WO2017010845A1 - Composition for preventing or treating staphylococcal infectious diseases - Google Patents

Composition for preventing or treating staphylococcal infectious diseases Download PDF

Info

Publication number
WO2017010845A1
WO2017010845A1 PCT/KR2016/007748 KR2016007748W WO2017010845A1 WO 2017010845 A1 WO2017010845 A1 WO 2017010845A1 KR 2016007748 W KR2016007748 W KR 2016007748W WO 2017010845 A1 WO2017010845 A1 WO 2017010845A1
Authority
WO
WIPO (PCT)
Prior art keywords
composition
pgn
wta
treatment
prevention
Prior art date
Application number
PCT/KR2016/007748
Other languages
French (fr)
Korean (ko)
Inventor
이복률
안동호
Original Assignee
재단법인 목암생명과학연구소
부산대학교 산학협력단
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 재단법인 목암생명과학연구소, 부산대학교 산학협력단 filed Critical 재단법인 목암생명과학연구소
Publication of WO2017010845A1 publication Critical patent/WO2017010845A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/14Peptides containing saccharide radicals; Derivatives thereof, e.g. bleomycin, phleomycin, muramylpeptides or vancomycin
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • C07K14/305Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Micrococcaceae (F)
    • C07K14/31Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Micrococcaceae (F) from Staphylococcus (G)

Definitions

  • the present invention relates to a pharmaceutical composition for the prevention or treatment of Staphylococcal infection disease.
  • Staphylococcus aureus is a human skin, soft tissue (soft tissue) and can lead to severe bloodstream infection (Lowy FD, The New England Journal of Medicine, 339: 520-532, 1998).
  • Staphylococcus aureus may be transformed into methicillin-resistant strain (methsaillin-resistant S. aureus , MRSA) resistant to beta-lactam family of methicillin. MRSA infections are difficult to treat and have a poor prognosis, which is a social problem.
  • CA-MRSA community-associated MRSA strains
  • H-MRSA hospital-associated MRSA strains
  • USA300 MRSA strain which is spreading in the United States, causes serious diseases in children or those with reduced immune function. Therefore, new vaccines or treatments having a prophylactic and therapeutic effect on MRSA infection are required.
  • the cell walls of Gram-positive bacteria are four components, including peptidoglycan (PGN), wall teichoic acid (WTA), lipoteicoic acid (LTA), and capsular polysaccharides (CP) It consists of.
  • PPN peptidoglycan
  • WTA wall teichoic acid
  • LTA lipoteicoic acid
  • CP capsular polysaccharides
  • the cell wall component of staphylococci is a glycopolymer, and its structure is very complicated, and it is difficult to separate and purify each as a single substance.
  • various cell wall components are exposed to the outside, which makes it difficult to identify which component acts as a ligand in the host's immune system.
  • WTA-PGN wall teichoic acid-attached peptidoglycan
  • Another object of the present invention to provide a method for preventing or treating staphylococcal infection disease using the composition.
  • Still another object of the present invention is to provide a method for preparing soluble dumpeichoic acid-attached peptidoglycan (WTA-PGN) which can be used as an active ingredient in the composition.
  • WTA-PGN soluble dumpeichoic acid-attached peptidoglycan
  • the present invention is a composition for the prevention or treatment of staphylococcal infection disease, which contains the wall teichoic acid-attached peptidoglycan (WTA-PGN) represented by the general formula 1 as an active ingredient to provide:
  • WTA-PGN wall teichoic acid-attached peptidoglycan
  • n is an integer of 10 to 50
  • m is an integer of 1 to 3
  • A is N-acetylmannosamine (ManNAc)
  • B is N-acetylglucosamine (GlcNAc)
  • O is An integer from 1 to 5
  • R 1 and R 2 are each independently hydroxy, tetrapeptide or pentapeptide
  • R 3 is hydroxy or N-acetylmuramic acid (MurNAc).
  • the present invention also provides a method for preventing or treating Staphylococcal infection disease, comprising administering the composition to a subject in need thereof.
  • the present invention provides: 1) to obtain a double-mutant strain is lgt (lipoprotein diacylglycerol transferase) and oatA (O-acetyl transferase) gene deletions from the wild type Staphylococcus aureus; 2) heating the double mutant strain to a temperature of 40 to 70 ° C; 3) adding ⁇ -lytic enzyme to the heated strain; 4) obtaining a soluble WTA-PGN containing fraction from the enzyme additive of step 3); 5) adding lysozyme or mutanolysin to the soluble WTA-PGN containing fractions; And 6) obtaining a soluble WTA-PGN from the enzyme additive of step 5).
  • lgt lipoprotein diacylglycerol transferase
  • oatA O-acetyl transferase
  • Soluble WTA-PGN represented by Formula 1 according to the present invention increases the production of IL-17A in the mouse body when administered to mice, the soluble WTA-PGN can be usefully used for the prevention and treatment of staphylococci have.
  • FIG. 1 is a graph showing the elution pattern of S. aureus T384 strain ( ⁇ lgt ⁇ oatA ) prepared in one embodiment of the present invention after treatment with ⁇ -lytic enzyme and HPLC separation.
  • Figure 2 is a graph showing the elution pattern separated by HPLC treatment of lysozyme to each fraction obtained by treatment with insoluble ⁇ -lytic enzyme to the heat-treated S. aureus T384 strain and separated by HPLC:
  • F An elution pattern separated by HPLC after treating lysozyme to the F fraction.
  • Figure 3 shows the results of the production of IL-17A after injection into the mouse each peak fraction obtained after the enzyme treatment:
  • FIG. 4 is a graph showing an elution pattern of Sephacryl-B-TSK-B fractions separated by a C18 reversed phase column.
  • FIG. 5 is a graph comparing the amount of IL-17A induced when mice were injected with each of the six fractions in which Sephacryl-B-TSK-B fractions were separated by a C18 reversed phase column (Con: Sephacryl-B-TSK).
  • -B fraction A: A fraction
  • B B fraction
  • C C fraction
  • D D fraction
  • E E fraction
  • F F fraction
  • FIG. 6 is a graph showing an elution pattern in which the C to E fractions separated from the C18 reverse phase column in Example 5 were re-separated into a C18 reverse phase column.
  • Figure 7 is a graph comparing the production of IL-17A induced when mice were injected with C-E fractions re-separated in a C18 reversed phase column (C: C fraction, D: D fraction, E: E fraction).
  • wall teichoic acid is one of the cell wall components of S. aureus , combined with a glycerol phosphate repeat unit and a ribitol phosphate repeat unit (N-acetyl refers to a glycopolymer consisting of mannosamine)-( ⁇ -1,3)-(N-acetylglucosamine).
  • peptidoglycan refers to a repeat glycopolymer of N-acetylmuriamic acid (MurNAc) and N-acetylglucosamine (GlcNAc) linked by stem-peptide bonds.
  • wall teichoic acid-attached peptidoglycan refers to a structure in which the wall teichoic acid and peptidoglycan are covalently bonded.
  • the present invention provides a composition for the prevention or treatment of Staphylococcal infection disease, which comprises aste active acid-attached peptidoglycan (WTA-PGN) represented by the following general formula (1):
  • n is an integer of 10 to 50
  • m is an integer of 1 to 3
  • A is N-acetylmannosamine (ManNAc)
  • B is N-acetylglucosamine (GlcNAc)
  • O is An integer from 1 to 5
  • R 1 and R 2 are each independently hydroxy, tetrapeptide or pentapeptide
  • R 3 is hydroxy or N-acetylmuramic acid (MurNAc).
  • n is an integer from 35 to 45
  • m is 3
  • A is ManNAc
  • B is GlcNAc
  • O is an integer from 1 to 5
  • R 1 and Each R 2 is independently hydroxy, tetrapeptide or pentapeptide and R 3 is hydroxy or MurNAc.
  • n 40, m is 3, A is ManNAc, B is GlcNAc, O is an integer from 1 to 5, R 1 and R 2 is Each independently is a tetrapeptide or pentapeptide and R 3 is hydroxy or MurNAc.
  • a and B may be linked to a ⁇ -position to each other.
  • the tetrapeptide is -X 1 -X 2 -X 3 -X 4 (SEQ ID NO: 1)
  • the pentapeptide is (SEQ ID NO: 2).
  • X 1 may be Ala or Gly
  • X 2 may be Glu or Asp
  • X 3 may be Lys, Arg, or His
  • X 4 may be Ala or Gly.
  • the tetrapeptide is-(L-Ala)-(D-Glu)-(L-Lys)-(D-Ala) (SEQ ID NO: 3), and the pentapeptide is (SEQ ID NO: 4).
  • the PGN structure of Formula 1 may be represented by the following formula (1).
  • the R 1 or R 2 of the WTA-PGN may form the R cross-linking with each other and any one of 1 or R 2 of other WTA-PGN,
  • the WTA-PGN along the two WTA-PGN may also be present in the form of a dimer combined with it.
  • the staphylococcus may be methicillin-resistant Staphylococcus aureus (MRSA), methicillin-sensitive Staphylococcus aureus (MSSA) or pathogenic staphylococcus.
  • MRSA methicillin-resistant Staphylococcus aureus
  • MSSA methicillin-sensitive Staphylococcus aureus
  • infectious diseases caused by Staphylococcus aureus include soft tissue infection, purulent arthritis, purulent osteomyelitis, otitis media, pneumonia, sepsis, acute respiratory tract infection, infection due to the use of a catheter, postoperative wound infection, bacteremia, Endocarditis and food poisoning.
  • composition according to the present invention may further comprise a pharmaceutically acceptable carrier, diluent and adjuvant in addition to the WTA-PGN.
  • Carriers used in the compositions according to the invention are selected based on the method and route of administration, and standard drug compositions, and include, for example, carrier proteins (ie, bovine serum albumin (BSA), egg white albumin (OVA), human serum albumin). (HSA) and keyhole limpet hemocyanin (KLH)), solubilizers (ie ethanol, polysorbate and Cremophor EL TM), isotonic agents, preservatives, antioxidants, excipients (ie lactose, starch, crystalline cellulose, mannitol) , Maltose, calcium hydrogen phosphate, light anhydrous silicic acid and calcium carbonate), binder (ie starch, polyvinylpyrrolidone, hydroxypropyl cellulose, ethyl cellulose, carboxymethyl cellulose and gum arabic), lubricant (ie magnesium stearate) , Talc, and cured oils, and the like) and stabilizers (ie, lactos
  • composition according to the present invention can be combined with a known KLH solution (Calbiotec, dissolving 125 mg per ml of 50% glycerol solution) as a carrier protein to enhance antigenicity.
  • KLH solution Calbiotec, dissolving 125 mg per ml of 50% glycerol solution
  • Diluents used in the compositions according to the invention can be selected based on the method and route of administration and the actual standard drug composition.
  • examples of diluents include water, physiological saline, phosphate buffered physiological saline, and bicarbonate solutions.
  • Adjuvants used in the compositions according to the invention may be selected based on the method and route of administration and the actual standard drug composition.
  • adjuvant include cholera toxin, E. coli dipyrotoxin (LT), liposomes and immune stimulatory complex (ISCOM).
  • the present invention also provides a method for preventing or treating Staphylococcal infection disease, comprising administering the composition to a subject in need thereof.
  • the subject may be a mammal, specifically a human.
  • the method can prevent or treat staphylococcal infections by simultaneously inducing opsonophagocytosis and phagocytosis in a subject.
  • the method may also increase the amount of IL-17A produced within 24 hours after administration of the composition according to the invention in a subject.
  • the administration may vary depending on the age, weight, sex and general state of health of the subject. Routes for such administration include oral and parenteral administration (eg, intravenous administration, arterial administration and topical administration), preferably parenteral administration.
  • parenteral administration eg, intravenous administration, arterial administration and topical administration
  • Formulations for oral and parenteral administration and methods for their preparation are known to those skilled in the art.
  • Formulations for oral and parenteral administration may be prepared by conventional procedures, eg, by admixing with the aforementioned pharmaceutically acceptable carriers.
  • Examples of formulations for oral administration include solid or liquid formulations such as solvents, tablets, granules, powders or capsules.
  • Examples of formulations for parenteral administration include solvents, suspensions, ointments, creams, suppositories, eye drops, nasal drops and ear drops.
  • biodegradable polymers eg, poly-D, L-lactide-co-glycoside or polyglycoside
  • buck base e.g, US patents
  • 5,417,986, 4,675,381 and 4,450,150 e.g., US patents
  • flavors and colorings may be added.
  • Suitable pharmaceutical carriers, diluents and pharmaceutically necessary materials for their use are described in Remington's Pharmaceutical Sciences.
  • the dosage of the composition according to the present invention is determined based on the type of adjuvant, the method and frequency of administration, and the desired effect and may generally be from 1 ⁇ g to 100 mg of WTA-PGN in a single adult dose.
  • the dosage may generally be from 1 ⁇ g to 1 mg of WTA-PGN in a single adult dose.
  • the administration can be administered several times if necessary.
  • the composition may be administered again by supplementing three times after the composition has been initially administered at regular intervals.
  • the compositions for the first and second reinforcements may be administered 8-12 weeks and 16-20 weeks after the first administration using the same formulation, respectively.
  • the present invention provides: 1) to obtain a double-mutant strain is lgt (lipoprotein diacylglycerol transferase) and oatA (O-acetyl transferase) gene deletions from the wild type Staphylococcus aureus; 2) heating the double mutant strain to a temperature of 40 to 70 ° C; 3) adding ⁇ -lytic enzyme to the heated strain; 4) obtaining a soluble WTA-PGN containing fraction from the enzyme additive of step 3); 5) adding lysozyme or mutanolysin to the soluble WTA-PGN containing fractions; And 6) obtaining a soluble WTA-PGN from the enzyme additive of step 5).
  • lgt lipoprotein diacylglycerol transferase
  • oatA O-acetyl transferase
  • Step 1) there is provided a method comprising obtaining a double-mutant strains and oatA lgt gene deletion from the wild-type staphylococci.
  • Double mutant strain ( ⁇ lgt ⁇ oatA obtained in the above step 1)) whereby the lgt gene deletion not likely to be contaminated by the lipoprotein can be obtained pure WTA-PGN.
  • the deletion of the oatA gene eliminates the acetyl group in the MurNAc residue of the PGN, and the resulting WTA-PGN can be easily degraded by the ⁇ -lytic enzyme of step 2).
  • Such double mutant strains can be obtained by known methods from wild type staphylococci, for example methicillin-resistant staphylococci (MRSA), methicillin-sensitive staphylococci (MSSA) or pathogenic staphylococci.
  • MRSA methicillin-resistant staphylococci
  • MSSA methicillin-sensitive staphylococci
  • the double mutant strain T363 strain of the lgt gene while having a resistance to play Oh, my God (phleomycin) defect (Nakayama M et al, Journal of Immunology 189:. 5903-591, 2012) and resistant to erythromycin T0003 strain (Park KH et al. , Journal of Biological Chemistry 285, 27167-27175, 2010) with oatA gene deficient can be prepared by transduction through phage 80.
  • Step 2) provides a step of heating the double mutant strain to a temperature of 40 to 70 °C.
  • the temperature may be 45 to 68 °C, 50 to 65 °C or 55 to 63 °C, in one embodiment of the present invention the temperature may be 60 °C.
  • step 2) may comprise the step of culturing the double mutant strain obtained in step 1), followed by heat treatment.
  • Step 3) provides a step of treating ⁇ -lytic enzyme on the heated strain.
  • the ⁇ -lytic enzyme plays a role in degrading pentaglycine ((Gly) 5) linkages linking the stem peptides present at the MurNAc residue of the insoluble WTA-PGN obtained in step 2), thereby insoluble WTA contained in the heated strain. Change PGN to soluble WTA-PGN.
  • Such ⁇ -lytic enzymes are commercially available or described in Li et al. , Journal of Biochemitry 122, 772-778, 1997, can be isolated and purified.
  • An example of a commercially available ⁇ -lytic enzyme is lysostaphin.
  • Step 3) may be carried out by suspending the strain heated in step 2) in a buffer, and then adding ⁇ -lytic enzyme and reacting with stirring at 30 to 40 ° C. for 10 to 14 hours.
  • Step 4) provides a step of obtaining a soluble WTA-PGN containing fraction from the enzyme additive of step 3).
  • This step can be carried out by passing the ⁇ -lytic enzyme additive through HPLC to obtain a fraction, and then selecting the fraction containing soluble WTA-PGN in the fraction. At this time, the fraction containing WTA-PGN can be confirmed by PAGE or silver nitrate staining method.
  • the column used in the HPLC purification may be HiTrap Q FF (GE Healthcare), which is an anion exchange resin that binds with the anion of WTA ribitol phosphate.
  • HiTrap Q FF GE Healthcare
  • Step 5 provides the step of adding lysozyme or mutanolysine to the soluble WTA-PGN containing fractions.
  • the lysozyme or mutanolysine transforms the polymeric PGN into an oligomeric PGN by breaking down the bond between MurNAc and GlcNAc of the PGN.
  • Step 5) may be carried out by suspending the soluble WTA-PGN obtained in step 4) in a buffer, and then adding lysozyme or mutanolysine for 10 to 14 hours with stirring at 30 to 40 ° C.
  • Step 6) provides a step of obtaining soluble WTA-PGN from the enzyme additive of step 5).
  • the step can be carried out by passing lysozyme or mutanolysine enzyme additive through HPLC to obtain a fraction, and then selecting the fraction containing soluble WTA-PGN from the fraction. At this time, the fraction containing WTA-PGN can be confirmed by PAGE or silver nitrate staining method.
  • the column used for HPLC purification may be HiTrap-Q (GE Healthcare).
  • the method according to the invention may further comprise further purifying the WTA-PGN after step 6).
  • Said further purification can be carried out by gel filtration chromatography or reverse phase liquid chromatography.
  • the soluble WTA-PGN prepared in step 6) can be further purified via gel filtration chromatography using Sephacryl S-200 HR column or reverse phase liquid chromatography using Symmetry Shield TM RP18 column. have.
  • the soluble WTA-PGN prepared in step 6) is further purified by gel filtration chromatography using Sephacryl S-200 HR column and reverse phase liquid chromatography using two Symmetry Shield TM RP18 columns. Can be.
  • the active fraction can be selected based on the amount of IL-17A produced after injection of each fraction obtained in the chromatography into the mouse abdominal cavity.
  • the strain can be used to isolate WTA, WTA-PGN and PGN without contamination by lipoproteins by deletion of the lgt gene.
  • the PGN MurNAc residue 6 position oxygen does not have an acetyl group, so that the isolated PGN can be easily degraded by lysozyme.
  • the stored strain was centrifuged at 6,000 rpm at 4 ° C. for 30 minutes to obtain pellets.
  • 20 ml of saline was added to resuspend the pellet, which was centrifuged for 5 minutes at 3,000 ⁇ g, 4 ° C. to obtain pellet.
  • the flask was stirred every 5 minutes during the heat treatment process so that the strain was uniformly heat treated.
  • the diluted solution of the strain was cooled to 30 ° C., which was centrifuged at 3,000 ⁇ g, 20 ° C. for 10 minutes to obtain pellets.
  • the pellet was suspended in 10 ml saline and centrifuged for 5 minutes under the same conditions as above to obtain the pellet.
  • the obtained pellet was washed three times with sterile distilled water, and the pellet was suspended in 3 ml of distilled water and lyophilized.
  • the concentrated sample was subjected to size exclusion chromatography on a Sephacryl S-100 column (1.6 cm x 87 cm) using 10 mM sodium citrate buffer (pH 6.0) containing 200 mM NaCl.
  • the eluate was measured for absorbance at 280 nm, and the fractions showing lytic activity among the fractions with high absorbance were collected and concentrated again.
  • Li et al. Based on the results of (1997), ⁇ -lytic enzyme fractions were selected from the concentrated fractions.
  • the selected fractions were subjected to size exclusion chromatography on a Superdex-75 column (1 cm ⁇ 30 cm) using 10 mM sodium citrate buffer (pH 6.0) containing 200 mM NaCl. As a result, ⁇ -lytic enzyme was finally obtained.
  • the obtained ⁇ -lytic enzyme was incubated with PGN suspension derived from Micrococcus luteus (ATCC 9341) or insoluble staphylococcus, and the lytic activity or lytic activity of the enzyme was confirmed.
  • enzymes obtained by Procise® Protein sequencer Cat. # 491-0, Applied Biosystems, USA
  • electrophoresis were identified.
  • the obtained enzyme was a ⁇ -lytic enzyme by confirming that the N-terminal sequence was S-P-N-G-L-L-Q-F-P-F (SEQ ID NO: 5) and the size was about 20 kDa.
  • the supernatant obtained was filtered with a filter having a pore size of 0.45 ⁇ m and the filtrate was HPLC (805 MANOMETRIC MODULE, 811C) equipped with a HiTrap Q FF column equilibrated with 20 mM Tris-HCl buffer (pH 7.0) (buffer A). DYNAMIC MIXER, 305 PUMP, 306 PUMP, 151 UV / VIS Detector, Gilson, USA). Thereafter, 20 mM Tris-HCl buffer (pH 7.0) (buffer B) containing 1 M NaCl was eluted by performing a linear gradient from 0 M to 1 M NaCl concentration. The concentration gradient of the buffer B was performed for 100 minutes in a gradient up to 80%, 1 minute in a gradient up to 100%, and then eluted at 100% for 10 minutes.
  • each fraction was named A, B, C, D, E and F, and each of these fractions was precipitated with acetone and then lyophilized.
  • Each lyophilized A to F fraction was suspended in 20 mM Tris-HCl buffer (pH 7.0). 12.5 ⁇ g of lysozyme (Cat. # 62970, Sigma-Aldrich Co. LLC., USA) was added to 1 mg of the lyophilized fraction and reacted with stirring at 180 rpm and 37 ° C. for 12 hours. The subsequent procedure was performed under the same conditions and methods as in Example ⁇ 3-2>, except that the HiTrap Q column was used.
  • Each of the peaks identified above were named as AL-A for the A peak obtained in the A fraction, and the same as the BL-A for the A peak obtained in the B fraction, and AL-A, BL-A, CL-B, and DL.
  • the -B, DL-C, EL-C and FL-B peaks were respectively precipitated with acetone and then lyophilized.
  • the mixed peaks (EL-C + FL-B) in which each of the CL-B, DL-B and DL-C peaks and the EL-C and FL-B peaks were mixed were prepared by lyophilization, respectively. It was. Meanwhile, C57BL / 6J female mice weighing 15 ⁇ 0.5 g at 5 weeks of age were purchased from Korea Research Institute of Bioscience and Biomedical Research Center (Ochang Campus, Korea) and were commercially available solid feeds (Cat. # 5L79, Orient Bio, Inc.). Korea) was adapted to the environment (20-25 ° C., humidity 55%) for one week while feeding. Six to twelve animals per group were divided into six groups per group in a completely randomized design to provide diet and drinking water by ad libitum. Body weight and dietary intake of each mouse were measured once daily and the lights were turned on and off at 12 hour intervals.
  • the amount of IL-17A is ELISA Ready-SET-Go!
  • the kit (Cat. # 88-7371-88, eBioscience, USA) was used to measure according to the manufacturer's protocol. As a result, a value obtained by correcting the absorbance value measured at 450 nm to the absorbance value measured at 550 nm was determined as the amount of final IL-17A.
  • Example ⁇ 3-3> The lyophilized fractions in Example ⁇ 3-3> were further purified using various columns.
  • the solvent was allowed to flow until the UV detector was stabilized under conditions of a flow rate of 0.3 ml / min, a sensitivity of 2, a peak width of 10.0 sec, and a UV absorbance of 220 nm so that there was no error due to impurity inflow during the experiment.
  • the injector was converted to the loaded state to inject the suspended peak fractions.
  • the eluent was obtained by converting the injector into the injection state before elution, and the elution conditions were the same as the equilibration conditions.
  • the obtained eluate was named 'Sepacryl-B fraction' and stored by lyophilization.
  • the Sephacryl-B fraction was further purified using a TSK G2000SW column. Specifically, 3.5 mg of the lyophilized Sephacryl-B fraction was suspended in 5 mM sodium phosphate buffer (pH 6.0) containing 100 mM NaCl to prepare a sample. On the other hand, the column was prepared by washing and equilibrating with the buffer. Equilibration was performed until the UV detector stabilized under conditions of flow rate 3 ml / min, sensitivity 0.5, peak width 10 seconds and UV absorbance 202 nm. After equilibration, the inlet was changed to a rod state to slowly inject the sample, and when the elution started, the inlet was changed to infusion to obtain an eluate. In this case, 5 mM sodium phosphate buffer (pH 6.0) containing 100 mM NaCl was used as the elution buffer.
  • 5 mM sodium phosphate buffer (pH 6.0) containing 100 mM NaCl was used as the e
  • the B fraction which most significantly increased the production of IL-17A was named 'Sepacryl-B-TSK-B fraction'. Lyophilized and stored.
  • a C18 reversed phase column was used to further purify the lyophilized Sephacryl-B-TSK-B fraction.
  • Example 5 Each fraction obtained in Example 5 was used to confirm IL-17A inducing activity in mice under the same conditions and methods as in Example 4, and the results are shown in FIG. 5.
  • Example 5 Using a fraction of 2 mg of the lyophilized C to E peak obtained in Example 5, a C18 reversed phase column was used in the same conditions and methods as in Example 5 to purify it more purely.
  • Example 7 WTA was first removed to confirm the structure of Fraction C, which showed a production increase activity of IL-17A.
  • lyophilized C fraction was dissolved in 50 ⁇ l of sterile distilled water. 50 ⁇ l of 25% TFA was added thereto and incubated with stirring at a temperature of 37 ° C. for 12 hours. After incubation, 100 ⁇ l of sterile distilled water was added to prepare a final 6.25% concentration of TFA. Thereafter, the Symmetry Shield TM RP18 column was used in the same conditions and methods as in Example 5 to purify only PGN free of WTA. At this time, the flow rate was carried out under the conditions of 1 mL / min, a sensitivity of 0.5, a column temperature of 40 ° C., and a UV absorbance of 203 nm. The concentration gradient of the mobile phase for elution was performed for 10 minutes at 0%, 50 minutes at gradient to 43.8%, 2 minutes at gradient to 100%, and then eluted at 100% for 10 minutes.
  • Example ⁇ 8-1> The eluate obtained in Example ⁇ 8-1> was loaded onto the column via an autoinjector in a Nano-flow (n) LC-MS / MS system.
  • nLC-MS / MS systems are high resolution hybrid masses with nLC (EASY-nLC 1000, Thermo Fisher Scientific, USA) with fritless electrospray (ESI) columns (150 ⁇ m ⁇ 100 mm, Nikkyo Technos Co. Ltd, Japan) It consists of a spectrometer (Q-Exactive mass spectrometer, Thermo Fisher Scientific, USA).
  • nLC separation was eluted using a 32% (v / v) acetonitrile solution containing 0.1% (v / v) formic acid as elution buffer for 30 minutes at a flow rate of 300 nL / min.
  • the eluate was sprayed online with a mass spectrometer at a pressure of 2.0 kPa in the bipolar state, and the analyzer was operated in data-dependent mode to automatically switch between MS and MS / MS acquisition.
  • a mass resolution of 35,000 was obtained at m / z 400 through full-scan mass spectra (m / z 300 to 2,000) measurements.
  • PGN having a structure represented by the following formula 1 was confirmed. It had five monosaccharide chains represented by GlcNAc-MurNAc-GlcNAc-MurNAc-GlcNAc and had one phosphate group bound to the fourth MurNAc residue.
  • the PGN of the C fraction had a molecular weight of 2167.91.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Organic Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Engineering & Computer Science (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The present invention relates to a pharmaceutical composition for preventing or treating staphylococcal infectious diseases. Specifically, the soluble WTA-PGN represented by general formula 1 according to the present invention increases the production of IL-17A in the body of mice when administered to the mice, and thus, the soluble WTA-PGN can be favorably used in the prevention and treatment of staphylococcus.

