WO2013189950A1 - Dibenzofuran derivatives with antibacterial and wound-healing activity - Google Patents

Dibenzofuran derivatives with antibacterial and wound-healing activity Download PDF

Info

Publication number
WO2013189950A1
WO2013189950A1 PCT/EP2013/062664 EP2013062664W WO2013189950A1 WO 2013189950 A1 WO2013189950 A1 WO 2013189950A1 EP 2013062664 W EP2013062664 W EP 2013062664W WO 2013189950 A1 WO2013189950 A1 WO 2013189950A1
Authority
WO
WIPO (PCT)
Prior art keywords
wound
cooh
usnic acid
mmol
compound
Prior art date
Application number
PCT/EP2013/062664
Other languages
French (fr)
Inventor
Luisella Verotta
Michela BRUNO
Beatrice TRUCCHI
Elia RANZATO
Simona MARTINOTTI
Silvia BONETTA
Sara BONETTA
Elisabetta CARRARO
Bruno BURLANDO
Akkol Esra KUPELI
Ipek SUNTAR
Hikmet KELES
Original Assignee
Universita' Degli Studi Di Milano
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 Universita' Degli Studi Di Milano filed Critical Universita' Degli Studi Di Milano
Publication of WO2013189950A1 publication Critical patent/WO2013189950A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61QSPECIFIC USE OF COSMETICS OR SIMILAR TOILETRY PREPARATIONS
    • A61Q19/00Preparations for care of the skin
    • A61Q19/08Anti-ageing preparations
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/34Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide
    • A61K31/343Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide condensed with a carbocyclic ring, e.g. coumaran, bufuralol, befunolol, clobenfurol, amiodarone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41781,3-Diazoles not condensed 1,3-diazoles and containing further heterocyclic rings, e.g. pilocarpine, nitrofurantoin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K8/00Cosmetics or similar toiletry preparations
    • A61K8/18Cosmetics or similar toiletry preparations characterised by the composition
    • A61K8/30Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds
    • A61K8/49Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds containing heterocyclic compounds
    • A61K8/4973Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds containing heterocyclic compounds with oxygen as the only hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61QSPECIFIC USE OF COSMETICS OR SIMILAR TOILETRY PREPARATIONS
    • A61Q17/00Barrier preparations; Preparations brought into direct contact with the skin for affording protection against external influences, e.g. sunlight, X-rays or other harmful rays, corrosive materials, bacteria or insect stings
    • A61Q17/005Antimicrobial preparations
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D307/00Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom
    • C07D307/77Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom ortho- or peri-condensed with carbocyclic rings or ring systems
    • C07D307/78Benzo [b] furans; Hydrogenated benzo [b] furans
    • C07D307/82Benzo [b] furans; Hydrogenated benzo [b] furans with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to carbon atoms of the hetero ring
    • C07D307/83Oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D307/00Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom
    • C07D307/77Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom ortho- or peri-condensed with carbocyclic rings or ring systems
    • C07D307/91Dibenzofurans; Hydrogenated dibenzofurans

Definitions

  • the present invention relates to dibenzofuran derivatives useful as antibacterial and wound-healing agents and for regenerative or anti-aging treatment of the skin tissues.
  • Wound infections represent a significant problem in many post-operative disorders and in burns, and are common in many developing countries due to poor conditions of hygiene.
  • Staphylococcus aureus, Streptococcus pyogenes, Escherichia coli, Pseudomonas aeruginosa, Streptococcus pneumoniae and Klebsiella pneumoniae are some important micro-organisms that cause wound infections (Mertz P. et al., 1993). Said infections are treated with a number of antibiotics which in many cases fail during the treatment due to the onset of resistance and/or toxicity.
  • Active ingredients for use on the skin in dermatological or cosmetic formulations with regenerative or anti-aging purposes should ideally have low cytotoxicity on the skin cells, high antibacterial activity against infectious or opportunistic pathogens, and good skin regeneration properties.
  • a large number of wound-healing products of natural, chemical or biotechnological origin are currently on the market. However, in most cases these products are complex mixtures used on an empirical basis, while their action mechanisms and active ingredients have not yet been clearly identified. For example, the best results in the regeneration of chronic or serious wounds are currently obtained in clinical practice with platelet derivatives containing various growth factors and a number of low-molecular- weight compounds (Ranzato et al., 2009).
  • Usnic acid is a dibenzofuran derivative produced by various species of lichen. Its chemical name is 2,6-diacetyl-7,9-dihydroxy-8,9b- dimethyldibenzofuran-l,3(2H,9bH)-dione, corresponding to the following formula:
  • (+) Usnic acid is commercially available, being easily extracted from lichens of species belonging to the genera Usnea and Ramalina, wherein it is present in percentages exceeding 26% (Reyim M. et al., 2010).
  • the antibacterial, antifungal, antiviral and anti-inflammatory properties of usnic acid are known, as are its consequent uses in the cosmetic and pharmaceutical fields, in particular as an active ingredient of creams, deodorants, toothpastes, mouthwashes, antibiotic ointments, vaginal creams, foot creams, powders and shampoos, or as a diet supplement.
  • Usnea densirotra tea is known as "Barba della Piedra", and is used for gargling.
  • usnic acid is used as both active ingredient and preservative.
  • PCT/EP2009/006960 discloses (+) usnic acid derivatives obtained by conjugating (+) usnic acid with biologically active molecules.
  • the compounds are useful for the prophylaxis and treatment of infections caused by protozoa of the genus Plasmodium.
  • (+)-usnic acid due to its properties of stimulating the wound closure of human HaCaT keratinocyte monolayers at subtoxic doses.
  • the action mechanism was correlated with its ability to promote cell motility (Burlando B. et al., 2009).
  • (+)-usnic acid sodium salt was indicated as a promoter of the wound-healing process, due to the secretion of growth factors and the increased rate of cell migration to a dose-dependent extent (Burlando B. et al., 2009).
  • Experiments effected in vivo had confirmed the ability of usnic acid sodium salt to accelerate healing of the epidermis, but this activity was not attributed to stimulation of proliferation (Jin J. et al., 2005).
  • Recent publications report the use of formulations based on usnic acid conveyed by liposomes to heal burn wounds (Nunes P.S. et al., 2011).
  • Usnic acid is universally known as an antibiotic. (+)- and (-) usnic acids are active on sensitive and resistant strains of Gram-positive bacteria (see the review by Ingolfsdottir K., 2002). Some metal complexes of usnic acid hydrazones have proved more active (Natic M. et al, 2004). Kokubun T. et al., 2007 reports the antibacterial activity of (+) usnic acid on MRSA (methicillin-resistant S. aureus) and MDR (multidrug-resistant) strains in addition to mupirocin-resistant strains.
  • MRSA methicillin-resistant S. aureus
  • MDR multidrug-resistant
  • Usnic acid is also a potent antioxidant, and has been extensively used in dermatological products and cosmetics as a preservative (Variati R. et al., 2010; Seifert P. et al., Cosmetic News, 1995).
  • the data regarding the toxicity of usnic acid mainly relate to the systemic use of weight loss preparations containing usnic acid (Guo L. et al., 2008).
  • PCT/EP2009/006960 and novel derivatives of (+)-usnic acid are useful as antibacterial and wound-healing agents and for regenerative or anti-aging treatment of the skin tissues.
  • the compounds according to the present invention are (+)-usnic acid derivatives obtained by nucleophilic addition on the triketone moiety of the molecule by amino groups of natural products, in particular amino acids or decarboxyamino acids, preferably those which contain chemical groups involved in cell-cell interaction, such as redox systems or adhesion mechanisms.
  • the object of the present invention is (+)-usnic acid derivatives having general formula (I):
  • X is selected from OH, SH, COOH and SO 3 H;
  • n and m independently of one another, are an integer from 1 to 10, preferably from 2 to 4; with the proviso that when X is OH or SH, the minimum value of n or m is 2;
  • the compounds of the invention of formula (I) and (II) have low cytotoxicity on the skin cells, high antibacterial activity against infectious or opportunistic pathogens and skin regeneration properties.
  • compositions are preferably administered in the form of semisolid preparations such as creams, salves, ointments or liquids, such as solutions.
  • the dose of active ingredient will depend on the form and administration route, solubility and other pharmacokinetic parameters of the compound.
  • a dose between 0.1 and 1000 mg, preferably between 0.1 and 500 mg, of active ingredient per administration unit is generally acceptable.
  • the daily amount of active ingredient and the duration of the treatment can vary, depending on the severity of the disorder and the characteristics of the patient to be treated.
  • the cells cultured in 12-well plates, were mechanically scratched with the tip of a sterile 0.1- 10 ⁇ , pipette, and then left to re-epithelialise for 24 h at 37°C in the presence of usnic acid (1.7 ⁇ g/mL), ME81 (2.2 ⁇ g/mL), PS8 (2.5 ⁇ g/mL), MB73 (2.1 ⁇ g/mL), and MB56 (2.0 ⁇ g/mL).
  • usnic acid 1.7 ⁇ g/mL
  • ME81 2.2 ⁇ g/mL
  • PS8 2.5 ⁇ g/mL
  • MB73 2.1 ⁇ g/mL
  • MB56 2.0 ⁇ g/mL
  • the crude product was purified by column chromatography (CH 2 Cl 2 /MeOH/EtOH 9:0.5:0.5 to 8:1:1, silica weight 50 g, volume fraction: 30 mL, fractions collected from the 43 th to the 49 th ) and then by reverse phase using Biotage SP1 instrument (SNAP KP-C18-HS 12 g, 35-70 ⁇ , flow rate 15 mL/min, ACN/H 2 O 20:80 to 80:20) to obtain 11 mg (0.019 mmol, 4%) of the desired product as yellow solid.
  • Biotage SP1 instrument SNAP KP-C18-HS 12 g, 35-70 ⁇ , flow rate 15 mL/min, ACN/H 2 O 20:80 to 80:20
  • N-methylmorpholine 15 L, 0.14 mmol
  • 4-(4,6-dimethoxy- 1,3,5- triazin-2-yl)-4-methylmo ⁇ holinium chloride 65 mg, 0.23 mmol
  • 4-Aminobutyric acid (12 mg, 0.12 mmol) was added and the reaction mixture was stirred at room temperature for 3 h and 30 min.
  • the solvent was removed under reduced pressure and the solid obtained was diluted with CH 2 C1 2 and extracted twice with 0.1 ⁇ HCl.
  • the antimicrobial assays were effected with the bacterial strains Staphylococcus aureus (Biogenetics) and Listeria innocua (ATCC 33090). Fresh pure cultures were used in the agar diffusion test. Culture broths maintained for a day in Tryptic Soy Broth (TSB) were seeded evenly on the surface of Muller- Hinton plates according to the Kirby-Bauer method. Usnic acid or derivatives thereof, dissolved in dimethyl sulphoxide (DMSO) (10 ⁇ ,, 500 ⁇ ), was adsorbed on sterile filter paper discs (Whatman No.l, diameter 6 mm) and positioned on the surface of the Muller-Hinton agar plate. DMSO (10 ⁇ ,) and ampicillin sodium salt (AMP) (10 uL, 500 ⁇ ) were used as solvent and control antibacterial respectively. The plates were incubated at 28°C for L. innocua and 37°C for S. aureus.
