine or an analog thereof, or is absent;
X13 is proline or an analog thereof, or is absent;
X14 is leucine or an analog thereof, or is absent;
X15 is proline or an analog thereof, or is absent;
X16 is isoleucine, leucine, or an analog thereof, or is absent;
X17 is proline, glutamine, or an analog thereof, or is absent;
X18 is isoleucine, leucine, phenylalanine, or an analog thereof, or is absent;
X19 is proline or an analog thereof, or is absent;
or a salt thereof.
2. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to claim 1 , wherein X1 is L-phenylalanine.
3. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to claim 1 or 2, wherein X2 is L-proline.
4. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of claims 1 to 3, wherein X3 is L-leucine.
5. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of claims 1 to 4, wherein X4 is absent.
6. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of claims 1 to 4, wherein X4 is L-glutamine.
7. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of claims 1 to 6, wherein X5 is absent.
8. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of claims 1 to 6, wherein X5 is L-glutamine, L-proline or L-leucine.
9. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of claims 1 to 8, wherein X6 is L-proline.
10. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of claims 1 to 9, wherein X7 is L-glutamine.
1 1 . The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of claims 1 to 10, wherein X8 is L-alanine.
12. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of claims 1 to 1 1 , wherein X9 is L-proline.
13. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of claims 1 to 12, wherein X10 is L-glutamine, L-leucine, L-serine, L-valine or L-proline, D-proline.
14. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of claims 1 to 13, wherein X1 1 , X12, X13, X14 and/or X15 are absent.
15. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of claims 1 to 13, wherein X1 1 , X12, X13, X14 and X15 are absent.
16. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of claims 1 to 13, wherein X1 1 is L-glutamine.
17. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of claims 1 to 14 and 16, wherein X12 is L-leucine.
18. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of claims 1 to 14, 16 and 17, wherein X13 is L-proline.
19. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of claims 1 to 14 and 16 to 18, wherein X14 is L-leucine.
20. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of claims 1 to 14 and 16 to 19, wherein X15 is L-proline.
21 . The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of claims 1 to 20, wherein X16 is L-isoleucine or L-leucine.
22. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of claims 1 to 20, wherein X16 is absent.
23. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of claims 1 to 22, wherein X17 is L-proline.
24. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of claims 1 to 23, wherein X18 is L-isoleucine, L-leucine or L-phenylalanine.
25. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of claims 1 to 24, wherein X18 is L-isoleucine, L-leucine or L-phenylalanine.
26. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of claims 1 to 25, wherein X19 is L-proline.
27. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of claims 1 to 26, wherein represents a peptide bond.
28. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of claims 1 to 27, which comprises a sequence of at least 7 or 8 residues from one of the following sequences: FPLPPQPP (SEQ ID NO:1), FPLPQAPLIPIP (SEQ ID NO:2), FPQPPQLQIP (SEQ ID NO:3), PLPPSPP (SEQ ID NO:4), FPFQQPPLIPFP (SEQ ID NO:5), FPPPVGLPLIPLP (SEQ ID NO:6), FPPPPQIPIP (SEQ ID NO:7), FPLPRQIPIP (SEQ ID NO:8), FPPPPQLPLIPIP (SEQ ID NO:9), FPLPQAPLIPIP (SEQ ID NO:10), or a variant thereof having at least 70% sequence identity with the at least 7 or 8 residues.
29. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to claim 28, which comprises a sequence of at least 7 or 8 residues from one of the following sequences: FPLPPQPP (SEQ ID NO: 1 ), FPLPQAPLIPIP (SEQ ID NO:2), FPQPPQLQIP (SEQ ID NO:3), PLPPSPP (SEQ ID NO:4), FPFQQPPLIPFP (SEQ ID NO:5), FPPPVGLPLIPLP (SEQ ID NO:6), FPPPPQIPIP (SEQ ID NO:7), FPLPRQIPIP (SEQ ID NO:8), FPPPPQLPLIPIP (SEQ ID NO:9), or FPLPQAPLIPIP (SEQ ID NO: 10).
30. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to claim 29, wherein the polypeptide, peptide or peptidomimetic comprises the sequence FPLPPQPP (SEQ ID NO: 1) or FPLPQAP (SEQ ID NO: 1 1), or a variant thereof having at least 70% sequence identity with these sequences.
31 . The isolated polypeptide, peptide, peptidomimetic or salt thereof according to claim 30, wherein the polypeptide, peptide or peptidomimetic comprises the sequence FPLPPQPP (SEQ ID NO: 1 ) or FPLPQAP (SEQ ID NO:1 1 ).
32. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of claims 1 to 31 , which comprises at least 10 contiguous amino acids from the amino acid sequences of any one of SEQ ID NOs: 12-29 and 62.
33. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to claim 32, comprises an amino acid sequence having at least 70% sequence identity with the sequences of any one of SEQ ID NOs: 12-29 and 62.
34. The isolated polypeptide, peptide, peptidomimetic or salt thereof according to claim 33, comprises with the amino acid sequences of any one of SEQ ID NOs: 12-29 and 62.
35. An isolated nucleic acid comprising a nucleotide sequence encoding the polypeptide or peptide of any one of claims 1 to 34.
36. A vector comprising the nucleic acid of claim 35.
37. A host cell comprising nucleic acid of claim 35 or the vector of claim 36.
38. A composition comprising the isolated polypeptide, peptide, peptidomimetic or salt thereof according to any one of claims 1 to 34, and one or more excipients.
39. The composition of claim 38, wherein the one or more excipients comprise a biodegradable polymer.
40. The composition of claim 38 or 39, which is in the form of a cream, a gel, a paste, a solution, a chewing gum, a patch, a film, or a strip.
41 . A method for treating a bacterial or fungal infection in a subject comprising administering to the subject an effective amount of the polypeptide, peptide, peptidomimetic or salt thereof according to any one of claims 1 to 34, or the composition of any one of claims 38 to 40.
42. The method of claim 41 , wherein the fungal infection is a yeast infection.
43. The method of claim 42, wherein the yeast is of the Candida genus.
44. The method of claim 43, wherein the yeast is of the Candida albicans species.
45. The method of claim 41 , wherein the bacterial infection is by Gram- bacteria.
46. The method of claim 45, wherein the Gram- bacteria are of the Porphyromonas, Prevotella, Fusobacterium, Escherichia and/or Treponema genus.
47. The method of claim 46, wherein the Gram- bacteria are of the Porphyromonas gingivalis, Fusobacterium nucleatum, Prevotella intermedia, Escherichia coli and/or Treponema denticola species.
48. The method of claim 41 , wherein the bacterial infection is by Gram+ bacteria.
G17200-00006
48
49. The method of claim 48, wherein the Gram+ bacteria are of the Bacillus genus.
50. The method of claim 49, wherein the Gram+ bacteria are of the Bacillus subtilis species.
51 . A method for treating a periodontal or gingival disease in a subject in need thereof, said method comprising contacting the gum from the subject with an effective amount of the polypeptide, peptide, peptidomimetic or salt thereof according to any one of claims 1 to 34, or the composition of any one of claims 38 to 40.
52. The method of claim 51 , wherein the composition is the composition of claim 40.
53. The method of claim 51 or 52, wherein the periodontal or gingival disease is peri- implantitis, gingivitis, periodontitis or peri-implant mucositis.
54. A method for cleaning or disinfecting a surface or an object comprising contacting the surface or the object with an effective amount of the polypeptide, peptide, peptidomimetic or salt thereof according to any one of claims 1 to 34, or the composition of any one of claims 38 to 40.
55. The method of claim 54, wherein the object is a dental or orthopedic implant.
56. The polypeptide, peptide, peptidomimetic or salt thereof according to any one of claims 1 to 34, or the composition of any one of claims 38 to 40 for treating a bacterial or fungal infection in a subject.
57. The polypeptide, peptide, peptidomimetic, salt or composition for use according to claim
56, wherein the fungal infection is a yeast infection.
58. The polypeptide, peptide, peptidomimetic, salt or composition for use according to claim
57, wherein the yeast is of the Candida genus.
59. The polypeptide, peptide, peptidomimetic, salt or composition for use according to claim
58, wherein the yeast is of the Candida albicans species.
60. The polypeptide, peptide, peptidomimetic, salt or composition for use according to claim 56, wherein the bacterial infection is by Gram- bacteria.
61 . The polypeptide, peptide, peptidomimetic, salt or composition for use according to claim
60, wherein the Gram- bacteria are of the Porphyromonas, Prevotella, Fusobacterium, Escherichia and/or Treponema genus.
62. The polypeptide, peptide, peptidomimetic, salt or composition for use according to claim
61 , wherein the Gram- bacteria are of the Porphyromonas gingivalis, Fusobacterium nucleatum, Prevotella intermedia, Escherichia coli and/or Treponema denticola species.
63. The polypeptide, peptide, peptidomimetic, salt or composition for use according to claim 56, wherein the bacterial infection is by Gram+ bacteria.
G17200-00006
49
64. The polypeptide, peptide, peptidomimetic, salt or composition for use according to claim
63, wherein the Gram+ bacteria are of the Bacillus genus.