Description

포도상구균 감염 질환의 예방 또는 치료용 조성물Composition for preventing or treating staphylococcal infection disease
본 발명은 포도상구균 감염 질환의 예방 또는 치료용 약학 조성물에 관한 것이다.The present invention relates to a pharmaceutical composition for the prevention or treatment of Staphylococcal infection disease.
포도상구균(Staphylococcus aureus)은 인간의 피부, 연조직(soft tissue) 및 혈류에서 중증 감염을 야기할 수 있다(Lowy FD, The New England Journal of Medicine, 339:520-532, 1998). 또한, 포도상구균은 여러 가지 원인에 의하여 베타-락탐 계열의 항생제인 메티실린에 대해 내성을 갖는 메티실린-내성 균주(methicillin-resistant S. aureus, MRSA)로 변형될 수 있다. 이러한 MRSA 감염은 치료가 어렵고 예후가 좋지 않아 사회적으로 큰 문제가 되고 있다. 특히, 병원에 입원한 적이 없는 어린이 등에게 지역사회 관련 MRSA 균주(community-associated MRSA, CA-MRSA)가 출현하여 종래의 병원 관련 MRSA 균주(hospital-associated MRSA, HA-MRSA)와 함께 그 치료의 어려움을 더욱 가중시키고 있다. 최근 미국에서 확산되고 있는 USA300 MRSA 균주는 어린이나 면역 기능이 저하된 사람들에게 심각한 질환을 유발하고 있는바, MRSA 감염에 대해 예방 및 치료 효과를 갖는 새로운 백신이나 치료법의 개발이 요구된다.Staph (Staphylococcus aureus) is a human skin, soft tissue (soft tissue) and can lead to severe bloodstream infection (Lowy FD, The New England Journal of Medicine, 339: 520-532, 1998). In addition, Staphylococcus aureus may be transformed into methicillin-resistant strain (methsaillin-resistant S. aureus , MRSA) resistant to beta-lactam family of methicillin. MRSA infections are difficult to treat and have a poor prognosis, which is a social problem. In particular, community-associated MRSA strains (CA-MRSA) have appeared in children who have never been admitted to the hospital, and the treatment of the treatment with conventional hospital-associated MRSA strains (HA-MRSA) It is adding to the difficulties. Recently, the USA300 MRSA strain, which is spreading in the United States, causes serious diseases in children or those with reduced immune function. Therefore, new vaccines or treatments having a prophylactic and therapeutic effect on MRSA infection are required.
종래 국외 연구진들이 포도상구균 백신 후보 물질들로 개발하려던 물질들은 전임상 실험에서 좋은 예후를 보였음에도 불구하고, 임상 단계에서 모두 실패하였기에, 최근까지도 임상적으로 유용하게 사용될 수 있는 포도상구균 백신은 개발되지 못하고 있는 실정이다.Previously, researchers who have tried to develop staphylococcal vaccine candidates have failed in clinical stages despite the good prognosis in preclinical experiments. Until recently, no clinically useful staphylococcus vaccines have been developed. There is a situation.
최근 연구들에 따르면, 포도상구균 백신으로서 기능하기 위해서는 체액성 면역(humoral immunity)과 세포성 면역(cellular immunity)이 모두 필요하고, T 세포 활성화에 의한 식균세포 작용기능(phagocytic cell effector function)을 촉진시켜야 한다는 사실이 제안되었다. 이때, 체액성 면역은 백신 물질에 대해 특이적인 혈청 항체를 생성함으로써 옵소닌식균작용(opsonophagocytosis)을 촉진하고, 세포성 면역은 MRSA 감염 초기 단계에 T 세포-매개 IL-17A를 생성하여 호중구 동원 및 식균작용(phagocytosis)을 촉진한다.Recent studies show that both humoral and cellular immunity are required to function as a staphylococcal vaccine and promote phagocytic cell effector function by T cell activation. It has been suggested that it should be done. At this time, humoral immunity promotes opsonophagocytosis by generating serum antibodies specific for the vaccine substance, and cellular immunity produces T cell-mediated IL-17A in the early stages of MRSA infection, leading to neutrophil recruitment and Promotes phagocytosis.
또한, 국외 연구진에 의해, 포도상구균이 감염된 마우스 모델에서 기억 γδ-T 세포의 수가 증가하여 면역 반응이 나타난다는 연구결과가 발표되었다(J. Immunol., 192(8):3697-708, 2014). 이 연구결과는, γδ-T 세포의 기억 반응이 포도상구균에 노출시 αβ-T 세포와 유사한 방식으로 유도되고, γδ-T 세포에 의해 생산되는 IL-17A가 강력한 포도상구균 치료 효과가 있음을 시사한다.In addition, foreign researchers reported that immune responses occur due to an increase in the number of memory γδ-T cells in a mouse model infected with Staphylococcus aureus ( J. Immunol. , 192 (8): 3697-708, 2014). . The findings suggest that the memory response of γδ-T cells is induced in a manner similar to αβ-T cells upon exposure to Staphylococcus, and that IL-17A produced by γδ-T cells has a potent staphylococcal treatment effect. do.
그람 음성 박테리아와 달리, 포도상구균과 같은 그람 양성 박테리아의 세포벽은 펩티도글리칸(PGN), 벽테이코산(WTA), 리포테이코산(LTA) 및 협막 다당류(CP)를 포함하는 4종의 성분으로 구성된다. 이들 세포벽 성분을 표적으로 하여 박테리아 감염을 예방 또는 치료하기 위한 물질을 개발할 수 있으나, 숙주의 면역계에서 인식하는 박테리아의 리간드 물질이 명확히 규명되지 않아, 이에 어려움을 겪고 있다.Unlike Gram-negative bacteria, the cell walls of Gram-positive bacteria, such as staphylococci, are four components, including peptidoglycan (PGN), wall teichoic acid (WTA), lipoteicoic acid (LTA), and capsular polysaccharides (CP) It consists of. Targeting these cell wall components can be developed to prevent or treat bacterial infections, but the ligand material of bacteria recognized by the host's immune system has not been clearly identified, which is a problem.
특히, 포도상구균의 세포벽 성분은 당고분자(glycopolymer)로서, 그 구조가 매우 복잡하고, 각각을 단일 물질로서 분리 및 정제하기 어렵다. 또한, 여러 종류의 세포벽 성분이 외부에 노출되어 있어 어떤 성분이 숙주의 면역계에서 리간드로 작용하는지 규명하기 어렵다.In particular, the cell wall component of staphylococci is a glycopolymer, and its structure is very complicated, and it is difficult to separate and purify each as a single substance. In addition, various cell wall components are exposed to the outside, which makes it difficult to identify which component acts as a ligand in the host's immune system.
이에, 본 발명자들은 포도상구균의 세포벽 성분 중 어떤 성분이 숙주의 면역계에서 리간드로 작용하는지에 대해 예의연구한 결과, 특정 구조식을 갖는 벽테이코산-부착된 펩티도글리칸(WTA-PGN)을 분리하고, 이러한 WTA-PGN이 포도상구균 감염 질환의 예방 또는 치료에 유용하다는 것을 확인함으로써 본 발명을 완성하였다.Therefore, the present inventors have diligently studied which component of the cell wall component of Staphylococcus aureus acts as a ligand in the host's immune system, and thus isolates the wall teichoic acid-attached peptidoglycan (WTA-PGN) having a specific structural formula. The present invention was completed by confirming that WTA-PGN is useful for preventing or treating Staphylococcal infection disease.
따라서, 본 발명의 목적은 포도상구균 감염 질환의 예방 또는 치료용 조성물을 제공하는 것이다.Accordingly, it is an object of the present invention to provide a composition for the prevention or treatment of Staphylococcal infection disease.
본 발명의 다른 목적은 상기 조성물을 이용하여 포도상구균 감염 질환의 예방 또는 치료방법을 제공하는 것이다.Another object of the present invention to provide a method for preventing or treating staphylococcal infection disease using the composition.
본 발명의 또 다른 목적은 상기 조성물에서 유효성분으로 사용될 수 있는 가용성 벽테이코산-부착된 펩티도글리칸(WTA-PGN)을 제조하는 방법을 제공하는 것이다.Still another object of the present invention is to provide a method for preparing soluble dumpeichoic acid-attached peptidoglycan (WTA-PGN) which can be used as an active ingredient in the composition.
상기 목적을 달성하기 위하여, 본 발명은 하기 일반식 1로 표시되는 벽테이코산-부착된 펩티도글리칸(WTA-PGN)을 유효성분으로 함유하는, 포도상구균 감염 질환의 예방 또는 치료용 조성물을 제공한다:In order to achieve the above object, the present invention is a composition for the prevention or treatment of staphylococcal infection disease, which contains the wall teichoic acid-attached peptidoglycan (WTA-PGN) represented by the general formula 1 as an active ingredient to provide:
[일반식 1][Formula 1]
Figure PCTKR2016007748-appb-I000001
Figure PCTKR2016007748-appb-I000001
상기 일반식 1에서, n은 10 내지 50의 정수이고, m은 1 내지 3의 정수이며, A는 N-아세틸만노사민(ManNAc)이고, B는 N-아세틸글루코사민(GlcNAc)이며, O는 1 내지 5의 정수이고, R1 및 R2는 각각 독립적으로 하이드록시, 테트라펩타이드 또는 펜타펩타이드이며, R3은 하이드록시 또는 N-아세틸뮤람산(MurNAc)이다.In Formula 1, n is an integer of 10 to 50, m is an integer of 1 to 3, A is N-acetylmannosamine (ManNAc), B is N-acetylglucosamine (GlcNAc), O is An integer from 1 to 5, R 1 and R 2 are each independently hydroxy, tetrapeptide or pentapeptide, and R 3 is hydroxy or N-acetylmuramic acid (MurNAc).
또한, 본 발명은 상기 조성물을 이를 필요로 하는 개체에 투여하는 것을 포함하는, 포도상구균 감염 질환의 예방 또는 치료 방법을 제공한다.The present invention also provides a method for preventing or treating Staphylococcal infection disease, comprising administering the composition to a subject in need thereof.
나아가, 본 발명은 1) 야생형 포도상구균으로부터 lgt(lipoprotein diacylglycerol transferase) 및 oatA(O-acetyl transferase) 유전자가 결실된 이중 돌연변이 균주를 수득하는 단계; 2) 상기 이중 돌연변이 균주를 40 내지 70℃의 온도로 가열하는 단계; 3) 상기 가열된 균주에 β-lytic 효소를 첨가하는 단계; 4) 상기 단계 3)의 효소 첨가물로부터 가용성 WTA-PGN 함유 분획을 수득하는 단계; 5) 상기 가용성 WTA-PGN 함유 분획에 라이소자임 또는 뮤타노라이신(mutanolysin)을 첨가하는 단계; 및 6) 상기 단계 5)의 효소 첨가물로부터 가용성 WTA-PGN을 수득하는 단계를 포함하는, 일반식 1로 표시되는 WTA-PGN을 제조하는 방법을 제공한다.Furthermore, the present invention provides: 1) to obtain a double-mutant strain is lgt (lipoprotein diacylglycerol transferase) and oatA (O-acetyl transferase) gene deletions from the wild type Staphylococcus aureus; 2) heating the double mutant strain to a temperature of 40 to 70 ° C; 3) adding β-lytic enzyme to the heated strain; 4) obtaining a soluble WTA-PGN containing fraction from the enzyme additive of step 3); 5) adding lysozyme or mutanolysin to the soluble WTA-PGN containing fractions; And 6) obtaining a soluble WTA-PGN from the enzyme additive of step 5).
본 발명에 따른 일반식 1로 표시되는 가용성 WTA-PGN은 이를 마우스에 투여하였을 때 마우스 체내의 IL-17A의 생산량을 증가시키므로, 상기 가용성 WTA-PGN은 포도상구균의 예방 및 치료에 유용하게 사용될 수 있다.Soluble WTA-PGN represented by Formula 1 according to the present invention increases the production of IL-17A in the mouse body when administered to mice, the soluble WTA-PGN can be usefully used for the prevention and treatment of staphylococci have.
도 1은 본 발명의 일실시예에서 제조된 S. aureus T384 균주(ΔlgtΔoatA)를 열처리한 뒤, β-lytic 효소를 처리하고 HPLC로 분리한 용출패턴을 나타낸 그래프이다.1 is a graph showing the elution pattern of S. aureus T384 strain ( ΔlgtΔoatA ) prepared in one embodiment of the present invention after treatment with β-lytic enzyme and HPLC separation.
도 2는 열처리한 S. aureus T384 균주에 불용성 β-lytic 효소를 처리하고 HPLC로 분리하여 얻은 각각의 분획에 라이소자임을 처리하고 HPLC로 분리한 용출패턴을 나타낸 그래프이다:Figure 2 is a graph showing the elution pattern separated by HPLC treatment of lysozyme to each fraction obtained by treatment with insoluble β-lytic enzyme to the heat-treated S. aureus T384 strain and separated by HPLC:
A: A 분획에 라이소자임을 처리한 뒤 HPLC로 분리한 용출패턴;A: an elution pattern separated by HPLC after treating lysozyme to the A fraction;
B: B 분획에 라이소자임을 처리한 뒤 HPLC로 분리한 용출패턴;B: an elution pattern separated by HPLC after treating lysozyme to the B fraction;
C: C 분획에 라이소자임을 처리한 뒤 HPLC로 분리한 용출패턴;C: an elution pattern separated by HPLC after treating lysozyme to the C fraction;
D: D 분획에 라이소자임을 처리한 뒤 HPLC로 분리한 용출패턴;D: an elution pattern separated by HPLC after treatment with lysozyme in the D fraction;
E: E 분획에 라이소자임을 처리한 뒤 HPLC로 분리한 용출패턴; 및E: an elution pattern separated by HPLC after treatment of lysozyme to the E fraction; And
F: F 분획에 라이소자임을 처리한 뒤 HPLC로 분리한 용출패턴.F: An elution pattern separated by HPLC after treating lysozyme to the F fraction.
도 3은 효소처리 후 수득한 각 피크 분획물을 마우스에 주사한 뒤, IL-17A의 생산량을 확인한 결과이다:Figure 3 shows the results of the production of IL-17A after injection into the mouse each peak fraction obtained after the enzyme treatment:
1: HPLC로 분리한 C 분획에 라이소자임을 처리하고 HPLC로 분리한 B 피크;1: B peak treated with lysozyme and separated by HPLC with C fraction separated by HPLC;
2: HPLC로 분리한 D 분획에 라이소자임을 처리하고 HPLC로 분리한 B 피크;2: B peak treated by lysozyme and separated by HPLC for the D fraction separated by HPLC;
3: HPLC로 분리한 D 분획에 라이소자임을 처리하고 HPLC로 분리한 C 피크; 및3: C peak treated by lysozyme with HPLC fractionated and separated by HPLC; And
4: HPLC로 분리한 E 및 F 분획 각각에 라이소자임을 처리하고 HPLC로 분리한 E 분획의 C 피크 및 F 분획의 B 피크의 혼합물.4: A mixture of the C peak of the E fraction and the B peak of the F fraction, each of which was treated with lysozyme and separated by HPLC for each of the E and F fractions separated by HPLC.
도 4는 세파크릴-B-TSK-B 분획을 C18 역상 컬럼으로 분리한 용출 패턴을 나타내는 그래프이다.4 is a graph showing an elution pattern of Sephacryl-B-TSK-B fractions separated by a C18 reversed phase column.
도 5는 세파크릴-B-TSK-B 분획을 C18 역상 컬럼으로 분리한 6개의 각 분획을 마우스에 주사하였을 때 유도되는 IL-17A의 생산량을 비교한 그래프이다(Con: 세파크릴-B-TSK-B 분획, A: A 분획, B: B 분획, C: C 분획, D: D 분획, E: E 분획, F: F 분획).FIG. 5 is a graph comparing the amount of IL-17A induced when mice were injected with each of the six fractions in which Sephacryl-B-TSK-B fractions were separated by a C18 reversed phase column (Con: Sephacryl-B-TSK). -B fraction, A: A fraction, B: B fraction, C: C fraction, D: D fraction, E: E fraction, F: F fraction).