  • DMSO dimethyl sulphoxide
  • the antimicrobial activities of the products tested were evaluated by measuring the diameter of the clear zone (without bacterial growth) round the discs with the absorbed compound.
  • the antibacterial activity was expressed in millimetres of bacterial growth inhibition zone. All the tests were effected in duplicate. The results are set out in Table 1.
  • MLC Minimum Lethal Concentration
  • the compounds of examples 10 and 2 which exhibited the greatest antimicrobial efficiency in the agar diffusion test of example 16 were selected together with usnic acid to evaluate the Minimum Inhibitory Concentration (MIC) and the Minimum Lethal Concentration (MLC).
  • the MIC and MLC were determined by the broth microdilution method (Cosentino et al., 2003; Elo et al., 2007), Briefly, the culture broths maintained overnight were prepared with Nutrient Broth (NB) for S. aureus and Tryptic Soy Broth (TSB) for L. innocua.
  • Usnic acid and the derivatives thereof were diluted at 12 different concentrations in the interval between 34-10 and 34 ⁇ g/nlL (double serial dilutions).
  • a volume of 10 ih was distributed in each of the two test tubes used (1.5 mL). Each test micro-organism was diluted to 10 5 CFU/ml, and the bacterial suspension was added (490 ⁇ ) to each test tube. The concentration of the bacterial suspension was confirmed by viable count on Tryptic Soy Agar (TSA). The concentrations of the substances tested were equivalent to 1.95-2000 ⁇ in the test tubes of the final tests.
  • TSA Tryptic Soy Agar
  • test tubes were incubated at 37°C for 24 h, and the MIC and MLC were evaluated. Bacterial growth was revealed by the presence of a pellet on the base of the test tube. The MICs were determined on the basis of the first test tube without a visible pellet of bacteria, in increasing order of concentration of the compound. A volume (100 ⁇ ) from each test tube used to evaluate the MIC was suspended in TSA to confirm the MIC values and establish the MLC. The number of surviving bacteria was determined after incubation.
  • the MIC value was taken as the lowest concentration of the compound able to produce a significant reduction in viability (>90%), while the MLC value was taken as the concentration at which a percentage > 99.9% of the initial inoculum was killed.
  • S. aureus suggests that this compound has a bacteriostatic effect, as previously observed (Lauterwein et al. 1995).
  • the compounds of examples 2 and 10 presented lower MLC values than usnic acid, indicating a prevalently bactericidal effect.
  • the MIC values were very similar for all the compounds tested on S. aureus, whereas the compounds of examples 2 and 10 presented lower MIC values on L. innocua.
  • the wound-healing capacity of usnic acid derivatives was tested on an in vitro wound-healing model consisting of HaCaT keratinocyte monolayers. Said cells represent an in vitro model of keratinocyte proliferation and migration.
  • the HaCaT cell line imitates many properties of normal epidermal keratinocytes, is not invasive and can differentiate under suitable experimental conditions (Petrussevska RT, et al. 1988; Schoop VM, et al. 1999).
  • the HaCaT cell line was previously used in studies of the wound-healing process as an in vitro model of the system of re-epithelialisation, a phase typical of the wound-healing process (Matsuura K, et al. 2007; Ranzato, E., et al. 2008).
  • the cells were maintained at 37°C, 5% CO 2 in DMEM culture medium supplemented with 10% foetal bovine serum (FBS, Euroclone, Pero, Italy) and 1% antibiotic mixture.
  • FBS foetal bovine serum
  • the scratch wound test was effected on confluent HaCaT monolayers by selecting the compounds with the most potent antibacterial activity, namely the compounds of examples 10 and 2 (PS8 and ME81 respectively), and those with the lowest toxicity on the keratinocytes, namely the compounds of examples 3 and 7 (ME56 and MB73 respectively).
  • the analysis was effected as described in Ranzato et al. (Ranzato, E., et al. 2009a).
  • the cells were grown to confluency in multiwell plates, and the cell layers were then scratched with the tip of a sterile 0.1-10 ⁇ , pipette. After washing away the cells in suspension, the cultures were incubated again with the medium and exposed to usnic acid and derivatives thereof, used at the concentration of 5 ⁇ for 24 h. Some samples, used as positive control, were exposed to a dose of 20% (v/v) of platelet lysate (PL) which, according to earlier studies, promotes wound- healing in those cells, and was obtained from blood samples as described in Ranzato et al. (Ranzato, E., et al. 2008).
  • PL platelet lysate
  • the cells were fixed with 3.7% formaldehyde in PBS for 30 min, and then stained with 0.1% toluidine blue at room temperature for 30 min (Figure 1, top image).
  • the size of the wound was measured at the time of the lesion and at the end of the treatment, using a Televal inverted microscope (Carl Zeiss Inc., Thornwood, NY) equipped with a digital video camera and NIH ImageJ software. Wound closing was determined as the difference in the size of the wound at 0 and 24 h.
  • the would-closure rate is the difference between the rim distance at zero time and after 24hours. Values are expressed as the percent of the would-closure rate obtained with platelet lysate Example 19
  • mice Male Sprague-Dawley rats (160-180 g) and Swiss albino mice (20-25 g) were supplied by the Saki Yenilli animal breeding laboratory (Ankara, Turkey). The animals were maintained under environmental conditions for 3 days for acclimatisation purposes. During the experiments they received a standard diet of pellets, and unlimited access to water. The study was authorised by the Institutional Animal Ethics Committee and performed in compliance with international legislation governing experiments on animals and biodiversity.
  • test samples were prepared using as base an ointment (carrier) consisting of glycol stearate, propylene glycol and liquid paraffin (3 :6: 1) at the concentration of 1 %. Aliquots of 0.5 g of each ointment to be tested were applied locally to the wound created with a surgical blade.
  • carrier consisting of glycol stearate, propylene glycol and liquid paraffin (3 :6: 1) at the concentration of 1 %.
  • the animals in the carrier group were only treated with the basic ointment, while the animals in the reference medicament group were treated with 0.5 g of Madecassol ® (Bayer, 00001199), which contains 1% of Centella asiatica extract.
  • a total of seven rats per group were anaesthetised with 0.15 cc of Ketalar ® (Shetty et al., 2006), and the dorsal region of the rats was shaved and cleaned with 70% alcohol.
  • Two linear incisions 5 cm long were made in the paravertebral region with a sterile blade on the shaven skin at a distance of 1.5 cm from the median dorsal line, on each side.
  • the wounds were closed with three surgical stitches at 1 cm intervals.
  • the ointments prepared with the test samples, the reference medicament (Madecassol ® ) or the basic ointment [glycol stearate: propylene glycol: liquid paraffin (3:6:1)] were applied locally to the wounds in the dorsal region in each group of animals once a day for 9 days. All the stitches were removed on the last day, and the ultimate tensile strength of the previously damaged skin was measured with a tensiometer (Zwick/Roell Z0.5, Germany) (Lodhi et al., 2006; Suguna et al., 2002).
  • mice The fur on the backs of the mice was shaved, creating a circular wound in the interscapular dorsal region of the animals through excision of the skin with a 5 mm biopsy drill (Nopa instruments, Germany); the wounds were left open (Tramontina et al., 2002).
  • test samples The test samples, the reference medicament (Madecassol ® , Bayer) and the ointments used as carrier were applied once a day until the wound had completely healed.
  • the progressive changes in the wound area were monitored with a camera (Fuji, S20 Pro, Japan) every two days.
  • the wound area was then evaluated with the use of the AutoCAD program.
  • the contraction of the wound was measured as the percentage reduction of the wound area.
  • a tissue sample was taken from the healed skin of each group of mice for histopathological examination (Sadaf et al., 2006).
  • Table 6 shows the contraction values in the development of wound healing on circular excision wound models for the groups treated with the carrier, the negative control, the compounds and the reference medicament.
  • Usnic acid and compounds MB73 and ME81 exhibited potential wound-healing activity, while the carrier and negative control groups did not exhibit statistically significant wound-healing activity.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Dermatology (AREA)
  • Birds (AREA)
  • Oncology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Gerontology & Geriatric Medicine (AREA)
  • Communicable Diseases (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)
  • Agricultural Chemicals And Associated Chemicals (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

Disclosed are dibenzofuran derivatives of usnic acid, compositions thereof and use thereof in dermatological or cosmetic formularions with antimicrobial, regenerative and anti-aging purposes.

Description

DIBENZOFURAN DERIVATIVES WITH ANTIBACTERIAL AND
WOUND-HEALING ACTIVITY
Field of the invention
The present invention relates to dibenzofuran derivatives useful as antibacterial and wound-healing agents and for regenerative or anti-aging treatment of the skin tissues.
Background of the invention
Wound infections represent a significant problem in many post-operative disorders and in burns, and are common in many developing countries due to poor conditions of hygiene. Staphylococcus aureus, Streptococcus pyogenes, Escherichia coli, Pseudomonas aeruginosa, Streptococcus pneumoniae and Klebsiella pneumoniae are some important micro-organisms that cause wound infections (Mertz P. et al., 1993). Said infections are treated with a number of antibiotics which in many cases fail during the treatment due to the onset of resistance and/or toxicity.
Active ingredients for use on the skin in dermatological or cosmetic formulations with regenerative or anti-aging purposes should ideally have low cytotoxicity on the skin cells, high antibacterial activity against infectious or opportunistic pathogens, and good skin regeneration properties. A large number of wound-healing products of natural, chemical or biotechnological origin are currently on the market. However, in most cases these products are complex mixtures used on an empirical basis, while their action mechanisms and active ingredients have not yet been clearly identified. For example, the best results in the regeneration of chronic or serious wounds are currently obtained in clinical practice with platelet derivatives containing various growth factors and a number of low-molecular- weight compounds (Ranzato et al., 2009).
There is consequently an urgent need to identify new chemical entities possessing antibiotic and tissue-remodelling properties, for use, either alone or in formulations with other drugs, in the treatment of topical infections such as infected burns, topical otitis (Aronovitz G.H., 2000), haemorrhoids, vaginal lesions, and infections of the oral cavity.
Usnic acid is a dibenzofuran derivative produced by various species of lichen. Its chemical name is 2,6-diacetyl-7,9-dihydroxy-8,9b- dimethyldibenzofuran-l,3(2H,9bH)-dione, corresponding to the following formula:
Figure imgf000003_0001
Usnic acid
The compound exists in nature in the enantiomeric (+) and (-) forms and as a racemic mixture. (+) Usnic acid is commercially available, being easily extracted from lichens of species belonging to the genera Usnea and Ramalina, wherein it is present in percentages exceeding 26% (Reyim M. et al., 2010).