65. The polypeptide, peptide, peptidomimetic, salt or composition for use according to claim
64, wherein the Gram+ bacteria are of the Bacillus subtilis species.
66. The polypeptide, peptide, peptidomimetic or salt thereof according to any one of claims 1 to 34, or the composition of any one of claims 38 to 40 for treating a periodontal or gingival disease in a subject.
67. The polypeptide, peptide, peptidomimetic, salt or composition for use according to claim
66. wherein the composition is the composition of claim 40.
68. The polypeptide, peptide, peptidomimetic, salt or composition for use according to claim
66 or 67, wherein the periodontal or gingival disease is peri-implantitis, gingivitis, periodontitis or peri-implant mucositis.
69. The polypeptide, peptide, peptidomimetic or salt thereof according to any one of claims 1 to 34, or the composition of any one of claims 38 to 40 for cleaning or disinfecting a surface or an object.
70. The polypeptide, peptide, peptidomimetic, salt or composition for use according to claim 69, wherein the object is a dental or orthopedic implant.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US201962829732P | 2019-04-05 | 2019-04-05 | |
US62/829,732 | 2019-04-05 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2020198834A1 true WO2020198834A1 (en) | 2020-10-08 |
Family
ID=72667551
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/CA2019/051839 WO2020198834A1 (en) | 2019-04-05 | 2019-12-18 | Treatment of microbial infections using scpppq1 proteins and derived peptides |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2020198834A1 (en) |
Citations (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2012122640A1 (en) * | 2011-03-17 | 2012-09-20 | Université de Montréal | Compositions, diagnostic methods using epithelial extracellular matrix proteins |
-
2019
- 2019-12-18 WO PCT/CA2019/051839 patent/WO2020198834A1/en active Application Filing
Patent Citations (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2012122640A1 (en) * | 2011-03-17 | 2012-09-20 | Université de Montréal | Compositions, diagnostic methods using epithelial extracellular matrix proteins |
Non-Patent Citations (2)
Title |
---|
DATABASE Nucleotide 6 February 2019 (2019-02-06), "Homo sapiens secretory calcium-binding phosphoprotein proline-glutamine rich (SCPPPQ1) mRNA, complete cds", XP055746750, retrieved from ncbi Database accession no. MK 322956.1 * |
FOUILLEN ET AL.: "Selective bacterial degradation of the extracellular matrix attaching the gingiva to the tooth", EUR. J. ORAL SCI., vol. 127, 22 June 2019 (2019-06-22), pages 313 - 322, XP055746757 * |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
AU2018202201B2 (en) | Peptides and their use | |
JP5335241B2 (en) | Antimicrobial hexapeptide | |
CN1925866B (en) | Treatment of bacterial infections | |
JP6683601B2 (en) | Antimicrobial peptide | |
EP2116603A2 (en) | Antimicrobial peptides | |
WO1997018827A1 (en) | Compositions and methods for the prevention and treatment of oral mucositis | |
US6025326A (en) | Compositions and methods for the prevention and treatment of oral mucositis | |
US11945846B2 (en) | Stapled intracellular-targeting antimicrobial peptides to treat infection | |
US10435437B2 (en) | Modified apidaecin derivatives as antibiotic peptides | |
US20200115426A1 (en) | Cationic intrinsically disordered antimicrobial peptides | |
CN110582507A (en) | Engineered antimicrobial amphiphilic peptides and methods of use | |
EP3809851A1 (en) | Lysins and derivatives thereof resensitize staphylococcus aureus and gram-positive bacteria to antibiotics | |
JP7076497B2 (en) | Cyclic antimicrobial pseudopeptide and its use | |
WO2020198834A1 (en) | Treatment of microbial infections using scpppq1 proteins and derived peptides | |
US20190375791A1 (en) | New d-configured cateslytin peptide | |
EP2197899B1 (en) | Cyclic peptides comprising at least one aza- beta3-aminoacyl residue and uses thereof | |
AU704851B2 (en) | Compositions and methods for the prevention and treatment of oral mucositis | |
CA3192533A1 (en) | Composition and use | |
De Zoysa | Synthesis and Structure-Activity Relationship Studies of Battacin Peptides as Potential Therapeutic Agents Against Bacterial Pathogens | |
MXPA98004079A (en) | Compositions and methods for the prevention and treatment of mucositis or |
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: 19922647 Country of ref document: EP Kind code of ref document: A1 |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
122 | Ep: pct application non-entry in european phase |
Ref document number: 19922647 Country of ref document: EP Kind code of ref document: A1 |