도 6은 실시예 5에서 C18 역상 컬럼으로 분리한 C 내지 E 분획을 C18 역상 컬럼으로 재분리한 용출 패턴을 나타내는 그래프이다.FIG. 6 is a graph showing an elution pattern in which the C to E fractions separated from the C18 reverse phase column in Example 5 were re-separated into a C18 reverse phase column.
도 7은 C18 역상 컬럼에서 재분리된 C 내지 E 분획을 마우스에 주사하였을 때 유도되는 IL-17A의 생산량을 비교한 그래프이다(C: C 분획, D: D 분획, E: E 분획).Figure 7 is a graph comparing the production of IL-17A induced when mice were injected with C-E fractions re-separated in a C18 reversed phase column (C: C fraction, D: D fraction, E: E fraction).
이하, 본 발명에 사용된 용어를 정의한다.Hereinafter, terms used in the present invention are defined.
본 명세서에서 사용된 용어, "벽테이코산(wall teichoic acid, WTA)"은 포도상구균(S. aureus)의 세포벽 성분 중 하나로서, 글리세롤 포스페이트 반복 단위 및 리비톨 포스페이트 반복 단위와 함께 (N-아세틸만노사민)-(β-1,3)-(N-아세틸글루코사민)으로 구성된 당중합체를 지칭한다. As used herein, the term "wall teichoic acid (WTA)" is one of the cell wall components of S. aureus , combined with a glycerol phosphate repeat unit and a ribitol phosphate repeat unit (N-acetyl Refers to a glycopolymer consisting of mannosamine)-(β-1,3)-(N-acetylglucosamine).
본 명세서에서 사용된 용어, "펩티도글리칸(peptidoglycan, PGN)"은 스템 펩타이드간 결합에 의해 연결된 N-아세틸뮤람산(MurNAc)과 N-아세틸글루코사민(GlcNAc)의 반복 당중합체를 지칭한다.As used herein, the term "peptidoglycan (PGN)" refers to a repeat glycopolymer of N-acetylmuriamic acid (MurNAc) and N-acetylglucosamine (GlcNAc) linked by stem-peptide bonds.
본 명세서에서 사용된 용어, "벽테이코산-부착된 펩티도글리칸(WTA-PGN)"은 벽테이코산과 펩티도글리칸이 공유결합된 구조를 지칭한다.As used herein, the term "wall teichoic acid-attached peptidoglycan (WTA-PGN)" refers to a structure in which the wall teichoic acid and peptidoglycan are covalently bonded.
이하, 본 발명을 상세히 설명한다.Hereinafter, the present invention will be described in detail.
본 발명은 하기 일반식 1로 표시되는 벽테이코산-부착된 펩티도글리칸(WTA-PGN)을 유효성분으로 함유하는, 포도상구균 감염 질환의 예방 또는 치료용 조성물을 제공한다:The present invention provides a composition for the prevention or treatment of Staphylococcal infection disease, which comprises aste active acid-attached peptidoglycan (WTA-PGN) represented by the following general formula (1):
[일반식 1][Formula 1]
Figure PCTKR2016007748-appb-I000002
Figure PCTKR2016007748-appb-I000002
상기 일반식 1에서, n은 10 내지 50의 정수이고, m은 1 내지 3의 정수이며, A는 N-아세틸만노사민(ManNAc)이고, B는 N-아세틸글루코사민(GlcNAc)이며, O는 1 내지 5의 정수이고, R1 및 R2는 각각 독립적으로 하이드록시, 테트라펩타이드 또는 펜타펩타이드이며, R3은 하이드록시 또는 N-아세틸뮤람산(MurNAc)이다.In Formula 1, n is an integer of 10 to 50, m is an integer of 1 to 3, A is N-acetylmannosamine (ManNAc), B is N-acetylglucosamine (GlcNAc), O is An integer from 1 to 5, R 1 and R 2 are each independently hydroxy, tetrapeptide or pentapeptide, and R 3 is hydroxy or N-acetylmuramic acid (MurNAc).
본 발명의 다른 실시예에서, 상기 일반식 1에서, n은 35 내지 45의 정수이고, m은 3이며, A는 ManNAc이고, B는 GlcNAc이며, O는 1 내지 5의 정수이고, R1 및 R2는 각각 독립적으로 하이드록시, 테트라펩타이드 또는 펜타펩타이드이며, R3은 하이드록시 또는 MurNAc이다.In another embodiment of the present invention, in formula 1, n is an integer from 35 to 45, m is 3, A is ManNAc, B is GlcNAc, O is an integer from 1 to 5, R 1 and Each R 2 is independently hydroxy, tetrapeptide or pentapeptide and R 3 is hydroxy or MurNAc.
본 발명의 또 다른 실시예에서, 상기 일반식 1에서, n은 40이고, m은 3이며, A는 ManNAc이고, B는 GlcNAc이며, O는 1 내지 5의 정수이고, R1 및 R2는 각각 독립적으로 테트라펩타이드 또는 펜타펩타이드이며, R3은 하이드록시 또는 MurNAc이다.In another embodiment of the present invention, in formula 1, n is 40, m is 3, A is ManNAc, B is GlcNAc, O is an integer from 1 to 5, R 1 and R 2 is Each independently is a tetrapeptide or pentapeptide and R 3 is hydroxy or MurNAc.
상기 일반식 1에서 A 및 B는 서로 β-위치로 연결될 수 있다. 또한, 상기 일반식 1에서 테트라펩타이드는 -X1-X2-X3-X4(서열번호 1)이고, 펜타펩타이드는
Figure PCTKR2016007748-appb-I000003
(서열번호 2)이다. 여기서, X1은 Ala 또는 Gly이며, X2는 Glu 또는 Asp이고, X3는 Lys, Arg 또는 His이며, X4는 Ala 또는 Gly일 수 있다. 본 발명의 일실시예에서, 상기 테트라펩타이드는 -(L-Ala)-(D-Glu)-(L-Lys)-(D-Ala)(서열번호 3)이고, 상기 펜타펩타이드는
Figure PCTKR2016007748-appb-I000004
(서열번호 4)일 수 있다.
In Formula 1, A and B may be linked to a β-position to each other. In addition, in the general formula 1, the tetrapeptide is -X 1 -X 2 -X 3 -X 4 (SEQ ID NO: 1), and the pentapeptide is
Figure PCTKR2016007748-appb-I000003
(SEQ ID NO: 2). Here, X 1 may be Ala or Gly, X 2 may be Glu or Asp, X 3 may be Lys, Arg, or His, and X 4 may be Ala or Gly. In one embodiment of the present invention, the tetrapeptide is-(L-Ala)-(D-Glu)-(L-Lys)-(D-Ala) (SEQ ID NO: 3), and the pentapeptide is
Figure PCTKR2016007748-appb-I000004
(SEQ ID NO: 4).
본 발명의 일실시예에서, 상기 일반식 1의 PGN 구조는 하기 식 1로 표시될 수 있다.In one embodiment of the present invention, the PGN structure of Formula 1 may be represented by the following formula (1).
[식 1][Equation 1]
Figure PCTKR2016007748-appb-I000005
Figure PCTKR2016007748-appb-I000005
본 발명의 다른 실시예에서, 상기 WTA-PGN의 R1 또는 R2는 다른 WTA-PGN의 R1 또는 R2 중 어느 하나와 서로 교차결합을 형성할 수 있으며, 이에 따라 WTA-PGN은 2개의 WTA-PGN이 결합된 다이머(dimer) 형태로도 존재할 수 있다.In a further embodiment of the invention, the R 1 or R 2 of the WTA-PGN may form the R cross-linking with each other and any one of 1 or R 2 of other WTA-PGN, The WTA-PGN along the two WTA-PGN may also be present in the form of a dimer combined with it.
상기 포도상구균은 메티실린-내성 포도상구균(methicillin-resistant Staphylococcus aureus, MRSA), 메티실린-민감성 포도상구균(methicillin-sensitive Staphylococcus aureus, MSSA) 또는 병원성 포도상구균일 수 있다. 또한, 상기 포도상구균에 의한 감염 질환은 연부조직 감염, 화농성 관절염, 화농성 골수염, 중이염, 폐렴, 패혈증, 급성 호흡기 감염(acute respiratory tract infection), 카테터의 사용으로 인한 감염, 수술 후 창상 감염, 균혈증, 심내막염 및 식중독으로 구성된 군으로부터 선택될 수 있다.The staphylococcus may be methicillin-resistant Staphylococcus aureus (MRSA), methicillin-sensitive Staphylococcus aureus (MSSA) or pathogenic staphylococcus. In addition, the infectious diseases caused by Staphylococcus aureus include soft tissue infection, purulent arthritis, purulent osteomyelitis, otitis media, pneumonia, sepsis, acute respiratory tract infection, infection due to the use of a catheter, postoperative wound infection, bacteremia, Endocarditis and food poisoning.
일례로, 본 발명에 따른 조성물은 상기 WTA-PGN 외에 약학적으로 허용가능한 담체, 희석제 및 보강제를 추가로 포함할 수 있다. In one example, the composition according to the present invention may further comprise a pharmaceutically acceptable carrier, diluent and adjuvant in addition to the WTA-PGN.
본 발명에 따른 조성물에 사용되는 담체는 투여 방법 및 경로, 그리고 표준 약물 조성물을 기초로 선택되고, 예를 들면, 담체 단백질(즉, 소혈청 알부민(BSA), 난백 알부민(OVA), 인간혈청 알부민(HSA) 및 키홀 림펫 헤모시아닌(KLH)), 용해제(즉, 에탄올, 폴리솔베이트 및 Cremophor EL™), 등장화제, 보존제, 항산화제, 부형제(즉, 락토스, 전분, 결정성 셀룰로오스, 만니톨, 말토스, 인산 수소 칼슘, 경무수 규산 및 탄산칼슘), 결합제(즉, 전분, 폴리비닐피롤리돈, 히드록시프로필셀룰로오스, 에틸셀룰로오스, 카복시메틸셀룰로오스 및 아라비아검), 윤활제(즉, 스테아린산 마그네슘, 탈크, 및 경화유 등) 및 안정화제(즉, 락토스, 만니톨, 말토스, 폴리솔베이트, 마크로졸, 폴리옥시에틸렌 경화 피마자유)일 수 있다. 필요에 따라, 글리세린, 디메틸아세트아미드, 70% 젖산 나트륨, 계면활성제 또는 염기성 물질 (즉, 수산화나트륨, 에틸렌디아민, 에탄올 아민, 중탄산나트륨, 알기닌, 메글루민 또는 트리스아미노메탄) 등을 포함할 수 있다. 구체적으로, 본 발명에 따른 조성물은 항원성을 강화하기 위하여, 담체 단백질로서 공지의 KLH 용액(Calbiotec, 50% 글리세롤 용액 1 ㎖ 당 125 ㎎을 용해시킴)과 결합될 수 있다.Carriers used in the compositions according to the invention are selected based on the method and route of administration, and standard drug compositions, and include, for example, carrier proteins (ie, bovine serum albumin (BSA), egg white albumin (OVA), human serum albumin). (HSA) and keyhole limpet hemocyanin (KLH)), solubilizers (ie ethanol, polysorbate and Cremophor EL ™), isotonic agents, preservatives, antioxidants, excipients (ie lactose, starch, crystalline cellulose, mannitol) , Maltose, calcium hydrogen phosphate, light anhydrous silicic acid and calcium carbonate), binder (ie starch, polyvinylpyrrolidone, hydroxypropyl cellulose, ethyl cellulose, carboxymethyl cellulose and gum arabic), lubricant (ie magnesium stearate) , Talc, and cured oils, and the like) and stabilizers (ie, lactose, mannitol, maltose, polysorbate, macrosol, polyoxyethylene cured castor oil). If desired, glycerin, dimethylacetamide, 70% sodium lactate, surfactants or basic substances (ie, sodium hydroxide, ethylenediamine, ethanol amine, sodium bicarbonate, arginine, meglumine or trisaminomethane) and the like can be included. have. Specifically, the composition according to the present invention can be combined with a known KLH solution (Calbiotec, dissolving 125 mg per ml of 50% glycerol solution) as a carrier protein to enhance antigenicity.
본 발명에 따른 조성물에 사용되는 희석제는 투여 방법 및 경로, 그리고 실제 표준 약물 조성물을 기초로 하여 선택될 수 있다. 희석제의 예로는 물, 생리 식염수, 인산염 완충 생리 식염수 및 중탄산염 용액 등이 있다.Diluents used in the compositions according to the invention can be selected based on the method and route of administration and the actual standard drug composition. Examples of diluents include water, physiological saline, phosphate buffered physiological saline, and bicarbonate solutions.
본 발명에 따른 조성물에 사용되는 보강제는 투여 방법 및 경로, 그리고 실제 표준 약물 조성물을 기초로 선택될 수 있다. 보강제의 예로는 콜레라 독소, 대장균의 이열성 장독소(LT), 리포좀 및 면역 자극성 복합체(ISCOM) 등이 있다.Adjuvants used in the compositions according to the invention may be selected based on the method and route of administration and the actual standard drug composition. Examples of adjuvant include cholera toxin, E. coli dipyrotoxin (LT), liposomes and immune stimulatory complex (ISCOM).
또한, 본 발명은 상기 조성물을 이를 필요로 하는 개체에 투여하는 것을 포함하는, 포도상구균 감염 질환의 예방 또는 치료 방법을 제공한다.The present invention also provides a method for preventing or treating Staphylococcal infection disease, comprising administering the composition to a subject in need thereof.
상기 개체는 포유동물, 구체적으로 인간일 수 있다.The subject may be a mammal, specifically a human.
상기 방법은 개체로부터 옵소닌식균작용(opsonophagocytosis) 및 식균작용(phagocytosis)을 동시에 유도함으로써, 포도상구균 감염질환을 예방 또는 치료할 수 있다. 또한, 상기 방법은 개체 내에서 본 발명에 따른 조성물의 투여 후 24시간 이내에 IL-17A 생성량을 증가시킬 수 있다.The method can prevent or treat staphylococcal infections by simultaneously inducing opsonophagocytosis and phagocytosis in a subject. The method may also increase the amount of IL-17A produced within 24 hours after administration of the composition according to the invention in a subject.
상기 투여는 투여대상의 연령, 체중, 성별 및 일반적인 건강상태에 따라 다를 수 있다. 상기 투여를 위한 경로는 경구투여 및 비경구 투여(예를 들면, 정맥 투여, 동맥 투여 및 국소 투여)가 있으며, 바람직하게는 비경구 투여일 수 있다.The administration may vary depending on the age, weight, sex and general state of health of the subject. Routes for such administration include oral and parenteral administration (eg, intravenous administration, arterial administration and topical administration), preferably parenteral administration.
경구 투여 및 비경구 투여용 제형 및 그것의 제조방법은 당업자에게 공지되어 있다. 경구 투여 및 비경구 투여용 제형은 통상적인 공정, 예를 들면, 앞서 언급한 약학적으로 허용가능한 담체와 혼합하여 제조될 수 있다. 경구 투여용 제형의 예로는 용제, 정제, 과립제, 가루약 또는 캡슐제와 같은 고체 또는 액체 제형을 포함한다. 비경구 투여용 제형의 예로는 용제, 현탁제, 연고제, 크림, 좌제, 안제, 점비제 및 점이제를 포함한다. 본 제제의 서방정 제조를 위해, 생물 분해성 폴리머(예를 들면, 폴리-D,L-락티드-코-그리코시드 또는 폴리글리코시드)를 버크 기제에 첨가할 수 있다(예를 들면, 미국특허 제5,417,986호, 제4,675,381호 및 제4,450,150호 참조). 경구 투여의 경우, 풍미제 및 착색료가 첨가될 수 있다. 적절한 약제학적 담체, 희석제와 그것의 사용을 위해 약제학적으로 필요한 물질은 문헌[Remington's Pharmaceutical Sciences]에 기재되어 있다.Formulations for oral and parenteral administration and methods for their preparation are known to those skilled in the art. Formulations for oral and parenteral administration may be prepared by conventional procedures, eg, by admixing with the aforementioned pharmaceutically acceptable carriers. Examples of formulations for oral administration include solid or liquid formulations such as solvents, tablets, granules, powders or capsules. Examples of formulations for parenteral administration include solvents, suspensions, ointments, creams, suppositories, eye drops, nasal drops and ear drops. For the preparation of sustained-release tablets of the present formulations, biodegradable polymers (eg, poly-D, L-lactide-co-glycoside or polyglycoside) can be added to the buck base (eg, US patents). 5,417,986, 4,675,381 and 4,450,150). For oral administration, flavors and colorings may be added. Suitable pharmaceutical carriers, diluents and pharmaceutically necessary materials for their use are described in Remington's Pharmaceutical Sciences.