The antibacterial, antifungal, antiviral and anti-inflammatory properties of usnic acid are known, as are its consequent uses in the cosmetic and pharmaceutical fields, in particular as an active ingredient of creams, deodorants, toothpastes, mouthwashes, antibiotic ointments, vaginal creams, foot creams, powders and shampoos, or as a diet supplement. In Argentina, Usnea densirotra tea is known as "Barba della Piedra", and is used for gargling. In these preparations, usnic acid is used as both active ingredient and preservative.
The use of usnic acid in combinatorial synthesis has been described (Tomasi et al., 2006). Other usnic acid derivatives are described in GB 800114.
PCT/EP2009/006960 discloses (+) usnic acid derivatives obtained by conjugating (+) usnic acid with biologically active molecules. The compounds are useful for the prophylaxis and treatment of infections caused by protozoa of the genus Plasmodium.
The potential wound-healing activity of (+)-usnic acid, due to its properties of stimulating the wound closure of human HaCaT keratinocyte monolayers at subtoxic doses, was recently described. The action mechanism was correlated with its ability to promote cell motility (Burlando B. et al., 2009). Earlier studies had indicated (+)-usnic acid sodium salt as a promoter of the wound-healing process, due to the secretion of growth factors and the increased rate of cell migration to a dose-dependent extent (Burlando B. et al., 2009). Experiments effected in vivo had confirmed the ability of usnic acid sodium salt to accelerate healing of the epidermis, but this activity was not attributed to stimulation of proliferation (Jin J. et al., 2005). Recent publications report the use of formulations based on usnic acid conveyed by liposomes to heal burn wounds (Nunes P.S. et al., 2011).
Usnic acid is known to has a number of properties which make it useful in the treatment of topical lesions. Lichens belonging to the genus that contains usnic acid have been used all over the world as remedies for systemic and topical use since ancient times.
Usnic acid is universally known as an antibiotic. (+)- and (-) usnic acids are active on sensitive and resistant strains of Gram-positive bacteria (see the review by Ingolfsdottir K., 2002). Some metal complexes of usnic acid hydrazones have proved more active (Natic M. et al, 2004). Kokubun T. et al., 2007 reports the antibacterial activity of (+) usnic acid on MRSA (methicillin-resistant S. aureus) and MDR (multidrug-resistant) strains in addition to mupirocin-resistant strains.
All the results obtained to date confirm a bacteriostatic rather than bactericidal action.
Usnic acid is also a potent antioxidant, and has been extensively used in dermatological products and cosmetics as a preservative (Variati R. et al., 2010; Seifert P. et al., Cosmetic News, 1995). The data regarding the toxicity of usnic acid mainly relate to the systemic use of weight loss preparations containing usnic acid (Guo L. et al., 2008).
Toxicity studies effected at the National Center for Toxicological Research (NCR), USA, have demonstrated that preparations based on Usnea lichen containing equivalent concentrations of usnic acid produce greater toxicity than pure usnic acid.
It has been reported that the contact dermatitis and allergenicity of usnic acid are modest, mainly due to the (-) enantiomer (Hasten B.M. et al., 1993), and probably correlated with its general toxicity.
The lipophilia of usnic acid, together with its systemic toxicity, is a characteristic which has limited its use. Usnic acid presents high lipophilia due to the intramolecular hydrogen bonds, which probably contributes to its toxicity, making the molecule permeable to the membranes. The problem of its very low solubility in water has been overcome in cosmetics by using complexes with liposomes or other formulations. (+) usnic acid is therefore a good candidate for investigation of the factors involved in its biological activity: it is easily obtainable and has recognised, proven activities in vitro, but presents limitations on use due to its toxicity in vivo (Guo L. et al., 2008).
Description of the invention
It has now been found that some (+)-usnic acid derivatives disclosed in
PCT/EP2009/006960 and novel derivatives of (+)-usnic acid are useful as antibacterial and wound-healing agents and for regenerative or anti-aging treatment of the skin tissues.
The compounds according to the present invention are (+)-usnic acid derivatives obtained by nucleophilic addition on the triketone moiety of the molecule by amino groups of natural products, in particular amino acids or decarboxyamino acids, preferably those which contain chemical groups involved in cell-cell interaction, such as redox systems or adhesion mechanisms. The object of the present invention is (+)-usnic acid derivatives having general formula (I):
Figure imgf000006_0001
(I)
wherein:
represents a single or double bond;
R is a residue selected from:
- H
- (C¾)nX
- (CH2)nC(O)NH(CH2)mX
Figure imgf000006_0002
Figure imgf000006_0003
Figure imgf000007_0001
wherein the symbol 1 represents the attachment point of the R group to the N(H) group;
X is selected from OH, SH, COOH and SO3H;
n and m, independently of one another, are an integer from 1 to 10, preferably from 2 to 4; with the proviso that when X is OH or SH, the minimum value of n or m is 2;
their enantiomers and pharmaceutically and cosmeceutically acceptable salts, for use as antibacterial and wound-healing agents and for regenerative or anti-aging treatment of skin tissues.
In one embodiment of the invention, in the compounds of formula (I) the R group is a residue selected from:
- H
- (CH2)2SH
- (CH2)2SO3H CH2COOH
(CH2)3COOH
(CH2)4OH
(CH2)3C(0)NH(CH2)3COOH
Figure imgf000008_0001
Figure imgf000009_0001
In a preferred embodiment, in the compounds of formula (I), R is (CH2)2SO3H.
In another embodiment the present invention provides novel compounds of general formula (II)
Figure imgf000009_0002
(II)
wherein:
represents a single or double bond;
R' is a residue selected from:
- (CH2)nX'
- (CH2)nC(0)NH(CH2)mX
Figure imgf000009_0003
Figure imgf000010_0001
wherein the symbol 1 represents the attachment point of the R' group to the N(H) group;
X is selected from OH, SH, COOH and SO3H;
X' is selected from OH, SH and COOH;
n and m, independently of one another, are an integer from 1 to 10, preferably from 2 to 4; with the following provisos:
when X or X' is OH or SH, the minimum value of n or m is 2;
when X' is COOH, n is different from 1 or 2;
when X' is OH, n is different from 4;
their enantiomers and pharmaceutically and cosmeceutically acceptable salts.
Salts of compounds (I) and (II) bearing acidic functions such as free carboxyl or sulfonyl groups, with Ag , Zn , Na , Mg and Ca are preferred.
In one embodiment of the invention, the R' group is selected from the group of:
H
- (CH2)2SH
CH2COOH
(CH2)3COOH
- (CH2)4OH
(CH2)3C(O)NH(CH2)3COOH
Figure imgf000011_0001
The compounds of the invention of formula (I) and (II) have better antibacterial activity and wound-healing properties than usnic acid. In many cases they also present an overall increase in solubility in water compared with this compound.
The antibacterial activity can be further improved by:
(i) conjugating the compounds (I) or (II) to antibiotic molecules. The conjugation involves suitable moieties present in the usnic acid scaffold of compounds (I) or (II), particularly the R and R' groups.
(ii) metal complexation with Zn, Cu, Fe, Au of the compounds of general formula I and II. The formation of complexes involves the enol system and the nitrogen present in the usnic acid scaffold of compounds (I) or (II) .
A further object of the invention is represented by the compounds of formula (II) for use as medicaments, in particular as antibacterial and wound- healing agents and for regenerative or anti-aging treatment of skin tissues.
The compounds of the invention of formula (I) or (II) can be obtained by reaction between a molar equivalent of (+)-usnic acid and an amount of 1 to 3 molar equivalents of an amine of formula R-NH2 (III) or R' NH2, (IV), wherein R, R' are as defined above.
The compounds of the invention wherein R or R' are the residue
Figure imgf000012_0001
can also be prepared by reacting (+)-usnic acid with a diamine of formula
H2N-(CH2)n-S-S-(CH2)n-NH2 (V). In this case, 0.5 molar equivalents of diamine (V) per 1 molar equivalent of (+)-usnic acid are used.
The reaction is generally effected in a solvent such as ethanol, methanol, methylene chloride or mixtures thereof, in the presence of an organic base such as triethylamine or pyridine, or an inorganic base such as potassium hydroxide. The reaction is carried out at the reflux temperature of the solvent mixture, for a time generally ranging from a few hours to a few days. The reaction is preferably carried out with anhydrous reagents and solvents, in an inert gas atmosphere.
The compounds of the invention of formula (I) and (II) have low cytotoxicity on the skin cells, high antibacterial activity against infectious or opportunistic pathogens and skin regeneration properties.
The compounds of the invention of formula (I) and (II) have inhibitory activity against Gram-positive bacteria, and in particular have proved effective against strains of S. aureus and L. innocua. Unlike usnic acid, which exhibits a prevalently bacteriostatic effect, the compounds of the invention surprisingly exhibit a bactericidal effect.
In tests effected in vitro on human keratinocytes, the compounds of the invention proved significantly less toxic than usnic acid.
In tests effected in vitro and in vivo, the compounds of the invention proved to have greater wound-healing capacity than usnic acid. In particular, the compounds of the invention are able to promote the reorganisation of a layer of keratinocytes in a way that recalls the epithelial-mesenchymal transition process, similarly to the process functionally described for a platelet lysate (Ranzato et al., 2008).
The compounds of the invention are therefore useful in preparations that stimulate the regeneration of skin wounds or in cosmetic preparations with an anti- aging effect.
Further objects of the present invention are pharmaceutical compositions for topical use or cosmetic preparations containing a compound of formula (I) or formula (II) as defined above, together with compatible excipients for pharmaceutical or cosmeceutical use.
The compositions are preferably administered in the form of semisolid preparations such as creams, salves, ointments or liquids, such as solutions. The dose of active ingredient will depend on the form and administration route, solubility and other pharmacokinetic parameters of the compound. A dose between 0.1 and 1000 mg, preferably between 0.1 and 500 mg, of active ingredient per administration unit is generally acceptable. The daily amount of active ingredient and the duration of the treatment can vary, depending on the severity of the disorder and the characteristics of the patient to be treated.
The pharmaceutical compositions may contain additional active ingredients besides compounds (I) or (II), particularly the antibiotics gentamycin, erythromycin, clindamycin and oxacillin, which have been found to interact positively with usnic acid (Segatore B, Bellio P, Setacci D, Brisdelli F, Piovano M, Garbarino JA, Nicoletti M, Amicosante G, Perilli M, Celenza G. "In vitro interaction of usnic acid in combination with antimicrobial agents against methicillin-resistant Staphylococcus aureus clinical isolates determined by FICI and ΔΕ model methods", Phytomedicine. 2012 Feb 15;19(3-4):341-7. doi: 10.1016/j.phymed.2011.10.012. Epub 2011 Nov 25).
Another aspect of the invention are compounds of formula (I) or (II) as defined above or pharmaceutical or cosmetic compositions containing them, for use in regenerative or anti-aging treatment of skin tissues, in particular for treatment of topical infections such as infected burns, topical otitis, haemorrhoids, vaginal lesions, and infections of the oral cavity.
The invention will now be illustrated by the following examples.