본 발명에 따른 조성물의 투여량은 보강제의 종류, 투여 방법 및 빈도, 및 원하는 효과를 기초로 결정되며 일반적으로 성인 1회 투여시 WTA-PGN 1 ㎍ 내지 100 ㎎일 수 있다. 본 발명의 조성물에 보강제가 첨가되는 경우, 투여량은 일반적으로 성인 1회 투여시 WTA-PGN 1 ㎍ 내지 1 ㎎일 수 있다. 상기 투여는 필요한 경우 수회 투여될 수 있다. 예를 들면, 일정 간격으로 초기에 조성물이 투여된 후 3회 보강하여 조성물이 다시 투여될 수 있다. 선택적으로, 첫 번째 및 두 번째 보강을 위한 조성물은 각각 동일한 제제를 사용하여 최초 투여 후 8 내지 12번째 주 및 16 내지 20번째 주에 투여될 수 있다.The dosage of the composition according to the present invention is determined based on the type of adjuvant, the method and frequency of administration, and the desired effect and may generally be from 1 μg to 100 mg of WTA-PGN in a single adult dose. When adjuvant is added to the composition of the present invention, the dosage may generally be from 1 μg to 1 mg of WTA-PGN in a single adult dose. The administration can be administered several times if necessary. For example, the composition may be administered again by supplementing three times after the composition has been initially administered at regular intervals. Optionally, the compositions for the first and second reinforcements may be administered 8-12 weeks and 16-20 weeks after the first administration using the same formulation, respectively.
나아가, 본 발명은 1) 야생형 포도상구균으로부터 lgt(lipoprotein diacylglycerol transferase) 및 oatA(O-acetyl transferase) 유전자가 결실된 이중 돌연변이 균주를 수득하는 단계; 2) 상기 이중 돌연변이 균주를 40 내지 70℃의 온도로 가열하는 단계; 3) 상기 가열된 균주에 β-lytic 효소를 첨가하는 단계; 4) 상기 단계 3)의 효소 첨가물로부터 가용성 WTA-PGN 함유 분획을 수득하는 단계; 5) 상기 가용성 WTA-PGN 함유 분획에 라이소자임 또는 뮤타노라이신(mutanolysin)을 첨가하는 단계; 및 6) 상기 단계 5)의 효소 첨가물로부터 가용성 WTA-PGN을 수득하는 단계를 포함하는, 일반식 1로 표시되는 WTA-PGN을 제조하는 방법을 제공한다.Furthermore, the present invention provides: 1) to obtain a double-mutant strain is lgt (lipoprotein diacylglycerol transferase) and oatA (O-acetyl transferase) gene deletions from the wild type Staphylococcus aureus; 2) heating the double mutant strain to a temperature of 40 to 70 ° C; 3) adding β-lytic enzyme to the heated strain; 4) obtaining a soluble WTA-PGN containing fraction from the enzyme additive of step 3); 5) adding lysozyme or mutanolysin to the soluble WTA-PGN containing fractions; And 6) obtaining a soluble WTA-PGN from the enzyme additive of step 5).
이하, 본 발명에 따른 일반식 1로 표시되는 WTA-PGN을 제조하는 방법을 상세히 설명한다. Hereinafter, a method of manufacturing WTA-PGN represented by the general formula 1 according to the present invention will be described in detail.
상기 단계 1)은 야생형 포도상구균으로부터 lgtoatA 유전자가 결실된 이중 돌연변이 균주를 수득하는 단계를 제공한다. Step 1) there is provided a method comprising obtaining a double-mutant strains and oatA lgt gene deletion from the wild-type staphylococci.
상기 단계 1)에서 수득되는 이중 돌연변이 균주(ΔlgtΔoatA)는 lgt 유전자가 결실됨으로써 지질단백에 의해 오염될 가능성이 없어 순수한 WTA-PGN을 수득할 수 있다. 또한, oatA 유전자가 결실됨으로써 PGN의 MurNAc 잔기에 아세틸기가 없고, 생성된 WTA-PGN가 단계 2)의 β-lytic 효소에 의해 쉽게 분해될 수 있다. Double mutant strain (ΔlgtΔoatA obtained in the above step 1)) whereby the lgt gene deletion not likely to be contaminated by the lipoprotein can be obtained pure WTA-PGN. In addition, the deletion of the oatA gene eliminates the acetyl group in the MurNAc residue of the PGN, and the resulting WTA-PGN can be easily degraded by the β-lytic enzyme of step 2).
상기 이중 돌연변이 균주는 야생형 포도상구균, 예를 들면 메티실린-내성 포도상구균(MRSA), 메티실린-민감성 포도상구균(MSSA) 또는 병원성 포도상구균으로부터 공지된 방법에 의해 수득될 수 있다. 예를 들면, 상기 이중 돌연변이 균주는 플레오마이신(phleomycin)에 내성을 가지면서 lgt 유전자가 결손된 T363 균주(Nakayama M et al., Journal of Immunology 189:5903-591, 2012)와 에리트로마이신에 내성을 가지면서 oatA 유전자가 결손된 T0003 균주(Park KH et al., Journal of Biological Chemistry 285, 27167-27175, 2010) 각각의 유전자를 파아지 80을 매개로 하여 형질도입하여 제조될 수 있다.Such double mutant strains can be obtained by known methods from wild type staphylococci, for example methicillin-resistant staphylococci (MRSA), methicillin-sensitive staphylococci (MSSA) or pathogenic staphylococci. For example, the double mutant strain T363 strain of the lgt gene while having a resistance to play Oh, my God (phleomycin) defect (Nakayama M et al, Journal of Immunology 189:. 5903-591, 2012) and resistant to erythromycin T0003 strain (Park KH et al. , Journal of Biological Chemistry 285, 27167-27175, 2010) with oatA gene deficient can be prepared by transduction through phage 80.
상기 단계 2)는 상기 이중 돌연변이 균주를 40 내지 70℃의 온도로 가열하는 단계를 제공한다.Step 2) provides a step of heating the double mutant strain to a temperature of 40 to 70 ℃.
상기 온도는 45 내지 68℃, 50 내지 65℃ 또는 55 내지 63℃일 수 있고, 본 발명의 일실시예에서 상기 온도는 60℃일 수 있다.The temperature may be 45 to 68 ℃, 50 to 65 ℃ or 55 to 63 ℃, in one embodiment of the present invention the temperature may be 60 ℃.
예를 들면, 상기 단계 2)는 단계 1)에서 수득한 이중 돌연변이 균주를 배양한 뒤, 가열처리하는 단계를 포함할 수 있다.For example, step 2) may comprise the step of culturing the double mutant strain obtained in step 1), followed by heat treatment.
상기 단계 3)은 상기 가열된 균주에 β-lytic 효소를 처리하는 단계를 제공한다. Step 3) provides a step of treating β-lytic enzyme on the heated strain.
상기 β-lytic 효소는 단계 2)에서 수득한 불용성 WTA-PGN의 MurNAc 잔기에 존재하는 스템펩타이드를 연결하는 펜타글리신((Gly)5) 결합을 분해하는 역할을 함으로써 가열된 균주에 포함된 불용성 WTA-PGN을 가용성 WTA-PGN으로 변화시킨다.The β-lytic enzyme plays a role in degrading pentaglycine ((Gly) 5) linkages linking the stem peptides present at the MurNAc residue of the insoluble WTA-PGN obtained in step 2), thereby insoluble WTA contained in the heated strain. Change PGN to soluble WTA-PGN.
상기 β-lytic 효소는 상업적으로 입수하거나 문헌[Li et al., Journal of Biochemitry 122, 772-778, 1997]에 기재된 방법에 따라 분리 및 정제될 수 있다. 상업적으로 입수 가능한 β-lytic 효소의 예로는 라이소스타핀(lysostaphin)이 있다.Such β-lytic enzymes are commercially available or described in Li et al. , Journal of Biochemitry 122, 772-778, 1997, can be isolated and purified. An example of a commercially available β-lytic enzyme is lysostaphin.
상기 단계 3)은 단계 2)에서 가열된 균주를 완충액에 현탁한 뒤, β-lytic 효소를 가하여 30 내지 40℃에서 10 내지 14시간 동안 교반하면서 반응시킴으로써 수행될 수 있다.Step 3) may be carried out by suspending the strain heated in step 2) in a buffer, and then adding β-lytic enzyme and reacting with stirring at 30 to 40 ° C. for 10 to 14 hours.
상기 단계 4)는 상기 단계 3)의 효소 첨가물로부터 가용성 WTA-PGN 함유 분획을 수득하는 단계를 제공한다.Step 4) provides a step of obtaining a soluble WTA-PGN containing fraction from the enzyme additive of step 3).
상기 단계는 β-lytic 효소 첨가물을 HPLC에 통과시켜 분획을 얻은 후, 상기 분획에서 가용성 WTA-PGN이 함유된 분획을 선택함으로써 수행될 수 있다. 이때, WTA-PGN이 함유된 분획은 PAGE 또는 질산은(silver nitrate) 염색 방법으로 확인할 수 있다.This step can be carried out by passing the β-lytic enzyme additive through HPLC to obtain a fraction, and then selecting the fraction containing soluble WTA-PGN in the fraction. At this time, the fraction containing WTA-PGN can be confirmed by PAGE or silver nitrate staining method.
상기 HPLC 정제에 사용되는 컬럼은, WTA의 리비톨 포스페이트의 음이온과 결합하는 음이온 교환수지인 HiTrap Q FF(GE Healthcare)일 수 있다.The column used in the HPLC purification may be HiTrap Q FF (GE Healthcare), which is an anion exchange resin that binds with the anion of WTA ribitol phosphate.
상기 단계 5)는 상기 가용성 WTA-PGN 함유 분획에 라이소자임 또는 뮤타노라이신(mutanolysin)을 첨가하는 단계를 제공한다.Step 5) provides the step of adding lysozyme or mutanolysine to the soluble WTA-PGN containing fractions.
상기 라이소자임 또는 뮤타노라이신은 PGN의 MurNAc 및 GlcNAc 사이의 결합을 분해함으로써, 중합체성 PGN을 올리고머성 PGN으로 변화시킨다.The lysozyme or mutanolysine transforms the polymeric PGN into an oligomeric PGN by breaking down the bond between MurNAc and GlcNAc of the PGN.
상기 단계 5)는 단계 4)에서 수득한 가용성 WTA-PGN을 완충액에 현탁한 뒤, 라이소자임 또는 뮤타노라이신을 가하여 30 내지 40℃에서 10 내지 14시간 동안 교반하면서 반응시킴으로써 수행될 수 있다.Step 5) may be carried out by suspending the soluble WTA-PGN obtained in step 4) in a buffer, and then adding lysozyme or mutanolysine for 10 to 14 hours with stirring at 30 to 40 ° C.
상기 단계 6)은 상기 단계 5)의 효소 첨가물로부터 가용성 WTA-PGN을 수득하는 단계를 제공한다.Step 6) provides a step of obtaining soluble WTA-PGN from the enzyme additive of step 5).
상기 단계는 라이소자임 또는 뮤타노라이신 효소 첨가물을 HPLC에 통과시켜 분획을 얻은 후, 상기 분획에서 가용성 WTA-PGN이 함유된 분획을 선택함으로써 수행될 수 있다. 이때, WTA-PGN이 함유된 분획은 PAGE 또는 질산은(silver nitrate) 염색 방법으로 확인할 수 있다.The step can be carried out by passing lysozyme or mutanolysine enzyme additive through HPLC to obtain a fraction, and then selecting the fraction containing soluble WTA-PGN from the fraction. At this time, the fraction containing WTA-PGN can be confirmed by PAGE or silver nitrate staining method.
상기 HPLC 정제에 사용되는 컬럼은 HiTrap-Q(GE Healthcare)일 수 있다.The column used for HPLC purification may be HiTrap-Q (GE Healthcare).
본 발명에 따른 방법은 단계 6) 이후에 WTA-PGN을 추가 정제하는 단계를 더 포함할 수 있다.The method according to the invention may further comprise further purifying the WTA-PGN after step 6).
상기 추가 정제는 겔 여과 크로마토그래피 또는 역상(reverse phase) 액체 크로마토그래피에 의해 수행될 수 있다. Said further purification can be carried out by gel filtration chromatography or reverse phase liquid chromatography.
본 발명의 일 실시예에서, 단계 6)에서 제조된 가용성 WTA-PGN은 세파크릴 S-200 HR 컬럼을 이용한 겔 여과 크로마토그래피 또는 Symmetry ShieldTM RP18 컬럼을 이용한 역상 액체 크로마토그래피를 거쳐 추가 정제될 수 있다. In one embodiment of the invention, the soluble WTA-PGN prepared in step 6) can be further purified via gel filtration chromatography using Sephacryl S-200 HR column or reverse phase liquid chromatography using Symmetry Shield RP18 column. have.
본 발명의 다른 실시예에서, 단계 6)에서 제조된 가용성 WTA-PGN은 세파크릴 S-200 HR 컬럼을 이용한 겔 여과 크로마토그래피와 2회의 Symmetry ShieldTM RP18 컬럼을 이용한 역상 액체 크로마토그래피를 거쳐 추가 정제될 수 있다.In another embodiment of the present invention, the soluble WTA-PGN prepared in step 6) is further purified by gel filtration chromatography using Sephacryl S-200 HR column and reverse phase liquid chromatography using two Symmetry Shield RP18 columns. Can be.
상기 크로마토그래피에서 얻어진 각 분획을 마우스 복강에 주사한 후 생산되는 IL-17A 생성량에 기초하여 활성 분획을 선택할 수 있다.The active fraction can be selected based on the amount of IL-17A produced after injection of each fraction obtained in the chromatography into the mouse abdominal cavity.
이하, 본 발명을 하기 실시예에 의거하여 상세히 설명한다. 단, 하기 실시예는 본 발명을 예시하는 것일 뿐, 본 발명이 하기 실시예에 의해 제한되지 않는다.Hereinafter, the present invention will be described in detail based on the following examples. However, the following examples are merely to illustrate the invention, the present invention is not limited by the following examples.
실시예 1. WTA 유도체를 획득하기 위한 균주의 제조Example 1 Preparation of Strains for Obtaining WTA Derivatives
WTA-PGN을 분리하기 위해, 문헌[Kazue Takahashi et al., Plos One 8: e69739, 2013]에 기재된 방법에 따라, S. aureus T384 균주(RN4220 ΔlgtΔoatA의 이중 돌연변이)를 제조하였다.To isolate WTA-PGN, Kazue Takahashi et al. , Plos One 8: e69739, 2013], S. aureus T384 strain (double mutation of RN4220 ΔlgtΔoatA ) was prepared.
상기 S. aureus T384 균주는 플레오마이신(phleomycin)에 내성을 가지면서 지질단백 디아실글리세롤 트랜스퍼라아제(lipoprotein diacylglycerol transferase, lgt) 유전자가 결손된 T363 균주(Nakayama M et al., Journal of Immunology 189:5903-591, 2012)와 에리트로마이신에 내성을 가지면서 O-아세틸트랜스퍼라아제(O-acetyl transferase, oatA) 유전자가 결손된 T0003 균주(Park KH et al., Journal of Biological Chemistry 285, 27167-27175, 2010) 각각의 유전자를 파아지 80을 매개로 형질도입하여 제조하였다. 상기 균주는 lgt 유전자가 결손됨으로써 지질단백에 의한 오염 없이 WTA, WTA-PGN 및 PGN을 분리하는데 사용될 수 있다. 또한, oatA 유전자의 결손으로 인해 PGN MurNAc 잔기 6 위치 산소에 아세틸기가 없으므로 분리된 PGN이 라이소자임에 의해 쉽게 분해될 수 있다.The S. aureus strains T384 player Oh, my God while having a resistance to (phleomycin) lipoprotein diacylglycerol transferase dehydratase (lipoprotein diacylglycerol transferase, lgt) T363 strain of the deficient gene (Nakayama M et al., Journal of Immunology 189 : 5903-591, 2012) and T0003 strains resistant to erythromycin and lacking the O-acetyl transferase ( oatA ) gene (Park KH et al. , Journal of Biological Chemistry 285, 27167-). 27175, 2010) each gene was prepared by transducing the phage 80. The strain can be used to isolate WTA, WTA-PGN and PGN without contamination by lipoproteins by deletion of the lgt gene. In addition, due to the lack of the oatA gene, the PGN MurNAc residue 6 position oxygen does not have an acetyl group, so that the isolated PGN can be easily degraded by lysozyme.
실시예 2. 균주의 열처리Example 2. Heat Treatment of Strains
실시예 1에서 제조된 S. aureus T384 균주로부터 문헌[(Park KH et al., Journal of Biological Chemistry 285, 27167-27175, 2010; Jung DJ et al., Journal of Immunology 189:4951-4959, 2012]의 방법을 변형하여 불용성 WTA-PGN을 수득하였다.From the S. aureus T384 strain prepared in Example 1 (Park KH et al. , Journal of Biological Chemistry 285, 27167-27175, 2010; Jung DJ et al. , Journal of Immunology 189: 4951-4959, 2012) The method was modified to give an insoluble WTA-PGN.
구체적으로, 실시예 1의 S. aureus T384 균주를 LB10 액상배지(pH 7.0 내지 7.2)에 접종하여 180 rpm, 37℃의 조건으로 12시간 동안 배양하였다. 상기 배양된 균주 1 ㎖을 500 ㎖의 LB10 액상배지에 분주하여 진탕배양기에서 상기와 동일한 조건으로 4시간 동안 배양한 뒤, 1시간 마다 600 ㎚에서 흡광도를 측정하였다. OD600nm=0.6 부근이 되었을 때 배양을 멈추고 균을 4℃에 보관하였다.Specifically, S. aureus T384 strain of Example 1 was inoculated in LB10 liquid medium (pH 7.0 to 7.2) and incubated for 12 hours at 180 rpm, 37 ℃ conditions. 1 ml of the cultured strain was dispensed into 500 ml LB10 liquid medium and incubated for 4 hours under the same conditions as above in a shaker, and then absorbance was measured at 600 nm every hour. When the OD 600nm = 0.6 was incubated and the culture was stopped at 4 ℃.
보관된 균주를 6,000 rpm, 4℃의 조건에서 30분 동안 원심분리하여 펠렛을 수득하였다. 20 ㎖의 식염수를 첨가하여 수득된 펠렛을 재현탁시키고, 이를 3,000 xg, 4℃의 조건에서 5분 동안 원심분리하여 펠렛을 수득하였다. 수득된 펠렛을 20 ㎖의 식염수에 재현탁하고, 이를 60℃의 980 ㎖의 식염수에 희석하여, 최종 OD600nm=0.3이 되도록 하여 30분 동안 열처리하였다. 열처리 과정에서 5분마다 플라스크를 교반시켜 균주가 균일하게 열처리되도록 하였다. 열처리 후, 균주의 희석액을 30℃가 되도록 냉각하고, 이를 3,000 xg, 20℃의 조건에서 10분 동안 원심분리하여 펠렛을 수득하였다. 펠렛을 10 ㎖의 식염수에 현탁하고, 상기와 동일한 조건으로 5분 동안 원심분리하여 펠렛을 수득하였다. 수득된 펠렛을 멸균된 증류수로 3회 세척하고, 최종 3 ㎖의 증류수에 펠렛을 현탁하여 동결건조하였다.The stored strain was centrifuged at 6,000 rpm at 4 ° C. for 30 minutes to obtain pellets. 20 ml of saline was added to resuspend the pellet, which was centrifuged for 5 minutes at 3,000 × g, 4 ° C. to obtain pellet. The obtained pellet was resuspended in 20 ml of saline, which was diluted in 980 ml of saline at 60 ° C. and heat-treated for 30 minutes to a final OD 600 nm = 0.3. The flask was stirred every 5 minutes during the heat treatment process so that the strain was uniformly heat treated. After the heat treatment, the diluted solution of the strain was cooled to 30 ° C., which was centrifuged at 3,000 × g, 20 ° C. for 10 minutes to obtain pellets. The pellet was suspended in 10 ml saline and centrifuged for 5 minutes under the same conditions as above to obtain the pellet. The obtained pellet was washed three times with sterile distilled water, and the pellet was suspended in 3 ml of distilled water and lyophilized.
실시예 3. β-lytic 효소 및 라이소자임의 처리Example 3 Treatment of β-lytic Enzymes and Lysozyme
<3-1> β-<3-1> β- lyticlytic 효소의 준비 Preparation of Enzymes
가용성 WTA-PGN을 제조하는데 필요한 β-lytic 효소를 문헌[Li et al., Journal of Biochemistry 122, 772-778, 1997]의 방법을 참조하여 제조하였다.Β-lytic enzymes required to prepare soluble WTA-PGN are described in Li et al. , Journal of Biochemistry 122, 772-778, 1997].
먼저, 5 g의 조 아크로모펩티다아제(crude achromopeptidase)를 500 ㎖의 소디움 시트레이트 완충액(pH 6.0)에 녹인 후, 15,000 rpm, 4℃의 조건으로 15분 동안 원심분리하였다. 상등액을 10 mM 소디움 시트레이트 완충액(pH 6.0)으로 평형화시킨 CM-세파로스 패스트 플로우 컬럼(3 ㎝×18 ㎝)에 로딩하고, 0 M에서 0.5 M의 NaCl 농도까지 선행 구배를 수행하여 용출하였다. 용출된 용액을 280 ㎚에서 흡광도를 측정하고, 용균활성을 나타내는 분획을 모아 농축하였다. 농축된 시료를 세파크릴 S-100 컬럼(1.6 ㎝×87 ㎝)에서 200 mM의 NaCl이 포함된 10 mM 소디움 시트레이트 완충액(pH 6.0)을 사용하여 크기 배제 크로마토그래피를 하였다. 용출액을 280 ㎚에서 흡광도를 측정하고, 흡광도가 높은 분획 중 용균활성을 나타내는 분획을 모아 다시 농축하였다. Li et al. (1997)의 결과를 바탕으로, 농축된 분획 중에서 β-lytic 효소 분획을 선별하였다. 선별된 분획을 수퍼덱스-75 컬럼(1 ㎝×30 ㎝)에서 200 mM의 NaCl이 포함된 10 mM 소디움 시트레이트 완충액(pH 6.0)을 사용하여 크기 배제 크로마토그래피를 수행하였다. 그 결과, 최종적으로 β-lytic 효소를 수득하였다.First, 5 g of crude achromopeptidase was dissolved in 500 ml of sodium citrate buffer (pH 6.0), and then centrifuged for 15 minutes at 15,000 rpm and 4 ° C. The supernatant was loaded into a CM-Sepharose fast flow column (3 cm x 18 cm) equilibrated with 10 mM sodium citrate buffer (pH 6.0) and eluted by performing a prior gradient from 0 M to 0.5 M NaCl concentration. The eluted solution was measured for absorbance at 280 nm, and the fractions showing lytic activity were collected and concentrated. The concentrated sample was subjected to size exclusion chromatography on a Sephacryl S-100 column (1.6 cm x 87 cm) using 10 mM sodium citrate buffer (pH 6.0) containing 200 mM NaCl. The eluate was measured for absorbance at 280 nm, and the fractions showing lytic activity among the fractions with high absorbance were collected and concentrated again. Li et al. Based on the results of (1997), β-lytic enzyme fractions were selected from the concentrated fractions. The selected fractions were subjected to size exclusion chromatography on a Superdex-75 column (1 cm × 30 cm) using 10 mM sodium citrate buffer (pH 6.0) containing 200 mM NaCl. As a result, β-lytic enzyme was finally obtained.
수득한 β-lytic 효소를 마이크로코커스 루테우스(ATCC 9341)나 불용성의 포도상구균에서 유래된 PGN 현탁액과 함께 배양하여, 상기 효소의 용균활성 또는 용해활성을 확인하였다. 또한, Procise Protein sequencer(Cat. #491-0, Applied Biosystems, 미국)나 전기영동의 방법으로 수득된 효소를 확인하였다. 그 결과, 수득된 효소는 N-말단의 서열이 S-P-N-G-L-L-Q-F-P-F이고(서열번호 5), 그 크기는 약 20 kDa임을 확인함으로써, β-lytic 효소임을 알 수 있었다.The obtained β-lytic enzyme was incubated with PGN suspension derived from Micrococcus luteus (ATCC 9341) or insoluble staphylococcus, and the lytic activity or lytic activity of the enzyme was confirmed. In addition, enzymes obtained by Procise® Protein sequencer (Cat. # 491-0, Applied Biosystems, USA) or electrophoresis were identified. As a result, it was found that the obtained enzyme was a β-lytic enzyme by confirming that the N-terminal sequence was S-P-N-G-L-L-Q-F-P-F (SEQ ID NO: 5) and the size was about 20 kDa.
<3-2> β-<3-2> β- lyticlytic 효소의 처리 Processing of enzymes
실시예 2에서 열처리한 균주 100 ㎎ 당 10 ㎖의 20 mM Tris-HCl 완충액(pH 7.0)에 현탁시키고, 원심분리하여 상등액을 제거하는 과정을 3회 반복하였다. 상등액의 pH가 7.0이 된 것을 확인하고, 열처리된 S. aureus T384 균주 100 ㎎ 당 약 355.57 ㎍의 β-lytic 효소를 첨가하였다. 이를 180 rpm, 37℃의 조건으로 12시간 동안 교반하면서 반응시켰다. 이후, 반응액을 60℃의 항온수조에 10분 동안 두어 효소를 비활성화시키고, 15,000 rpm, 4℃의 조건으로 10분 동안 원심분리하여 상등액을 수득하였다. 수득된 상등액을 0.45 ㎛의 포어 사이즈를 갖는 필터로 여과하고, 여과액을 20 mM Tris-HCl 완충액(pH 7.0)(완충액 A)으로 평형화된 HiTrap Q FF 컬럼이 장착된 HPLC(805 MANOMETRIC MODULE, 811C DYNAMIC MIXER, 305 PUMP, 306 PUMP, 151 UV/VIS Detector, Gilson, 미국)에 주입하였다. 그 후, 1 M NaCl이 포함된 20 mM Tris-HCl 완충액(pH 7.0)(완충액 B)을 0 M에서 1 M의 NaCl 농도까지 선형 구배를 수행하여 용출하였다. 완충액 B의 농도구배는 80%까지 구배로 100분, 100%까지 구배로 1분 동안 수행한 뒤, 100%로 10분간 용출시켰다.The process of suspending in 10 ml of 20 mM Tris-HCl buffer (pH 7.0) per 100 mg of the strain heat treated in Example 2 and centrifugation to remove the supernatant was repeated three times. It was confirmed that the pH of the supernatant was 7.0, and about 355.57 μg of β-lytic enzyme was added per 100 mg of the heat-treated S. aureus T384 strain. It was reacted with stirring at 180 rpm and 37 ° C. for 12 hours. Thereafter, the reaction solution was placed in a constant temperature water bath at 60 ° C. for 10 minutes to deactivate the enzyme, and centrifuged at 15,000 rpm and 4 ° C. for 10 minutes to obtain a supernatant. The supernatant obtained was filtered with a filter having a pore size of 0.45 μm and the filtrate was HPLC (805 MANOMETRIC MODULE, 811C) equipped with a HiTrap Q FF column equilibrated with 20 mM Tris-HCl buffer (pH 7.0) (buffer A). DYNAMIC MIXER, 305 PUMP, 306 PUMP, 151 UV / VIS Detector, Gilson, USA). Thereafter, 20 mM Tris-HCl buffer (pH 7.0) (buffer B) containing 1 M NaCl was eluted by performing a linear gradient from 0 M to 1 M NaCl concentration. The concentration gradient of the buffer B was performed for 100 minutes in a gradient up to 80%, 1 minute in a gradient up to 100%, and then eluted at 100% for 10 minutes.
용출 결과, 도 1에서 보는 바와 같이, 총 6개의 분획을 수득하였다. 각각의 분획을 A, B, C, D, E 및 F로 명명하고, 이들 각각의 분획을 아세톤으로 침전시킨 후, 동결건조하였다.As a result of the elution, a total of 6 fractions were obtained as shown in FIG. 1. Each fraction was named A, B, C, D, E and F, and each of these fractions was precipitated with acetone and then lyophilized.
<3-3> <3-3> 라이소자임의Lysozyme 처리 process
동결건조된 A 내지 F 분획 각각을 20 mM Tris-HCl 완충액(pH 7.0)에 현탁하였다. 여기에 동결건조된 분획 1 ㎎ 당 12.5 ㎍의 라이소자임(Cat. #62970, Sigma-Aldrich Co. LLC., 미국)을 첨가하여 180 rpm, 37℃의 조건으로 12시간 동안 교반하면서 반응시켰다. 이후의 과정은 HiTrap Q 컬럼을 사용한 것을 제외하고는, 실시예 <3-2>와 동일한 조건 및 방법으로 수행하였다.Each lyophilized A to F fraction was suspended in 20 mM Tris-HCl buffer (pH 7.0). 12.5 μg of lysozyme (Cat. # 62970, Sigma-Aldrich Co. LLC., USA) was added to 1 mg of the lyophilized fraction and reacted with stirring at 180 rpm and 37 ° C. for 12 hours. The subsequent procedure was performed under the same conditions and methods as in Example <3-2>, except that the HiTrap Q column was used.
용출 결과, 도 2에서 보는 바와 같이, 각각의 분획에 따라 1 내지 3개의 피크가 확인되었다.As a result of the elution, as shown in FIG. 2, one to three peaks were identified according to each fraction.
상기 확인된 각각의 피크를 A 분획에서 수득한 A 피크이면 AL-A, B 분획에서 수득한 A 피크이면 BL-A와 같은 방법으로 명명하였고, AL-A, BL-A, CL-B, DL-B, DL-C, EL-C 및 FL-B 피크를 각각 아세톤으로 침전시킨 후, 동결건조하였다.Each of the peaks identified above were named as AL-A for the A peak obtained in the A fraction, and the same as the BL-A for the A peak obtained in the B fraction, and AL-A, BL-A, CL-B, and DL. The -B, DL-C, EL-C and FL-B peaks were respectively precipitated with acetone and then lyophilized.
실시예 4. 피크별 IL-17A 생산량 증가 효과 확인Example 4. Confirmation of IL-17A Production Increase Effect by Peak
실시예 <3-3>에서 수득한 피크별 IL-17A의 생산량 증가 효과를 확인하였다.It was confirmed the effect of increasing the yield of IL-17A for each peak obtained in Example <3-3>.
구체적으로, 수득된 피크 중, CL-B, DL-B 및 DL-C 피크 각각과 EL-C 및 FL-B 피크를 혼합한 혼합 피크(EL-C+FL-B)를 각각 동결건조하여 준비하였다. 한편, 생후 5주령에 체중 15±0.5 g인 C57BL/6J 암컷 마우스를 한국생명공학연구원 의생명마우스센터(오창캠퍼스, 한국)로부터 구입하여 시판 고형사료(Cat. #5L79, (주)오리엔트바이오, 한국)를 먹이면서 1주일 동안 환경(20 내지 25℃, 습도 55%)에 적응시켰다. 체중에 따른 난괴법(completely randomized design)으로 그룹당 6 내지 12마리씩 나누어 그룹별로 6마리씩 사육용 케이지에 넣어 자유급식(ad libitum)으로 식이 및 식수를 제공하였다. 각 마우스의 체중 및 식이섭취량은 매일 1회 측정하였고, 12시간 간격으로 조명을 점등 및 소등하였다.Specifically, among the obtained peaks, the mixed peaks (EL-C + FL-B) in which each of the CL-B, DL-B and DL-C peaks and the EL-C and FL-B peaks were mixed were prepared by lyophilization, respectively. It was. Meanwhile, C57BL / 6J female mice weighing 15 ± 0.5 g at 5 weeks of age were purchased from Korea Research Institute of Bioscience and Biomedical Research Center (Ochang Campus, Korea) and were commercially available solid feeds (Cat. # 5L79, Orient Bio, Inc.). Korea) was adapted to the environment (20-25 ° C., humidity 55%) for one week while feeding. Six to twelve animals per group were divided into six groups per group in a completely randomized design to provide diet and drinking water by ad libitum. Body weight and dietary intake of each mouse were measured once daily and the lights were turned on and off at 12 hour intervals.
마우스에 상기 준비한 동결건조된 각각의 피크 분획물 200 ㎍을 10 ㎎/㎖의 농도로 LPS-free water에 현탁하고, 상기 현탁액 20 ㎕를 80 ㎕의 PBS(Cat. #17-516Q, LONZA, 미국)에 희석하여 총 100 ㎕의 희석액을 마우스 복강 내로 주입하였다. 주입 6시간 후 마우스를 희생하여 마우스의 복강 침출액을 분리하였다. 이를 위하여, 먼저 2 ㎖의 Ca2 +/Mg2 + free-PBS로 마우스의 복강을 세척한 뒤, 복강 침출액(peritoneal lavage fluid)을 200 xg, 4℃ 조건으로 10분 동안 원심분리하여 상등액을 모아 -80℃에 보관하였다. IL-17A의 양은 ELISA Ready-SET-Go! 키트(Cat. #88-7371-88, eBioscience, 미국)를 사용하여 제조사의 프로토콜대로 측정하였다. 그 결과, 450 ㎚에서 측정한 흡광도 값을 550 ㎚에서 측정한 흡광도 값으로 보정한 값을 최종 IL-17A의 양으로 결정하였다.200 μg of each of the prepared lyophilized peak fractions was suspended in LPS-free water at a concentration of 10 mg / ml, and 20 μl of the suspension was added to 80 μl of PBS (Cat. # 17-516Q, LONZA, USA). A total of 100 μl of diluent was injected into the mouse intraperitoneal by diluting. Six hours after the injection, the mice were sacrificed to separate the peritoneal leachate from the mice. To this end, the first 2 ㎖ of Ca 2 + / Mg 2 + as free-PBS after washing the abdominal cavity of the mouse, collecting the supernatant by centrifugation for 10 minutes in the abdominal cavity leachate (peritoneal lavage fluid) 200 xg, 4 ℃ conditions Store at -80 ° C. The amount of IL-17A is ELISA Ready-SET-Go! The kit (Cat. # 88-7371-88, eBioscience, USA) was used to measure according to the manufacturer's protocol. As a result, a value obtained by correcting the absorbance value measured at 450 nm to the absorbance value measured at 550 nm was determined as the amount of final IL-17A.