Description of Figure
Regeneration of scratch wounds of confluent HaCaT monolayers. The cells, cultured in 12-well plates, were mechanically scratched with the tip of a sterile 0.1- 10 μΐ, pipette, and then left to re-epithelialise for 24 h at 37°C in the presence of usnic acid (1.7 μg/mL), ME81 (2.2 μg/mL), PS8 (2.5 μg/mL), MB73 (2.1 μg/mL), and MB56 (2.0 μg/mL). One sample was exposed to 20% platelet lysate (PL) as positive control.
Top panel. Representative image showing HaCaT monolayers subjected to a scratch wound, incubated under control conditions (A) or in the presence of MB56 (B), and then stained with toluidine blue and observed 24 h after the wound. Scale = 200 μπι.
Bottom panel. Chart showing wound-healing measurements, expressed as the difference between the size of the wound after 0 and 6 h. The bars represent the mean±SD of two independent experiments, each with n=20. The mean of the controls was set at 100%. Different letters on the bars indicate groups significantly different from one another according to the Tukey test (p < 0.01).
Examples
Example 1
Synthesis of the conjugate with arginine (PS2)
Figure imgf000015_0001
202 mg (1.16 mmol) of L-arginine was added to a suspension of (+)-usnic acid (400 mg, 1.16 mmol) in absolute EtOH (15 mL) and re fluxed under N2 for 5 h, then left under stirring at room temperature for another 15 h. The solid obtained by concentration under vacuum was crystallised from diisopropyl ether/EtOH 9:1, affording 0.516 g (89%) of a pale yellow solid. M.p.: 232-234°C.
1H NMR 400 MHz (DMSO-<¾ δ 1.58 (5H, m, CH3-13, CH2-4') 1.81 (2H, m, CH2-3') 1.92 (3H, s, CH3-16) 2.57 (3H, s, CH3-15) 2.57 (3H, s, CH3-18) 3.13 (2H, m, CH2-5') 4.26 (1H, m, CH2-2'), 5.69 (1H, s, CH-4), 7.62 (3H, br s), 8.89 (1H, br s) 12.43 (1H, br s, OH-10) 13.28 (1H, br s, OH-8) 13.35 (1H, br s, NH).
13C NMR 400 MHz (DMSO-i¾ δ 7.81 (C-16), 19.36 (C-15), 25.14 (C-4'), 30.45 (C-3'), 31.39 (C-18), 32.48 (C-13), 56.68 (C-12), 56.68 (C-5'), 58.87 (C-2'), 101.19 (C-7), 101.97 (C-2), 102.90 (C-4), 105.56 (C-l l), 106.81 (C-9), 156.09 (C- 6), 157.68 (C-6'), 158.08 (C-10), 163.08 (C-8), 172.54 (C-5), 172.77 (C-1'), 173.03 (C-14), 188.2 (C-3), 197.38 (C-1), 200.95 (C-17).
HRMS (ESI), (positive) m/z 501.1 [M+H]+, calculated for C24H28N4O8, 500.19.
Example 2
Synthesis of conjugate with taurine (ME81)
Figure imgf000016_0001
375 mg (3 mmol) of taurine was dissolved in aqueous EtOH (10 mL, 1:1), KOH was added to pH~9-10, and the reaction mixture was refluxed for 10 min on a water bath. A suspension of (+)-usnic acid (344 mg, 1 mmol) in absolute EtOH (5 mL) was then added in portions over 30 min, and the mixture was refluxed for 1 day. The reaction mixture was concentrated under low pressure. The crude product was then diluted with CH2C12 and washed with IN HC1 to pH 4. The aqueous phases were combined, and the solid obtained after concentration under low pressure was washed with a MeOH/H2O 9:1 solution. After removal of the taurine in suspension by filtration, the filtered solution was concentrated under low pressure, and the desired solid was crystallised from diisopropyl ether/EtOH 9:1, affording 50 mg (11%) of a brown compound.
1H NMR 400 MHz (DMSO-< ) δ 1.65 (3H, s, CH3-13), 1.98 (3H, s, CH3-16), 2.60 (3H, s, CH3-15), 2.65 (3H, s, CH3-18), 2.79 (2H, t, J = 6.4 Hz, CH2-1'), 3.80 (2H, m, CH2-2'), 5.86 (1H, s, H-4), 7.70 (1H, br s, SO3H), 12.43 (1H, s, OH-10), 12.92 (1H, br s, OH-8), 13.38 (1H, s, NH).
13C NMR 400 MHz (DMSO-< ) δ 8.0 (C-16), 9.15, 18.5 (C-15), 31.5 (C-18), 32.1 (C-13), 40.7 (CH2), 50.0 (CH2), 56.6 (C-12), 101.3 (C-7), 102.2 (C-2), 102.9 (C-4), 105.7 (C-l l), 106.7 (C-9), 156.4 (C-6), 158.3 (C-10), 162.3 (C-8), 173.3 (C- 5), 175.1 (C-14), 188.7 (C-3), 197.3 (C-l), 201.4 (C-17).
HRMS (ESI), (negative) m/z 450.1 [M-H]\ calculated for C20H19NO9S,451.1.
Example 3
Synthesis of conjugate with cysteamine (MB56)
Procedure A.
Figure imgf000017_0001
A solution of cysteamine (224 mg, 2.90 mmol) in 6 mL of anhydrous MeOH was added under N2 to a suspension of (+)-usnic acid (1 g, 2.90 mmol) in 6 mL of anhydrous CH2C12 and 30 mL of anhydrous MeOH. The reaction mixture was refluxed at 65°C for 4 h, and then at room temperature overnight and again at 65°C for 2 h. The solvent was removed under low pressure and the solid obtained was purified by flash-chromatography (toluene/EtOAc 8:2) until the desired product was obtained as a yellow solid (511 mg, 1.27 mmol, 44%). M.p.: 142- 143°C.
1H-NMR 400 MHz (CDC13) δ ppm 1.63 (1H, t, J = 8.6 Hz, SH), 1.71 (3H, CH3-13), 2.10 (3H, s, CH3-16), 2.65 (3H, s, CH3-15), 2.68 (3H, s, CH3-18), 2.86 (2H, m, CH2-2'), 3.71 (2H, m, CH2-1'), 5.80 (1H, s, CH-4), 11.91 (1H, br s, OHIO), 13.36 (1H, s, OH-8), 13.74 (1H, br s, NH).
When D2O was added, the signal at 2.86 ppm became a triplet (J = 6.4 Hz), and the triplet at 1.63 ppm disappeared.
13C-NMR 100 MHz (CDC13) 5 ppm 8.16 (C-16), 19.13 (C-15), 24.46 (C-2'), 31.95 (C-18), 32.63 (C-13), 47.38 (C-l'), 57.95 (C-12), 102.06 (C-2 + C-7), 103.00 (C-4), 105.67 (C-l l), 108.69 (C-9), 156.52 (C-6), 158.88 (C-10), 164.17 (C-8), 174.97 (C-5), 175.65 (C-14), 191.54 (C-3), 199.14
(C-l), 201.33 (C-17).
LC-MS (ESI), positive, m/z 403.9 [M + H]+' 425.8 [M + Naf, calculated for C20H21NO6S 403.1. Procedure B.
Triphenylphosphine (1.310 g, 4.99 mmol) and 18 mL of water under helium were added to a solution of MB90 of example 4 (2.680 g, 3.33 mmol) in 120 mL of acetone. The reaction mixture was left under stirring at room temperature for 4 days. The solvent was removed under low pressure, and the solid obtained was purified by flash-chromatography (toluene/EtOAc 9.5:0.5) until the desired product was obtained as a yellow solid (1.233 g, 3.056 mmol, 92%).
Example 4
Synthesis of dimer conjugate with cysteamine (MB90)
Figure imgf000018_0001
24 μΐ of anhydrous pyridine was added to a solution of cystamine dihydrochloride (65 mg, 0.29 mmol) in 3 mL of absolute EtOH. After 30 min stirring at room temperature, a suspension of usnic acid (200 mg, 0.58 mmol) in 3 mL of absolute EtOH was added, and pyridine was then added until completely dissolved. The reaction mixture was refluxed under nitrogen for 8 h. The solvent was evaporated under low pressure, and the solid obtained was acidified with IN HC1 to pH 4, and then extracted twice with EtOAc. The organic phases were combined and concentrated under low pressure, affording 173.2 mg of a pale yellow solid with a quantitative yield.
1H-NM 400 MHz (CDC13) δ ppm 1.68 (3H, CH3-13), 2.07 (3H, s, CH3- 16), 2.64 (3H, s, CH3-15), 2.66 (3H, s, CH3-18), 3.00 (2H, t, J = 6.5 Hz, CH2-2'), 3.87 (2H, m, CH2-1'), 5.76 (1H, s, CH-4), 11.81 (1H, br s, OH- 10), 13.33 (1H, s, OH-8), 13.72 (1H, br s, NH). 1JC-NMR 100 MHz (CDC13) δ ppm 8.18 (C- 16), 19.14 (C-15), 31.94 (C- 18), 32.66 (C-13), 37.26 (C-2'), 42.98 (C-l '), 58.14 (C-12), 102.04 (C-2 + C-7), 102.92 (C-4), 105.58 (C-l l), 108.73 (C-9), 156.48 (C-6), 158.80 (C-10), 164.22 (C-8), 174.97 (C-5), 175.69 (C-14), 191.63 (C-3), 199.20 (C-l), 201.30 (C-17).
LC-MS (ESI), positive, m/z 805.2 [M + H]+, calculated for C4oH4oN2O12S2
804.2.
Example 5
Synthesis of conjugate with asparagine (PS5)
Figure imgf000019_0001
79 mg (0.60 mmol) of L-asparagine was added to a suspension of (+)-usnic acid (207 mg 0.60 mmol) in absolute EtOH (15 mL). The reaction mixture was refluxed under N2 for 4 h, and then at room temperature for a further 15 h. The solid obtained by concentration under vacuum was crystallised from diisopropyl ether/EtOH 9: 1, giving 217 g of a yellow solid (79%). Mp: 221-223°C.
1H NMR 400 MHz (DMSO-<¾ S ppm 1.65 (3H, s, CH3-13), 1.96 (3H, s, CH3-16), 2.72 (3H, s, CH3-15) 2.73 (3H, s, CH3-I8), 2.76 (1H, m, CH2-3 '), 2.88 (1H, m, CH2-3'), 4.98 (1H, m, CH-2'), 5.84 (1H, s, CH-4), 12.21 (1H, s, OH), 13.38 (2H, m, OH, NH).
13C NMR 400 MHz (DMSO-^): S ppm 7.8 (C-16), 19.0 (C-15), 30.9 (C- 18), 32.1 (C-13), 37.6 (C-3'), 53.4 (C-2'), 56.74 (C-12), 101.3 (C-7), 102.3 (C-2), 102.8 (C-4), 105.63 (C-l l), 106.64 (C-9), 156.2 (C-6), 158.2 (C-10), 163.1 (C-8), 171.2 (C-l ', C-4'), 173.0 (C-5), 174.7 (C-14), 189.2 (C-3), 198.0 (C-l), 201.3 (C- 17). HRMS (ESI), negative, m/z 457.12 [M - H]", calculated for C22H22N2O9, 458.13.