그 결과, 도 3에 나타난 바와 같이, DL-B 피크를 동결건조하여 복강에 주사한 마우스에서 유의적으로 높은 수준의 IL-17A가 생산되는 것을 확인하였다.As a result, as shown in Figure 3, it was confirmed that significantly high levels of IL-17A are produced in mice injected with the abdominal cavity by lyophilizing the DL-B peak.
실시예 5. 가용성 WTA-PGN의 정제-(1)Example 5 Purification of Soluble WTA-PGN— (1)
실시예 <3-3>에서 동결건조된 분획물을 다양한 컬럼을 이용하여 추가로 정제하였다.The lyophilized fractions in Example <3-3> were further purified using various columns.
먼저, CL-B 피크 분획물 50 ㎎을 400 ㎕의 증류수에 현탁하고, 현탁액을 HPLC 장치(805 MANOMETRIC MODULE, 811C DYNAMIC MIXER, 305 PUMP, 306 PUMP, 151 UV/VIS Detector, Gilson, 미국)에 주입하였다. 주입 전에 세파크릴 S-200 HR 컬럼(Cat.#17-0584-01, GE Healthcare Life Sciences, 영국)을 연결하여 증류수로 세척하고, 멸균 증류수로 평형화시켰다. 또한, 실험 시 불순물 유입에 따른 오차가 없도록 유속 0.3 ㎖/분, 민감도 2, 피크폭 10.0초 및 UV 흡광도 220 ㎚의 조건으로 UV 검출기가 안정화될 때까지 용매를 흘려주었다. UV 값이 안정화된 뒤, 주입기를 로드 상태로 변환시켜 상기 현탁된 피크 분획물을 주입하였다. 용출 전에 주입기를 주입 상태로 변환시켜 용출액을 수득하였으며, 용출 조건은 평형화 조건과 동일하게 하였다. 수득된 용출액을 ‘세파크릴-B 분획’이라 명명하고 동결건조하여 보관하였다.First, 50 mg of CL-B peak fraction was suspended in 400 μl of distilled water, and the suspension was injected into an HPLC apparatus (805 MANOMETRIC MODULE, 811C DYNAMIC MIXER, 305 PUMP, 306 PUMP, 151 UV / VIS Detector, Gilson, USA). . Before injection, Sephacryl S-200 HR columns (Cat. # 17-0584-01, GE Healthcare Life Sciences, UK) were connected and washed with distilled water and equilibrated with sterile distilled water. In addition, the solvent was allowed to flow until the UV detector was stabilized under conditions of a flow rate of 0.3 ml / min, a sensitivity of 2, a peak width of 10.0 sec, and a UV absorbance of 220 nm so that there was no error due to impurity inflow during the experiment. After the UV values had stabilized, the injector was converted to the loaded state to inject the suspended peak fractions. The eluent was obtained by converting the injector into the injection state before elution, and the elution conditions were the same as the equilibration conditions. The obtained eluate was named 'Sepacryl-B fraction' and stored by lyophilization.
이후, TSK G2000SW 컬럼을 이용하여 세파크릴-B 분획을 더 정제하였다. 구체적으로, 동결건조된 세파크릴-B 분획 3.5 ㎎을 100 mM의 NaCl이 포함된 5 mM 인산나트륨 완충액(pH 6.0)에 현탁하여 시료를 준비하였다. 한편, 컬럼은 상기 완충액으로 세척 및 평형화시켜 준비하였다. 유속 3 ㎖/분, 민감도 0.5, 피크 너비 10 초 및 UV 흡광도 202 ㎚의 조건으로 UV 검출기가 안정화될 때까지 평형화시켰다. 평형화시킨 뒤, 주입구를 로드상태로 변환시켜 시료를 천천히 주입하고, 용출을 시작할 때 주입구를 주입으로 변환시켜 용출액을 수득하였다. 이때, 용출 완충액으로는 상기 100 mM의 NaCl이 포함된 5 mM 인산나트륨 완충액(pH 6.0)을 사용하였다.Thereafter, the Sephacryl-B fraction was further purified using a TSK G2000SW column. Specifically, 3.5 mg of the lyophilized Sephacryl-B fraction was suspended in 5 mM sodium phosphate buffer (pH 6.0) containing 100 mM NaCl to prepare a sample. On the other hand, the column was prepared by washing and equilibrating with the buffer. Equilibration was performed until the UV detector stabilized under conditions of flow rate 3 ml / min, sensitivity 0.5, peak width 10 seconds and UV absorbance 202 nm. After equilibration, the inlet was changed to a rod state to slowly inject the sample, and when the elution started, the inlet was changed to infusion to obtain an eluate. In this case, 5 mM sodium phosphate buffer (pH 6.0) containing 100 mM NaCl was used as the elution buffer.
수득된 분획물의 IL-17A 생산량을 실시예 4와 동일한 조건 및 방법으로 확인한 뒤, IL-17A의 생산량을 가장 유의적으로 증가시킨 B 분획을 ‘세파크릴-B-TSK-B 분획’이라 명명하고 동결건조하여 보관하였다.After confirming the IL-17A production amount of the obtained fraction by the same conditions and methods as in Example 4, the B fraction which most significantly increased the production of IL-17A was named 'Sepacryl-B-TSK-B fraction'. Lyophilized and stored.
동결건조된 세파크릴-B-TSK-B 분획을 더 정제하기 위하여 C18 역상 컬럼을 이용하였다.A C18 reversed phase column was used to further purify the lyophilized Sephacryl-B-TSK-B fraction.
구체적으로, 동결건조된 세파크릴-B-TSK-B 분획 3 ㎎을 0.05%(v/v)의 트리플로오로아세트산(TFA, Cat. #204-1771, Wako, 일본) 완충액(증류수 1 ℓ 중 0.5 ㎖ TFA) 200 ㎕에 현탁하여 시료를 준비하였다. 유속 1 ㎖/분, 민감도 1, 컬럼 온도 40℃ 및 UV 흡광도는 202 ㎚의 조건으로 UV 검출기를 안정화시킨 뒤, 용출을 수행하였다. 용출을 위한 이동상의 농도 구배는 0%로 10분, 41.7%까지 구배로 25분 및 100%까지 구배로 2분 동안 수행한 뒤, 100%로 10분간 용출시켰다. 이때, 이동상 용매로서 0.05%(v/v)의 트리플로오로아세트산/40%(v/v)의 아세토니트릴(Cat. #AH015-4, Burdick & Jackson, 미국) 완충액(증류수 600 ㎖, 아세토니트릴 400 ㎖ 및 0.5 ㎖ TFA)이 사용되었다.Specifically, 3 mg of lyophilized Sephacryl-B-TSK-B fraction was added to 0.05% (v / v) of trifluoroacetic acid (TFA, Cat. # 204-1771, Wako, Japan) buffer (1 liter of distilled water). Samples were prepared by suspending in 200 μl of 0.5 mL TFA). Flow rate 1 ml / min, sensitivity 1, column temperature 40 ° C. and UV absorbance were stabilized at 202 nm under UV detector, followed by elution. The concentration gradient of the mobile phase for elution was performed for 10 minutes at 0%, 25 minutes with gradients up to 41.7% and 2 minutes with gradients up to 100%, followed by elution at 100% for 10 minutes. At this time, 0.05% (v / v) of trifluoroacetic acid / 40% (v / v) of acetonitrile (Cat. # AH015-4, Burdick & Jackson, USA) buffer (600 mL of distilled water, acetonitrile) as a mobile phase solvent 400 ml and 0.5 ml TFA) were used.
그 결과, 도 4에 나타난 바와 같이, A 내지 F의 6개 피크로 분리된 분획이 확인되었다.As a result, as shown in FIG. 4, a fraction separated into six peaks of A to F was confirmed.
실시예 6. 피크별 IL-17A 생산량 증가 효과 확인Example 6 Confirmation of the Effect of IL-17A Production Increase by Peak
실시예 5에서 수득된 각 분획을 이용하여 실시예 4와 동일한 조건 및 방법으로 마우스에서의 IL-17A 유도 활성을 확인하여, 그 결과를 도 5에 나타내었다.Each fraction obtained in Example 5 was used to confirm IL-17A inducing activity in mice under the same conditions and methods as in Example 4, and the results are shown in FIG. 5.
도 5에 나타난 바와 같이, C 피크의 분획을 동결건조하여 복강에 주사한 마우스에서 유의적으로 높은 수준의 IL-17A가 생산되는 것을 확인하였다.As shown in FIG. 5, it was confirmed that significantly high levels of IL-17A were produced in mice injected into the abdominal cavity by lyophilizing the fraction of the C peak.
실시예 7. 가용성 WTA-PGN의 정제-(2)Example 7. Purification of Soluble WTA-PGN— (2)
실시예 5에서 수득한 동결건조된 C 내지 E 피크의 분획 2 ㎎을 사용하여, 이를 보다 순수하게 정제하기 위하여 실시예 5와 동일한 조건 및 방법으로 C18 역상 컬럼을 이용하였다. Using a fraction of 2 mg of the lyophilized C to E peak obtained in Example 5, a C18 reversed phase column was used in the same conditions and methods as in Example 5 to purify it more purely.
그 결과, 도 6에 나타난 바와 같이, C, D 및 E의 3개 피크로 분리된 분획이 확인되었고, 이들 분획을 이용하여 실시예 4와 동일한 조건 및 방법으로 마우스에서의 IL-17A 유도 활성을 확인한 결과, 도 7에 나타난 바와 같이, C 분획에서 가장 많은 양의 IL-17A가 생산된 것을 확인하였다.As a result, as shown in FIG. 6, fractions separated into three peaks of C, D, and E were identified, and IL-17A-induced activity in mice was observed under the same conditions and methods as in Example 4 using these fractions. As a result, as shown in Figure 7, it was confirmed that the highest amount of IL-17A produced in the C fraction.
실시예 8. PGN의 구조 확인Example 8 Confirmation of Structure of PGN
<8-1> <8-1> WTA의Of WTA 제거 remove
실시예 7에서 IL-17A의 생산량 증가 활성을 나타낸 분획 C의 구조를 확인하기 위하여, 먼저 WTA를 제거하였다.In Example 7, WTA was first removed to confirm the structure of Fraction C, which showed a production increase activity of IL-17A.
구체적으로, 동결건조된 C 분획 3 ㎎을 50 ㎕의 멸균 증류수에 용해시켰다. 여기에 50 ㎕의 25% TFA를 첨가하고, 37℃의 온도에서 12시간 동안 교반하며 배양하였다. 배양 후, 100 ㎕의 멸균 증류수를 첨가하여, 최종 6.25% 농도의 TFA가 되도록 준비하였다. 이후, WTA가 제거된 PGN만을 정제하기 위하여 실시예 5와 동일한 조건 및 방법으로 Symmetry Shield™ RP18 컬럼을 이용하였다. 이때, 유속 1 ㎖/분, 민감도 0.5, 컬럼 온도 40℃ 및 UV 흡광도 203 ㎚의 조건으로 수행하였다. 용출을 위한 이동상의 농도구배는 0%로 10분, 43.8%까지 구배로 50분, 100%까지 구배로 2분 동안 수행한 뒤, 100%로 10분간 용출시켰다.Specifically, 3 mg of lyophilized C fraction was dissolved in 50 μl of sterile distilled water. 50 μl of 25% TFA was added thereto and incubated with stirring at a temperature of 37 ° C. for 12 hours. After incubation, 100 μl of sterile distilled water was added to prepare a final 6.25% concentration of TFA. Thereafter, the Symmetry Shield ™ RP18 column was used in the same conditions and methods as in Example 5 to purify only PGN free of WTA. At this time, the flow rate was carried out under the conditions of 1 mL / min, a sensitivity of 0.5, a column temperature of 40 ° C., and a UV absorbance of 203 nm. The concentration gradient of the mobile phase for elution was performed for 10 minutes at 0%, 50 minutes at gradient to 43.8%, 2 minutes at gradient to 100%, and then eluted at 100% for 10 minutes.
<8-2> <8-2> PGN의Of PGN 구조 확인 Check structure
실시예 <8-1>에서 수득한 용출액을 Nano-flow (n) LC-MS/MS 시스템의 자동 주입기를 통해 컬럼에 로딩하였다.The eluate obtained in Example <8-1> was loaded onto the column via an autoinjector in a Nano-flow (n) LC-MS / MS system.
nLC-MS/MS 시스템은 frit-less electrospray(ESI) 컬럼(150 ㎛×100 ㎜, Nikkyo Technos Co. Ltd, 일본)을 가진 nLC(EASY-nLC 1000, Thermo Fisher Scientific, 미국)와 고해상도의 혼성 질량 분광계(Q-Exactive mass spectrometer, Thermo Fisher Scientific, 미국)로 구성된다.nLC-MS / MS systems are high resolution hybrid masses with nLC (EASY-nLC 1000, Thermo Fisher Scientific, USA) with fritless electrospray (ESI) columns (150 μm × 100 mm, Nikkyo Technos Co. Ltd, Japan) It consists of a spectrometer (Q-Exactive mass spectrometer, Thermo Fisher Scientific, USA).
nLC 분리는 유속을 300 nℓ/분으로 하여 30분 동안 0.1%(v/v)의 포름산을 포함하는 32%(v/v)의 아세토니트릴 용액을 용출 완충액으로 이용하여 용출하였다. 상기 용출액은 양극성 상태에서 2.0 ㎸의 압력으로 질량 분석기로 온라인 분무되었고, 분석기는 MS와 MS/MS acquisition 사이에서 자동으로 전환되도록 데이타-의존적 모드로 작동시켰다. 그 결과, 전체-검사 질량 스펙트라(full-scan mass spectra, m/z 300 내지 2,000) 측정을 통해, m/z 400에서 35,000의 mass resolution을 얻었다. 조사 스캔(survey scan)에서 강도가 10,000 counts/s 이상인 10개의 가장 강한 질량 피크를 확인하고, 각각의 피크를 분획화하기 위해 3 m/z window 이내에서 분리하였다. 질량 피크는 보정된 충돌 에너지 15에서 고에너지 충돌 분리(high-energy collisional dissociation)로 분획화하였다. MS/MS 측정을 위해서는 m/z 400에서 17,500의 mass resolution을 사용하였다.nLC separation was eluted using a 32% (v / v) acetonitrile solution containing 0.1% (v / v) formic acid as elution buffer for 30 minutes at a flow rate of 300 nL / min. The eluate was sprayed online with a mass spectrometer at a pressure of 2.0 kPa in the bipolar state, and the analyzer was operated in data-dependent mode to automatically switch between MS and MS / MS acquisition. As a result, a mass resolution of 35,000 was obtained at m / z 400 through full-scan mass spectra (m / z 300 to 2,000) measurements. The ten strongest mass peaks with an intensity of 10,000 counts / s or more were identified in the survey scan, and each peak was separated within 3 m / z window to fractionate. Mass peaks were fractionated by high-energy collisional dissociation at corrected collision energy 15. For MS / MS measurements, a mass resolution of 17,500 at m / z 400 was used.
그 결과, 하기 식 1로 표시되는 구조를 갖는 PGN이 확인되었다. 이는 GlcNAc-MurNAc-GlcNAc-MurNAc-GlcNAc로 표시되는 다섯 개의 단당 사슬을 가지면서 4번째 MurNAc 잔기에 인산기가 하나 결합되어 있었다. 상기 C 분획의 PGN은 분자량이 2167.91이었다.As a result, PGN having a structure represented by the following formula 1 was confirmed. It had five monosaccharide chains represented by GlcNAc-MurNAc-GlcNAc-MurNAc-GlcNAc and had one phosphate group bound to the fourth MurNAc residue. The PGN of the C fraction had a molecular weight of 2167.91.
[식 1][Equation 1]
Figure PCTKR2016007748-appb-I000006
Figure PCTKR2016007748-appb-I000006