Example 6
Synthesis of conjugate with glycine (PS12)
Figure imgf000020_0001
145 mg (1.16 mmol) of L-arginine was added to a suspension of (+)-usnic acid (400 mg, 1.16 mmol), triethylamine (324 μΕ, 2.32 mmol) in water (3 mL) and absolute EtOH (15 mL); the reaction mixture was refluxed under nitrogen for 4 h, then left under stirring at room temperature for another 15 h. The solid obtained by concentration under vacuum was crystallised from diisopropyl ether/EtOH 9:1, affording 375 g (78%) of a yellow solid. Mp: 210-212°C.
1H NMR 400 MHz (DMSO- 5) δ ppm 1.16 (9H, t, J= 7.2 Hz, CH3-3'), 1.62 (3H, s, CH3-13), 1.94 (3H, s, CH3-16), 2.54 (3H, s, CH3-15), 2.61 (3H, s, CH3-18), 3.01 (6H, q, J= 7.2 Hz, CH2-4'), 4.06 (2H, m, CH2-1'), 5.81 (1H, s, CH-4), 12.43 (1H, br s, OH), 12.94 (1H, s, OH), 13.38 (1H, br s, NH).
13C NMR 400 MHz (DMSO- ) δ ppm 7.9 (C-16), 8.9 (C-3'), 19.7 (C-15), 31.4 (C-18), 32.23 (C-13), 45.49 (C-4'), 47.9 (C-1'), 56.5 (C-12), 101.2 (C-7), 102.1 (C-2), 103.0 (C-4), 105.7 (C-l l), 106.7 (C-9), 156.3 (C-6), 158.2 (C-10), 162.9 (C-8), 169.6 (C-2'), 172.9 (C-5), 173.6 (C-14), 188.4 (C-3), 197.6 (C-1), 201.3 (C-17).
HRMS (ESI), negative, m/z 400.10 [M - H]", calculated for C20H19NO8,
401.11.
Elemental analysis; C 57.75%, H 6.75%, N 5.41%, calculated for C20H19NO8 x Et3N x H2O, C 59.99%, H 6.97%, N 5.38%.
Example 7
Synthesis of conjugate with 4-aminobutyric acid (MB 73)
Figure imgf000021_0001
4-aminobutyric acid (1.348 g, 13.07 mmol) was added to a solution of (+)- usnic acid (5.0 g, 14.52 mmol) in 150 mL of 96% EtOH (dried on molecular sieves) and 10 mL of triethylamine, and the reaction mixture was stirred under nitrogen at 80°C for 3 h, and then at room temperature overnight and again at 80°C for 5 h.
The solvent was removed under low pressure and the solid obtained was taken up with CH2C12 and extracted with 0.1N HC1. The aqueous phase was extracted again with CH2C12, and the combined organic phases were washed with a NaCl saturated solution, dried on Na2SO4, filtered and evaporated under low pressure. The solid obtained was suspended in diisopropyl ether/96% EtOH 9:1 (50 mL) and refluxed for 15 min. The suspension was filtered, and 3.878 g (9.03 mmol, 69%) of a yellow solid was obtained. M.p.: 202-205°C.
1H-NMR 400 MHz (DMSO-d6) δ ppm 1.66 (3H, s, CH3-13), 1.86 (2H, q, J = 7.2 Hz, CH2-2'), 1.98 (3H, s, CH3-16), 2.36 (2H, t, J = 7.2 Hz, CH2-3'), 2.59 (3H, s, CH3-15), 2.65 (3H, s, CH3-18), 3.59 (2H, m, CH2-1'), 5.89 (1H, s, CH-4), 12.23 (1H, br s, COOH), 12.34 (1H, s, OH-10), 13.05 (1H, t, J = 5.2 Hz, NH), 13.42 (1H, s, OH-8).
13C-NMR 100 MHz (DMSO-d6) δ ppm 7.48 (C-16), 18.08 (C-15), 23.97 (C- 2'), 30.66 (C-3'), 30.98 (C-18), 31.73 (C-13), 42.72 (C-l'), 56.30 (C-12), 100.81 (C-7), 101.60 (C-2), 102.28 (C-4), 105.10 (C-l l), 106.30 (C-9), 155.69 (C-6), 157.66 (C-10), 162.52 (C-8), 172.92 (C-5), 173.69 (COOH), 174.98 (C-14), 188.95 (C-3), 197.23 (C-l), 200.81 (C-17).
Example 8
Synthesis of conjugate with 4-aminobutanol (BT20)
Figure imgf000022_0001
A solution of 4-amino-l-butanol (800 iL, 714 mg, 8.68 mmol) in 4 mL of EtOH was added to a suspension of (+)-usnic acid (3.0 g, 8.71 mmol) in 100 mL of 96% EtOH (dried on molecular sieves). The reaction mixture was stirred at 80°C for 5 h under N2. The solvent was then removed under low pressure, and the crude product obtained was purified with a chromatography column (CH2Cl2/MeOH 98:2, weight of silica: 180 g, volume of fractions: 60 mL, fractions collected: 28 to 38), the desired product being obtained as a pale yellow solid (2.4 mg, 5.78 mmol, 66%). M.p.: 76°C. K +348.4 (c 0.40, CHC13).
1H-NMR 400 MHz (CDC13) δ ppm 1.52 (1H, m, OH), 1.73 (3H, s, CH3-13), 1.71-1.78 (2H, m, CH2-3'), 1.85-1.91 (2H, m, CH2-2'), 2.12 (3H, s, CH3-16), 2.66 (3H, s, CH3-15), 2.70 (3H, s, CH3-18), 3.57 (2H, m, CH2-1'), 3.77 (2H, dd, J = 11.0, 5.8), 5.80 (1H, s, CH-4), 11.99 (1H, s, OH-10), 13.38 (1H, s, OH-8 and 1H, s, ΝΉ).
When D2O was added, the signal at 3.77 ppm became a triplet (J = 6.0 Hz), and the triplet at 1.52 disappeared.
13C-NMR 100 MHz (CDC13) δ ppm 8.13 (C-16), 18.99 (C-15), 26.34 (C-3'), 30.27 (C-2'). 31.90 (C-18), 32.68 (C-13), 44.50 (C-4'), 57.72 (C-12), 6102.88 (C- 2), 103.10 (C-4), 102.62 (C-l'), 102.02 (C-7), 105.76 (C-l l), 108.62 (C-9), 156.55 (C-6), 158.94 (C-10), 164.13 (C-8), 174.74 (C-5), 175.54 (C-14), 190.96 (C-3), 198.88 (C-l), 201.34 (C-17). MS (EI) 415, calculated for C22H25N07415.
Example 9
Synthesis of conjugate with L-histidine (PS6)
Figure imgf000023_0001
0.205 g (0.85 mmol) of L-histidine was added to a suspension of (+)-usnic acid (0.296 g, 0.85 mmol), triethylamine (236 μΐ,, 1.7 mmol) in water (3 mL) and absolute EtOH. The reaction mixture was refluxed under nitrogen for 4 h, and then at room temperature for a further 15 h. The solid obtained by concentration under vacuum was crystallised from diisopropyl ether/EtOH 9:1 affording 0.322 g (78%) of a yellow solid. M.p.: 209-211°C.
1H NMR 400 MHz (DMSO- ) δ ppm 1.16 (9H, t, J= 8.2 Hz, CH3-7'), 1.61 (3H, s, CH3-13), 1.94 (3H, s, CH3-16), 2.50 (3H, s, CH3-15), 2.61 (3H, s, CH3-18), 2.95-3.06 (7H, m, CH2-8\ CH2-3'), 3.18 (1H, m, CH2-3'), 4.60 (1H, m, CH-2'), 5.80 (1H, s, CH-4), 6.83 (1H, s, CH-5'), 7.69 (1H, d, J = 0.8 Hz, CH-6'), 12.39 (1H, br s, OH), 13.25 (1H, br s, OH) 13.38 (1H, br s, NH).
13C NMR 400 MHz (DMSO- δ ppm 8.0 (C-16), 8.9 (C-7'), 18.9 (C-15), 31.3 (C-3'), 31.4 (C-18) 32.1 (C-13), 45.6 (C-8'), 56.5 (C-12), 58.8 (C-2'), 101.2 (C-7), 102.0 (C-2), 102.9 (C-4), 105.6 (C-l l), 106.6 (C-9), 118.0 (C-5'), 132.8 (C- 4'), 135.3 (C-6' 156.2 (C-6), 158.2 (C-10), 162.9 (C-8), 171.8 (C-Γ), 173.0 (C- 5), 173.9 (C-14), 188.9 (C-3), 197.9 (C-l), 201.19 (C-17). HRMS (ESI), positive, m/z 504.1 [M + Na]+calculated for C24H23N3O8, 481.15.
Elemental analysis; C 58.73%, H 6.41%, N 8.80%, calculated for C24H23N3O8 x 2H2O C 58.24%, H 6.84%, N 9.06%. Example 10
Synthesis of conjugate with L-tyrosine (PS8)
Figure imgf000024_0001
0.157 g (0.87 mmol) of L-tyrosine was added to a suspension of (+)-usnic acid (0.300 g, 0.87 mmol) in absolute EtOH (10 mL) and water (3 mL). The reaction mixture was refluxed under N2 for 6 h, and then at room temperature for a further 15 h. The solid obtained by concentration under vacuum was crystallised from diisopropyl ether/EtOH 9:1, affording 0.406 g of a yellow solid (92%). M.p.: 183-185°C.
1H NMR 400 MHz (DMSO-c , δ ppm 1.63 (3H, s, CH3-13), 1.96 (3H, s, CH3-16), 2.36 (3H, s, CH3-15), 2.62 (3H, s, CH3-18), 3.01 (1H, dd, J= 7.2, 14 Hz, H-3'), 3.15 (1H, dd, J= 4.8, 14 Hz, H-3'), 4.95 (1H, m, CH-2'), 5.85 (1H, s, CH- 4), 6.65 (2H, d, J = 8.4 Hz, CH-5', CH-9'), 6.97 (2H, d, J = 8.4 Hz, CH-6', CH- 8'), 12.11 (1H, s, OH), 13.28 (2H, m, OH, NH).
13C M 400 MHz (DMSO- ) δ ppm 7.7 (C-16), 18.9 (C-15), 31.4 (C-18), 32.0 (C-13), 37.9 (C-3'), 57.1 (C-12), 58.4 (C-2'), 101.2 (C-7), 102.4 (C-2), 103.0 (C-4), 105.6 (C-l l), 107.2 (C-9), 116.0 (C-6\ C-8'), 126.1 (C-4'), 131.3 (C-5', C- 9'), 157.2 (C-7'), 157.3(C-6), 158.60 (C-10), 163.0 (C-8), 171.5 (C-l '), 173.4 (C- 5), 174.9 (C-14), 189.2 (C-3), 198.0 (C-l), 201.4 (C-17).
HRMS (ESI), negative, m/z 506.14 [M - H]\ calculated for C27H25NO9, 507.15.