Claims (14)

  1. 하기 일반식 1로 표시되는 벽테이코산-부착된 펩티도글리칸(WTA-PGN)을 유효성분으로 함유하는, 포도상구균 감염 질환의 예방 또는 치료용 조성물:A composition for the prevention or treatment of Staphylococcal infection disease, which comprises a wall teichoic acid-attached peptidoglycan (WTA-PGN) represented by Formula 1 as an active ingredient:
    [일반식 1][Formula 1]
    Figure PCTKR2016007748-appb-I000007
    Figure PCTKR2016007748-appb-I000007
    상기 일반식 1에서, n은 10 내지 50의 정수이고, m은 1 내지 3의 정수이며, A는 N-아세틸만노사민(ManNAc)이고, B는 N-아세틸글루코사민(GlcNAc)이며, In Formula 1, n is an integer of 10 to 50, m is an integer of 1 to 3, A is N-acetylmannosamine (ManNAc), B is N-acetylglucosamine (GlcNAc),
    O는 1 내지 5의 정수이고, R1 및 R2는 각각 독립적으로 하이드록시, 테트라펩타이드 또는 펜타펩타이드이며, R3은 하이드록시 또는 N-아세틸뮤람산(MurNAc)이다.O is an integer from 1 to 5, R 1 and R 2 are each independently hydroxy, tetrapeptide or pentapeptide, and R 3 is hydroxy or N-acetylmuramic acid (MurNAc).
  2. 제1항에 있어서, 상기 일반식 1에서 A 및 B는 서로 β-위치로 연결된 것인, 포도상구균 감염 질환의 예방 또는 치료용 조성물.According to claim 1, wherein in Formula 1 A and B are linked to each other in the β-position, composition for the prevention or treatment of staphylococcal infection disease.
  3. 제1항에 있어서, 상기 일반식 1에서, n은 35 내지 45의 정수이고, m은 3이며, A는 ManNAc이고, B는 GlcNAc이며, O는 1 내지 5의 정수이고, R1 및 R2는 각각 독립적으로 하이드록시, 테트라펩타이드 또는 펜타펩타이드이며, R3은 하이드록시 또는 MurNAc인, 포도상구균 감염 질환의 예방 또는 치료용 조성물.The compound of claim 1, wherein in Formula 1, n is an integer of 35 to 45, m is 3, A is ManNAc, B is GlcNAc, O is an integer of 1 to 5, R 1 and R 2 Are each independently hydroxy, tetrapeptide or pentapeptide, R 3 is hydroxy or MurNAc, composition for the prevention or treatment of Staphylococcal infection disease.
  4. 제3항에 있어서, 상기 일반식 1에서, n은 40이고, m은 3이며, A는 ManNAc이고, B는 GlcNAc이며, O는 1 내지 5의 정수이고, R1 및 R2는 각각 독립적으로 테트라펩타이드 또는 펜타펩타이드이며, R3은 하이드록시 또는 MurNAc인, 포도상구균 감염 질환의 예방 또는 치료용 조성물.The method of claim 3, wherein in Formula 1, n is 40, m is 3, A is ManNAc, B is GlcNAc, O is an integer of 1 to 5, R 1 and R 2 are each independently Tetrapeptide or pentapeptide, R 3 is hydroxy or MurNAc, composition for the prevention or treatment of staphylococcal infection disease.
  5. 제4항에 있어서, 상기 테트라펩타이드는 -X1-X2-X3-X4이고, 여기서 X1은 Ala 또는 Gly이고, X2는 Glu 또는 Asp이며, X3는 Lys, Arg 또는 His이고, X4는 Ala 또는 Gly인, 포도상구균 감염 질환의 예방 또는 치료용 조성물.The method of claim 4, wherein the tetrapeptide is -X 1 -X 2 -X 3 -X 4 , wherein X 1 is Ala or Gly, X 2 is Glu or Asp, and X 3 is Lys, Arg or His , X 4 is Ala or Gly, composition for the prevention or treatment of staphylococcal infection disease.
  6. 제5항에 있어서, 상기 테트라펩타이드는 -(L-Ala)-(D-Glu)-(L-Lys)-(D-Ala)인, 포도상구균 감염 질환의 예방 또는 치료용 조성물.The composition of claim 5, wherein the tetrapeptide is-(L-Ala)-(D-Glu)-(L-Lys)-(D-Ala).
  7. 제4항에 있어서, 상기 펜타펩타이드는
    Figure PCTKR2016007748-appb-I000008
    이고, 여기서 X1은 Ala 또는 Gly이고, X2는 Glu 또는 Asp이며, X3는 Lys, Arg 또는 His이고, X4는 Ala 또는 Gly인, 포도상구균 감염 질환의 예방 또는 치료용 조성물.
    The method according to claim 4, wherein the pentapeptide is
    Figure PCTKR2016007748-appb-I000008
    Wherein X 1 is Ala or Gly, X 2 is Glu or Asp, X 3 is Lys, Arg or His, and X 4 is Ala or Gly.
  8. 제7항에 있어서, 상기 펜타펩타이드는
    Figure PCTKR2016007748-appb-I000009
    인, 포도상구균 감염 질환의 예방 또는 치료용 조성물.
    The method of claim 7, wherein the pentapeptide is
    Figure PCTKR2016007748-appb-I000009
    Phosphorus, staphylococcal infection disease prevention or treatment composition.
  9. 제1항에 있어서, 상기 포도상구균은 메티실린-내성 포도상구균(methicillin-resistant Staphylococcus aureus, MRSA), 메티실린-민감성 포도상구균(methicillin-sensitive Staphylococcus aureus, MSSA) 또는 병원성 포도상구균인, 포도상구균 감염 질환의 예방 또는 치료용 조성물.The method of claim 1, wherein the staphylococcus is Staphylococcus aureus (methicillin-resistant Staphylococcus aureus , MRSA), methicillin-sensitive Staphylococcus aureus (MSSA) or Staphylococcus aureus Composition for preventing or treating a disease.
  10. 제1항에 있어서, 상기 포도상구균 감염 질환은 연부조직 감염, 화농성 관절염, 화농성 골수염, 중이염, 폐렴, 패혈증, 급성 호흡기 감염(acute respiratory tract infection), 카테터의 사용으로 인한 감염, 수술 후 창상 감염, 균혈증, 심내막염 및 식중독으로 구성된 군으로부터 선택되는, 포도상구균 감염 질환의 예방 또는 치료용 조성물.The method of claim 1, wherein the staphylococcal infection disease is soft tissue infection, purulent arthritis, purulent osteomyelitis, otitis media, pneumonia, sepsis, acute respiratory tract infection, infection due to the use of a catheter, wound infection after surgery, Composition for the prevention or treatment of staphylococcal infection disease, selected from the group consisting of bacteremia, endocarditis and food poisoning.
  11. 제1항 내지 제10항 중 어느 한 항의 조성물을 이를 필요로 하는 개체에 투여하는 것을 포함하는, 포도상구균 감염 질환의 예방 또는 치료 방법.A method for preventing or treating a Staphylococcal infection disease, comprising administering the composition of any one of claims 1 to 10 to a subject in need thereof.
  12. 제11항에 있어서, 상기 방법은 옵소닌식균작용(opsonophagocytosis) 및 식균작용(phagocytosis)을 동시에 유도하는 것인, 포도상구균 감염 질환의 예방 또는 치료용 조성물.The method of claim 11, wherein the method induces opsonophagocytosis and phagocytosis at the same time, the composition for preventing or treating staphylococcal infection disease.
  13. 제11항에 있어서, 상기 방법은 상기 조성물의 투여 후 24시간 이내에 개체 내에서 IL-17A 생성량을 증가시키는 것인, 포도상구균 감염 질환의 예방 또는 치료용 조성물.The composition of claim 11, wherein the method increases IL-17A production in the subject within 24 hours after administration of the composition.
  14. 1) 야생형 포도상구균으로부터 lgt(lipoprotein diacylglycerol transferase) 및 oatA(O-acetyl transferase) 유전자가 결실된 이중 돌연변이 균주를 수득하는 단계;1) from wild-type staphylococcal lgt (lipoprotein diacylglycerol transferase) and oatA (O-acetyl transferase) gene comprising: obtaining a double deletion mutant strain;
    2) 상기 이중 돌연변이 균주를 40 내지 70℃의 온도로 가열하는 단계;2) heating the double mutant strain to a temperature of 40 to 70 ° C;
    3) 상기 가열된 균주에 β-lytic 효소를 첨가하는 단계;3) adding β-lytic enzyme to the heated strain;
    4) 상기 단계 3)의 효소 첨가물로부터 가용성 WTA-PGN 함유 분획을 수득하는 단계;4) obtaining a soluble WTA-PGN containing fraction from the enzyme additive of step 3);
    5) 상기 가용성 WTA-PGN 함유 분획에 라이소자임 또는 뮤타노라이신(mutanolysin)을 첨가하는 단계; 및5) adding lysozyme or mutanolysin to the soluble WTA-PGN containing fractions; And
    6) 상기 단계 5)의 효소 첨가물로부터 가용성 WTA-PGN을 수득하는 단계를 포함하는, 일반식 1로 표시되는 WTA-PGN을 제조하는 방법.6) A method for preparing WTA-PGN represented by the general formula 1, comprising the step of obtaining soluble WTA-PGN from the enzyme additive of step 5).
PCT/KR2016/007748 2015-07-15 2016-07-15 Composition for preventing or treating staphylococcal infectious diseases WO2017010845A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201562192656P 2015-07-15 2015-07-15
US62/192,656 2015-07-15

Publications (1)

Publication Number Publication Date
WO2017010845A1 true WO2017010845A1 (en) 2017-01-19

Family

ID=57758178

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2016/007748 WO2017010845A1 (en) 2015-07-15 2016-07-15 Composition for preventing or treating staphylococcal infectious diseases

Country Status (1)

Country Link
WO (1) WO2017010845A1 (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040247605A1 (en) * 2002-12-02 2004-12-09 Kokai-Kun John Fitzgerald Wall teichoic acid as a target for anti-staphylococcal therapies and vaccines
KR20100056510A (en) * 2007-09-11 2010-05-27 몬도바이오테크 래보래토리즈 아게 Use of urodilatin as a therapeutic agent
KR101062525B1 (en) * 2002-11-12 2011-09-06 더 브리검 앤드 우먼즈 하스피털, 인크. Polysaccharide Vaccine Against Staphylococcal Infections
KR20110124060A (en) * 2010-05-10 2011-11-16 부산대학교 산학협력단 Vaccine composition comprising wta as effective component
WO2013168965A2 (en) * 2012-05-07 2013-11-14 목암생명공학연구소 Vaccine composition for preventing staphylococcus aureus infection

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR101062525B1 (en) * 2002-11-12 2011-09-06 더 브리검 앤드 우먼즈 하스피털, 인크. Polysaccharide Vaccine Against Staphylococcal Infections
US20040247605A1 (en) * 2002-12-02 2004-12-09 Kokai-Kun John Fitzgerald Wall teichoic acid as a target for anti-staphylococcal therapies and vaccines
KR20100056510A (en) * 2007-09-11 2010-05-27 몬도바이오테크 래보래토리즈 아게 Use of urodilatin as a therapeutic agent
KR20110124060A (en) * 2010-05-10 2011-11-16 부산대학교 산학협력단 Vaccine composition comprising wta as effective component
WO2013168965A2 (en) * 2012-05-07 2013-11-14 목암생명공학연구소 Vaccine composition for preventing staphylococcus aureus infection

Similar Documents

Publication Publication Date Title
EP2907813B1 (en) Antibacterial pharmaceutical compositions
RU2506272C2 (en) Lantibiotic carboxyamide derivatives with enhanced antibacterial activity
JP3590638B2 (en) Glycopeptide derivatives
WO2012144790A1 (en) Cyclic peptide from nonomuraea sp., process for the production thereof, and pharmaceutical composition for the prevention or treatment of mycobacteria related disease comprising the same
WO2016108394A1 (en) Composition comprising neoagarooligosaccharide as active ingredient, for prevention or treatment of sepsis or septic shock
SU1205772A3 (en) Method of producing cephalosporines
EP1644382B1 (en) Cross-linked glycopeptide-cephalosporin antibiotics
DE4411025A1 (en) New lipopeptide A1437 derivs. with modified acyl gp.
EP1633759B1 (en) Cross-linked glycopeptide-cephalosporin antibiotics
WO2016195336A1 (en) Antibacterial peptide targeting mycobacterium tuberculosis toxin-antitoxin system, and use thereof
KR860001280B1 (en) Method of preparing noacyl glycoptide derivatives
WO2019039826A1 (en) Heterocephalus glaber-derived antimicrobial polypeptide having potent antimicrobial activity against gram-negative bacteria and pharmaceutical composition comprising same
Malabarba et al. Synthesis and biological properties of N63-carboxamides of teicoplanin antibiotics. Structure-activity relationships
US5057520A (en) 7-(peptidylpyrolidinyl)naphthyridine antibacterial compounds
WO2019190137A1 (en) 1,2-diacylglycerol compound, preparation method therefor, and immunomodulator containing same as active ingredient
US20180201646A1 (en) New bicyclic lipolantipeptide, preparation and use as antimicrobial agent
US6143739A (en) Basic proline-amide derivatives of GE 2270 and GE 2270-like antibiotics
WO2017010845A1 (en) Composition for preventing or treating staphylococcal infectious diseases
WO2017116005A1 (en) Small rna gene for regulating extracellular vesicle generation and host immune response, and use thereof
AU616053B2 (en) Amino acid quinoline and naphthyridine derivatives
WO2019235705A1 (en) Vaccine composition comprising recombinant protein of staphylococcus aureus attenuated enterotoxin and cytotoxin
WO2019004674A1 (en) Acetylcholine receptor-binding peptide
GB2182937A (en) Common antigen (psc-a)to pseudomonas aeruginosa
WO2016111574A1 (en) Novel cyclic depsipeptide-based compound, method for preparing same, and antibacterial pharmaceutical composition containing same as active ingredient
JPH02290870A (en) Enantiomerically pure 7-(3-amino-1- pyrrolidinyl)-quinolone- and naphthyridonecarboxylic acid

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: 16824761

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: 16824761

Country of ref document: EP

Kind code of ref document: A1