Example 11
Synthesis of conjugate with tryptamine (PS9)
Figure imgf000025_0001
0.139 g (0.87 mmol) of tryptamine was added to a suspension of (+)-usnic acid (0.300 g, 0.87 mmol) in absolute EtOH (10 mL). The reaction mixture was refluxed for 5 h, and then at room temperature for another 16 h. The solid obtained after concentration under vacuum was crystallised from diisopropyl ether/EtOH 9:1, affording 0.393 g (93%) of a yellow solid. M.p.: 218-220°C.
1H NMR 400 MHz (CDC13) δ ppm 1.69 (3H, s, CH3-13), 2.12 (3H, s, CH3- 16), 2.55 (3H, s, CH3-15), 2.69 (3H, s, CH3-18), 3.22 (2H, m, CH2-10'), 3.82 (2H, m, CH2-11 '), 5.78 (1H, s, CH 4), 7.17-7.24 (3H, m, CH-5', CH-6', CH-2'), 7.41 (1H, d, J = 8.0 Hz, CH-7'), 7.60 (1H, d, J= 7.8 Hz, CH-4'), 8.38 (1H, s, NH-Ar), 11.98 (1H, br s, OH), 13.36 (2H, m, NH, OH).
13C NMR 400 MHz (CDC13) δ ppm 7.4 (C-16), 18.5 (C-15), 25.1 (C-10'), 31.1 (C-18), 32.1 (C-13), 44.46 (C-Ι Γ), 101.26 (C-7), 102.4 (C-4), 105.16 (C-2), 107.94 (C-l l), 110.7 (C-9), 110.9 (C-3'), 111.6 (C-7'), 118.1 (C-6'), 119.7 (C-4'), 122.4 (C-5' 123.0 (C-2'), 126.7 (C-3'a), 136.4 (C-7'a), 155.7 (C-6), 158.5 (C- 10), 163.7 (C-8), 174.9 (C-5), 175.0 (C-14), 188.8(C-3), 198.2 (C-l), 200.7 (C-17).
HRMS (ESI), negative, m/z 485.17 [M - H]~, calculated for C28H26N2O6, 486.18.
Example 12
Synthesis of conjugate with ammonia (BT51)
Figure imgf000025_0002
15 0.4 mL of concentrated ammonium hydroxide was added to a suspension of (+)-usnic acid (300 mg, 0.87 mmol) in 3 mL of absolute ethanol, and the reaction mixture was heated at 80°C under N2 for 2 h. After cooling in an ice bath, the yellow solution was concentrated to about a third of the original solution, acidified with IN HC1 and extracted twice with ethyl acetate. The combined organic phases were washed with water and a NaCl saturated solution, dried on Na2SO4, filtered and evaporated. The crude product was then crystallised from chloroform to obtain 230 mg (0.67%, 77%) of a pale yellow solid.
1H-NMR 400 MHz (DMSO-<¾ δ ppm 1.64 (3H, s, CH5-13), 1.98 (3Η, s, CH3-16), 2.53 (3Η, s, CH 15), 2.65 (3Η, s, CH3-18), 5.87 (1Η, s, CH-4), 9.83 (1Η, br s, NH), 11.54 (1Η, br s, NH), 12.32 (1Η, s, OH-10), 13.42 (1Η, s, OH-8). When D2O was added, the signals at 9.38 ppm, 11.54 ppm, 12.32 ppm and 13.42 ppm disappeared.
13C-NMR 100 MHz (OMSO-d6) 6 ppm 8.60 (C-16), 25.63 (C-15), 32.11 (C- 18), 32.79 (C-13), 57.17 (C-12), 101.95 (C-7), 102.36 (C-2), 103.64 (C-4), 106.21 (C-l l), 107.40 (C-9), 156.77 (C-6), 158.77 (C-10), 163.62 (C-8), 174.07 (C-5), 176.94 (C-14), 189.59 (C-3), 198.69 (C-l), 201.94 (C-17).
MS (EI), positive, mlz found: 343 [Μ+'], calculated for C18H17NO6: 343.
Example 13
Synthesis of (i?)-2-acetamido-3-((4-(((^)-l-((if)-6-acetyl-7,9-dihydroxy-
8,9b-dimethy 1- 1 ,3-dioxo- 1 ,9b-dihy drodibenzo [b,d\ f uran-2 (3H)- ylidene)ethyl)amino)butanoyl)thio)propanoic acid (BT100).
Figure imgf000026_0001
In an oven dried round bottomed flask, 8 (200 mg, 0.47 mmol) was dissolved in 2.5 mL of anhydrous THF (previously degassed by sparging helium). 4-methylmorpholine (62 μΐ., 0.56 mmol) and 4-(4,6-dimethoxy-l,3,5-triazin-2- yl)morpholine (130 mg, 0.47 mmol) were added and the reaction mixture was stirred at rt for 30 min. N-acetyl-L-cysteine (77 mg, 0.47 mmol) was added and the reaction mixture was stirred at rt for 7 h, then overnight. Starting material was still visible, so 4-methylmorpholine (26 μΐ,, 0.235 mmol) and 4-(4,6-dimethoxy-l,3,5- triazin-2-yl)morpholine (65 mg, 0.235 mmol) were added and, after 30 min also N- acetyl-L-cysteine (38 mg, 0.235 mmol) was added. The reaction mixture was stirred for 2 days. The reaction mixture was diluted with CH2C12, then washed five times with brine. The combined organic layers were dried over Na2SO4, filtered and evaporated to dryness to give 234 mg of crude product as yellow solid. The crude product was purified by column chromatography (CH2Cl2/MeOH/EtOH 9:0.5:0.5 to 8:1:1, silica weight 50 g, volume fraction: 30 mL, fractions collected from the 43th to the 49th) and then by reverse phase using Biotage SP1 instrument (SNAP KP-C18-HS 12 g, 35-70 μιη, flow rate 15 mL/min, ACN/H2O 20:80 to 80:20) to obtain 11 mg (0.019 mmol, 4%) of the desired product as yellow solid.
[4° +120 (c 0.14, MeOH).
1H NMR 400 MHz (DMSO--4) δ ppm 1.66 (s, 3H, CH3-13), 1.80 (s, 3H, CH3CONH), 1.89-1.93 (m, 2H, CH2-3'), 1.98 (s, 3H, CH3-16), 2.58 (s, 3H, CH3- 15), 2.65 (s, 3H, CH3-18), 2.64-2.69 (m, 2H, CH2-2'), 3.04-3.09 (dd, J=12.6 Hz, J=6.2 Hz, IH, SCH2), 3.37-3.41 (dd, J=12.6 Hz, J=4.6 Hz, IH, SCH2), 3.55-3.58 (m, 2H, CH2-4'), 4.02-4.07 (m, IH, NHCH), 5.89 (s, IH, CH-4), 7.51 (d, J=6.8 Hz, IH, NHCH), 12.27 (br s, IH), 13.04 (br s, IH, NH), 13.41 (br s, IH), 13.45 (s, IH).
13C NMR 100 MHz (DMSO- ) δ ppm 8.65 (C-16), 19.26 (C-15), 23.82 (CH3CONH), 25.57 (C-3'), 32.17 (C-18), 32.82 (C-13), 32.92 (SCH2), 41.28 (C- 2'), 43.62 (C-4'), 53.59 (NHCH), 57.46 (C-12), 102.01 (C-7), 102.80 (C-2), 103.49 (C-4), 106.31 (C-l l), 107.44 (C-9), 156.90 (C-6), 158.84 (C-10), 163.63 (C-8), 169.79 (CH3CONH), 172.42 (COOH), 174.14 (C-5), 176.18 (C-14), 190.09 (C-3), 198.38 (C-l), 199.14 (C-l'), 202.06 (C-17).
LC-MS (ESI), negative, m/z found: 572.9 [M-H]~, calcd for C27H3oN2O10S:
574.2.
Example 14
Synthesis of (i?^E -4-(4-((l-(6-acetyl-7,9-dihydroxy-8,9b-dimethyl-l,3- dioxo-1 ,9b-dihydrodibenzo [b,d\ furan-2(3H)- ylidene)ethyl)amino)butanamido)butanoic acid (BT115).
Figure imgf000028_0001
To a solution of 9 (50 mg, 0.12 mmol) in 1.5 mL of anhydrous THF under nitrogen, N-methylmorpholine (15 L, 0.14 mmol) was added and the reaction mixture was stirred at room temperature. After 30 min, 4-(4,6-dimethoxy- 1,3,5- triazin-2-yl)-4-methylmoφholinium chloride (65 mg, 0.23 mmol)was added and the reaction mixture was stirred at room temperature for 15 min. 4-Aminobutyric acid (12 mg, 0.12 mmol) was added and the reaction mixture was stirred at room temperature for 3 h and 30 min. The solvent was removed under reduced pressure and the solid obtained was diluted with CH2C12 and extracted twice with 0.1 Ν HCl. The combined organic layers were washed with brine, dried over Na2SO4 and evaporated to dryness to obtain 64 mg of orange solid. The crude product was then purified by flash-chromatography using Biotage SP1 instrument (prepacked 21X55 mm SNAP silica gel cartridge, silica weight 10 g, flow rate 10 mL/min, CH3OH/CH2Cl2 0:100 to 10:90) to obtain 15 mg (0.029 mmol, 25%) of orange solid (R 0.31, eluent: CH3OH/CH2Cl2 1 :9). [aft +231 (c 0.09, CHC13).
1H NM 400 MHz (DMSO- ) δ ppm 1.58-1.62 (m, 2H, CH2-3"), 1.66 (s, 3H, CH3-13), 1.82-1.89 (m, 2H, CH2-3'), 1.98 (s, 3H, CH3-16), 2.18-2.22 (m, 4H, CH2-2" and CH2-2'), 2.59 (s, 3H, CH3-15), 2.64 (s, 3H, CH3-18), 3.03-3.08 (m, 2H, CH2-4"), 3.53-3.57 (m, 2H, CH2-4'), 5.88 (s, 1H, CH-4), 7.94 (br s, 1H, H- CO), 12.32 (s, 1H, OH-10), 13.04 (br s, 1H, NH), 13.41 (s, 1H, OH-8).
13C NMR 100 MHz (DMSO- ) ppm 8.65 (C-16), 19.25 (C-15), 25.72 (C- 3'), 25.80 (C-3), 32.17 (C-18), 32.51 (C-2), 32.84 (C-13), 33.22 (C-2'), 39.14 (C- 4), 44.19 (C-4'), 57.45 (C-12), 102.02 (C-7), 102.76 (C-2), 103.49 (C-4), 106.31 (C-11), 107.46 (C-9), 156.92 (C-6), 158.86 (C-10), 163.66 (C-8), 172.12 (C-1 '), 174.16 (C-5), 175.72 (C-1"), 176.11 (C-14), 190.08 (C-3), 198.36 (C-1), 202.07 (C-17).
MS (ESI), negative, m/z found: 513.4 [M-H]\ calcd for C26H30N2O9: 514.2.
Example 15
The following compounds were prepared by reacting usnic acid with the suitable amine precursor, substantially following the procedures described in examples 1-12.
Figure imgf000029_0001
Example 16
Antimicrobial efficacy in the agar diffusion test
The antimicrobial assays were effected with the bacterial strains Staphylococcus aureus (Biogenetics) and Listeria innocua (ATCC 33090). Fresh pure cultures were used in the agar diffusion test. Culture broths maintained for a day in Tryptic Soy Broth (TSB) were seeded evenly on the surface of Muller- Hinton plates according to the Kirby-Bauer method. Usnic acid or derivatives thereof, dissolved in dimethyl sulphoxide (DMSO) (10 μΐ,, 500 μΜ), was adsorbed on sterile filter paper discs (Whatman No.l, diameter 6 mm) and positioned on the surface of the Muller-Hinton agar plate. DMSO (10 μί,) and ampicillin sodium salt (AMP) (10 uL, 500 μΜ) were used as solvent and control antibacterial respectively. The plates were incubated at 28°C for L. innocua and 37°C for S. aureus.
The antimicrobial activities of the products tested were evaluated by measuring the diameter of the clear zone (without bacterial growth) round the discs with the absorbed compound. The antibacterial activity was expressed in millimetres of bacterial growth inhibition zone. All the tests were effected in duplicate. The results are set out in Table 1.
Table 1. Antibacterial activity of usnic acid and derivatives thereof according to the disc diffusion test. (ND = no inhibition).
Figure imgf000030_0001
The compounds of examples 1, 11 and 6 (PS2, PS9 and PS12 respectively) exhibited an inhibitory effect on L. innocua, whereas the compounds of examples 10 and 2 (PS8 and ME81 respectively) and usnic acid proved effective against both S. aureus and L. innocua, The results obtained demonstrate the bacterial inhibition activity of the compounds of the invention on Gram-positive bacteria, similarly to usnic acid. Usnic acid and the derivatives thereof presented lower antibacterial activity against S. aureus than ampicillin sodium salt, and a similar or greater effect against L. innocua.
Example 17
Evaluation of the Minimum Inhibitory Concentration (MIC) and the
Minimum Lethal Concentration (MLC).
The compounds of examples 10 and 2 (PS8 and ME81) which exhibited the greatest antimicrobial efficiency in the agar diffusion test of example 16 were selected together with usnic acid to evaluate the Minimum Inhibitory Concentration (MIC) and the Minimum Lethal Concentration (MLC). The MIC and MLC were determined by the broth microdilution method (Cosentino et al., 2003; Elo et al., 2007), Briefly, the culture broths maintained overnight were prepared with Nutrient Broth (NB) for S. aureus and Tryptic Soy Broth (TSB) for L. innocua. Usnic acid and the derivatives thereof were diluted at 12 different concentrations in the interval between 34-10 and 34 μg/nlL (double serial dilutions). A volume of 10 ih was distributed in each of the two test tubes used (1.5 mL). Each test micro-organism was diluted to 105 CFU/ml, and the bacterial suspension was added (490 μΕ) to each test tube. The concentration of the bacterial suspension was confirmed by viable count on Tryptic Soy Agar (TSA). The concentrations of the substances tested were equivalent to 1.95-2000 μΜ in the test tubes of the final tests.
The test tubes were incubated at 37°C for 24 h, and the MIC and MLC were evaluated. Bacterial growth was revealed by the presence of a pellet on the base of the test tube. The MICs were determined on the basis of the first test tube without a visible pellet of bacteria, in increasing order of concentration of the compound. A volume (100 ί) from each test tube used to evaluate the MIC was suspended in TSA to confirm the MIC values and establish the MLC. The number of surviving bacteria was determined after incubation.
The MIC value was taken as the lowest concentration of the compound able to produce a significant reduction in viability (>90%), while the MLC value was taken as the concentration at which a percentage > 99.9% of the initial inoculum was killed.
All the measurements were performed in duplicate. The results are set out in Table 2.
Table 2. MIC and MLC of usnic acid and derivatives thereof on S. aureus and !-, innocua
Figure imgf000032_0001
The difference between the MIC and MLC values of usnic acid on
S. aureus suggests that this compound has a bacteriostatic effect, as previously observed (Lauterwein et al. 1995). The compounds of examples 2 and 10 presented lower MLC values than usnic acid, indicating a prevalently bactericidal effect. The MIC values were very similar for all the compounds tested on S. aureus, whereas the compounds of examples 2 and 10 presented lower MIC values on L. innocua.
Example 18
Evaluation of wound-healing activity in vitro on HaCaT keratinocytes
Background The wound-healing capacity of usnic acid derivatives was tested on an in vitro wound-healing model consisting of HaCaT keratinocyte monolayers. Said cells represent an in vitro model of keratinocyte proliferation and migration. The HaCaT cell line imitates many properties of normal epidermal keratinocytes, is not invasive and can differentiate under suitable experimental conditions (Petrussevska RT, et al. 1988; Schoop VM, et al. 1999). The HaCaT cell line was previously used in studies of the wound-healing process as an in vitro model of the system of re-epithelialisation, a phase typical of the wound-healing process (Matsuura K, et al. 2007; Ranzato, E., et al. 2008).
Chemical reagents and cell cultures in vitro
All the reagents were supplied by Sigma Chemical Co. unless otherwise indicated. The cells were maintained at 37°C, 5% CO2 in DMEM culture medium supplemented with 10% foetal bovine serum (FBS, Euroclone, Pero, Italy) and 1% antibiotic mixture.
Cell viability assay
Cell viability was assayed on HaCaT cells and human epithelial carcinoma cells A431. The Neutral Red Uptake (NRU) test was used for this analysis, according to the method reported by Borenfreund et al. (Borenfreund, E., et al. 1989). Said cell viability assay is based on incorporation of the neutral red stain in the lysosomes of the viable cells after incubation with the test agent. Briefly, the cells were seeded in 96-well plates (20,000 cells per well), grown for 24 h before the experiment and then exposed for 24 h to different concentrations of usnic acid or derivatives thereof. After removal of the medium, an 0.05% neutral red solution was added to each well, followed by incubation for 3 h at 37°C. The cells were then washed with PBS, and a 1% solution of glacial acetic acid in 50% ethanol was added to fix the cells and extract the neutral red stain incorporated in the lysosomes. The absorbance of the supernatants was measured at 540 run with a microplate reader. The estimates of IC50 and IC05 and their 95% confidence intervals (95% CI) were obtained with a Microsoft Excel® application developed by CSIRO, Australia, based on the logistical function:
/ = !
0 + exp(-ra - (log(D) - log(IC50))
wherein /is cell viability (on a 0-1 scale) dependent on dose D, and m is the slope of the curve or Hill coefficient (Barnes, M., et al., R., and Stevens, D., 2003).
The results are set out in Table 3.
Table 3. In vitro toxicity of usnic acid and derivatives thereof on HaCaT keratinocytes, as estimated with NRU endpoint after 24 h exposure.
Figure imgf000034_0001
PS2 (1) 163 (108-244) 16 (8.0-30)
ME81 (2) 47 (39-58) —
ME56 (3) 155 (116-207) 43 (18-103)
MB73 (7) 150 (140-161) 67 (55-80)
Usnic acid 24 (18-32) 0.5 (0.2-1.3) IC50 = median effective concentration; IC05 = toxicity threshold; the 95%
CIs are shown in brackets
The compounds of examples 1, 2, 3 and 7 (PS2, ME81, MB56 and MB73 respectively) proved less toxic than usnic acid.
Scratch wound test
The scratch wound test was effected on confluent HaCaT monolayers by selecting the compounds with the most potent antibacterial activity, namely the compounds of examples 10 and 2 (PS8 and ME81 respectively), and those with the lowest toxicity on the keratinocytes, namely the compounds of examples 3 and 7 (ME56 and MB73 respectively). The analysis was effected as described in Ranzato et al. (Ranzato, E., et al. 2009a).
The cells were grown to confluency in multiwell plates, and the cell layers were then scratched with the tip of a sterile 0.1-10 μΐ, pipette. After washing away the cells in suspension, the cultures were incubated again with the medium and exposed to usnic acid and derivatives thereof, used at the concentration of 5 μΜ for 24 h. Some samples, used as positive control, were exposed to a dose of 20% (v/v) of platelet lysate (PL) which, according to earlier studies, promotes wound- healing in those cells, and was obtained from blood samples as described in Ranzato et al. (Ranzato, E., et al. 2008).
After the treatments, the cells were fixed with 3.7% formaldehyde in PBS for 30 min, and then stained with 0.1% toluidine blue at room temperature for 30 min (Figure 1, top image). The size of the wound was measured at the time of the lesion and at the end of the treatment, using a Televal inverted microscope (Carl Zeiss Inc., Thornwood, NY) equipped with a digital video camera and NIH ImageJ software. Wound closing was determined as the difference in the size of the wound at 0 and 24 h.
Usnic acid presented a significant wound-closure effect, as previously observed (Burlando, B, et al. 2009), while all the compounds of the invention tested presented greater wound-closing capacity (Figure 1, bottom panel). In particular, the compound of example 7 (GABA-usnic derivative MB73) stimulated faster wound-healing, almost equalling the wound-closure capacity of platelet lysate.
Table 4 Wound-closure of keratinocytes subjected to scratch wound assay in the presence of different compounds
Figure imgf000036_0001
The would-closure rate is the difference between the rim distance at zero time and after 24hours. Values are expressed as the percent of the would-closure rate obtained with platelet lysate Example 19
Evaluation of wound-healing activity in vivo
Animals
Male Sprague-Dawley rats (160-180 g) and Swiss albino mice (20-25 g) were supplied by the Saki Yenilli animal breeding laboratory (Ankara, Turkey). The animals were maintained under environmental conditions for 3 days for acclimatisation purposes. During the experiments they received a standard diet of pellets, and unlimited access to water. The study was authorised by the Institutional Animal Ethics Committee and performed in compliance with international legislation governing experiments on animals and biodiversity.
Preparation of test samples for the bioassays
Incision and excision wound models were used to evaluate wound-healing activity. For the wound in the in vivo models, the test samples were prepared using as base an ointment (carrier) consisting of glycol stearate, propylene glycol and liquid paraffin (3 :6: 1) at the concentration of 1 %. Aliquots of 0.5 g of each ointment to be tested were applied locally to the wound created with a surgical blade.
The animals in the carrier group were only treated with the basic ointment, while the animals in the reference medicament group were treated with 0.5 g of Madecassol® (Bayer, 00001199), which contains 1% of Centella asiatica extract.
Linear incision wound model
A total of seven rats per group were anaesthetised with 0.15 cc of Ketalar® (Shetty et al., 2006), and the dorsal region of the rats was shaved and cleaned with 70% alcohol. Two linear incisions 5 cm long were made in the paravertebral region with a sterile blade on the shaven skin at a distance of 1.5 cm from the median dorsal line, on each side. The wounds were closed with three surgical stitches at 1 cm intervals.
The ointments prepared with the test samples, the reference medicament (Madecassol®) or the basic ointment [glycol stearate: propylene glycol: liquid paraffin (3:6:1)] were applied locally to the wounds in the dorsal region in each group of animals once a day for 9 days. All the stitches were removed on the last day, and the ultimate tensile strength of the previously damaged skin was measured with a tensiometer (Zwick/Roell Z0.5, Germany) (Lodhi et al., 2006; Suguna et al., 2002).
Circular excision wound model
This model was used to monitor the process of contraction of the wound and the wound closure time. Each group of animals (seven animals in each) was anaesthetised with 0.01 cc of Ketalar®.
The fur on the backs of the mice was shaved, creating a circular wound in the interscapular dorsal region of the animals through excision of the skin with a 5 mm biopsy drill (Nopa instruments, Germany); the wounds were left open (Tramontina et al., 2002).
The test samples, the reference medicament (Madecassol®, Bayer) and the ointments used as carrier were applied once a day until the wound had completely healed. The progressive changes in the wound area were monitored with a camera (Fuji, S20 Pro, Japan) every two days. The wound area was then evaluated with the use of the AutoCAD program. The contraction of the wound was measured as the percentage reduction of the wound area. A tissue sample was taken from the healed skin of each group of mice for histopathological examination (Sadaf et al., 2006).
Statistical data analysis
Statistical analysis of the percentage data relating to anti-inflammatory and wound-healing activity was performed with analysis of variance (ANOVA). Values ofp < 0.05 were considered statistically significant.
The histopathological data were non-quantitative, so no statistical tests were performed. Results
Tables 5 and 6 show the results of the experiment.
As shown in Table 5, topical application of the ointment prepared with the compound of example 7 (MB73) on incised wounds presented the best result in terms of the ultimate tensile strength of the wound: 47.6% (p < 0.01) on the tenth day. Moreover, usnic acid and the compound of example 2 (ME81) generally proved more efficient (31.9% and 29.3% respectively) in the linear incision wound model.
Table 6 shows the contraction values in the development of wound healing on circular excision wound models for the groups treated with the carrier, the negative control, the compounds and the reference medicament. Usnic acid and compounds MB73 and ME81 exhibited potential wound-healing activity, while the carrier and negative control groups did not exhibit statistically significant wound-healing activity.
The wound contractions, on the eighth and tenth days respectively, were
52.42% (p < 0.01) and 82.95% (p < 0,001) for the group treated with the compound MB73, 30.36% (p < 0.05) and 52.19% (p < 0.01) for the Group treated with usnic acid, 37.55% (p < 0.05) and 40.83%(p < 0.01) for the group treated with compound ME81, and were compared with the reference medicament Madecassol® [72.24% (p < 0.01) -100% (p < 0.001)].
Table 5. Effect of ointments containing usnic acid and derivatives thereof on the linear incision wound model
Material (Example) Statistical Mean ± (Ultimate tensile
S.E.M. strength %)
Carrier 11.75 ± 2.48 10.6
Negative control 10.62 ± 2.63 -
Usnic acid 15.50 ± 2.14 31.9**
MB73 (7) 17.34 ± 1.92 47.6**
ME81 (2) 15.19 ± 2.02 29.3*
Madecassol 18.87 ± 1.39 60.6*** *: p < 0.05; **: p < 0.01; ***: p < 0,001; S.E.M.: standard error of the mean Percentage values of ultimate tensile strength: the group treated with the carrier was compared with the negative control group; the compounds and the reference medicament were compared with the group treated with the carrier.
Table 6. Effect of ointments containing usnic acid and derivatives thereof on the circular excision wound model
Figure imgf000040_0001
*: p < 0.05; **: p < 0.01; ***: p < 0.001; S.E.M.: standard error of the mean Percentage values of ultimate tensile strength: the group treated with the was compared with the negative control group; the compounds and the reference medicament were compared with the group treated with the carrier. Bibliography
Aronovitz G.H. (2000). Antimicrobial therapy of acute otitis media: review of treatment recommendations. Clin. Ther. 22(1): 29-39.
Barnes, M., Correll, R., and Stevens, D., 2003. A simple spreadsheet for estimating low-effect concentrations and associated logistic dose response curves.
Solutions to pollution: Program abstract book. The Society of Environmental
Toxicology and Chemistry Asia/Pacific - Australasian Society of Ecotoxicology.
SETAC ASE ASIA PACIFIC.
Borenfreund, E., Babich, H., and Martin- Alguacil, N., 1989. Effect of methylazoxymethanol acetate on bluegill sunfish cell cultures in vitro. Ecotoxicol.
Environ. Saf 17, 297-307.
Burlando B., Ranzato E., Volante A., Appendino G., Pollastro F., Verotta L.
(2009). Antiproliferative Effects on Tumor Cells and Promotion of Keratinocytes
Wound Healing by Different Lichen Compounds. Planta Med. 75 (6): 607-613. - Cosentino S., Barra A., Pisano B., Cabizza M., Pirisi FM., Palmas F.
(2003). Composition and antimicrobial properties of Sardinian Juniperus essential oils against foodborne pathogens and spoilage microorganisms. J. Food Prot.
66(7):1288-91.Guo L., Shi Q., Fang J.-L., Mei N., Afshan A.A., Lewis S.M.,
Leakey J.E.A., Frankos V.H. (2008). Review of usnic acid and usnea barbata toxicity. J. Environ. Sci & Health, Part C: Environ. Carcin. Eco. R. 26 (4): 317-
338.
Elo H., Matikainen J., Pelttari E. (2007). Potent activity of the lichen antibiotic (+)-usnic acid against clinical isolates of vancomycin-resistant enterococci and methicillin-resistant Staphylococcus aureus. Naturwissenschaften. 94(6):465-8.
Hausen B.M., Emde L., Marks V. (1993). An investigation of the allergenic constituents of Cladonia stellaris (Opiz) Pous & Vezda ('silver moss', 'reindeer moss' or 'reindeer lichen'). Contact dermatitis 28(2): 70-6. Jin J., Dong Y., He L. (2005). The study on skin wound healing promoting action of sodium usnic acid. Zhong Yao Cai = Journal of Chinese medicinal materials 28(2): 109-111.
Ingolfsdottir K. (2002). Usnic acid. Phytochemistry 61 (7): 729-736.
- Kokubun T., Shiu W.K.P., Gibbons S. (2007). Inhibitory activities of lichen-derived compounds against methicillin- and multidrug-resistant Staphylococcus aureus. Planta Med. 73 (2): 176-179.
Matsuura K, Kuratani T, Gondo T et al. Promotion of skin epithelial cell migration and wound healing by a 2-benzazepine derivative. Eur J Pharmacol. 2007; 563: 83-87.
Lauterwein M., Oethinger M., Belsner K., Peters T., Marre R. (1995). In vitro activities of the lichen secondary metabolites vulpinic acid, (+)-usnic acid and (-)-usnic acid against aerobic and anaerobic microorganisms. Antimicrob. Agents Chemother. 39:2541-2543.
- Lodhi, S., Pawar, R.S., Jain, A.P., Singhai, A.K., 2006. Wound healing potential of Tephrosia purpurea (Linn.) Pers. in rats. Journal of Ethnopharmacology 108, 204-210.
Mertz P., Ovington L. (1993). Wound healing microbiology. Dermatol. Clin. 11(4): 739-747.
- Natic M., Tesic Z., Andelkovic K., Brceski I., Radulovic S., Manic S., Sladic D. (2004). Synthesis and Biological Activity of Pd(II) and Cu(II) Complexes with Acylhydrazones of Usnic Acid. Syn. React. Inorg. Met. 34 (1): 101-113.
Nunes P.S., Albuquerque-Junior R.L.C., Cavalcante D.R.R., Dantas M.D.M., Cardoso, J.C., Bezerra M.S., Souza J.C.C., Serafmi M.R., Quintas- Junior L.J., Bonjardim L.R., Araujo, A.A.S. (2011). Collagen-based films containing liposome-loaded usnic acid as dressing for dermal burn healing. J. Biomed. Biotech. 1-9. Petrussevska RT, Breitkreutz D et al. Normal keratinization in a spontaneously immortalized aneuploid human keratinocyte cell line. J. Cell Biol. 1988; 106: 761-771.
Ranzato et al. Br J Dermatol 2008 Sep; 159:537-45
- Ranzato et al. 2009. Platelet Derivatives: A New Horizon in Regenerative Medicine. In: Leon V. Berhardt (Ed) Advances in Medicine and Biology, Volume 11, Nova Science Publishers, Inc.).
Ranzato, E., Mazzucco, L., Patrone, M., and Burlando, B., 2009a. Platelet lysate promotes in vitro wound scratch closure of human dermal fibroblasts: different roles of cell calcium, P38, ERK and PI3K/AKT. J Cell Mol Med 13, 2030-2038.
Reyim M., Abdulla A. (2010). Extraction technology of usnic acid from Cladonia cornuta (L.) Hoffm and content determination. Zhongguo Niangzao 11 : 122-124.
- Sadaf, F., Saleem, R., Ahmed, M., Ahmad, S.L, Navaid-ul, Z., 2006. Healing potential of cream containing extract of Sphaeranthus indicius on dermal wounds in guinea pigs. Journal of Ethnopharmacology 107, 161-163.
Schoop VM, Mirancea N, Fusenig NE. Epidermal organization and differentiation of HaCaT keratinocytes in organotypic coculture with human dermal fibroblasts. J. Invest Dermatol. 1999; 112: 343-353.
Seifert P., Bertrand C. (1995). Usnic acid: a natural preservative from lichens. Cosmetic News 18(102): 169-72.
Shetty, B.S., Udupa, S.L., Udupa, A.L., Somayaji, S.N., 2006. Effect of Centella asiatica L (Umbelliferae) on normal and dexamethasone-suppressed wound healing in wistar albino rats. The International Journal of Lower Extremity Wounds 5, 137-143.
Suguna, L., Singh, S., Sivakumar, P., Sampath, P., Chandrakasan, G., 2002. Influence of Terminalia chebula on dermal wound healing in rats. Phytotherapy Research 16, 227-231.
Tomasi S. et al. Solid-Phase Synthesis of Polyfunctionalized Natural Products: Application to Usnic Acid, a Bioactive Lichen Compound. J. Comb. Chem. 2006, 8, 1 1-14.
- Tramontina, V.A., Machado, M.A., Nogueira Filho Gda R., Kim, S.H., Vizzioli, M.R., Toledo, S., 2002. Effect of bismuth subgallate (local hemostatic agent) on wound healing in rats. Histological and histometric findings. Brazilian Dental Journal 13, 11-16.
Variati R. (2010). Usnic acid derivatives, a family of natural derived bacteriostatic substances and their use in cosmetic products. Olaj, Szappan, Kozmetika 59(1): 11- 16.

Claims

1. Compound of general formula (I) :
Figure imgf000045_0001
(I) wherein:
represents a single or double bond; R is a residue selected from:
- H
- (CH2)nX
- (CH2)nC(O)NH(CH2)mX
Figure imgf000045_0002
Figure imgf000045_0003
Figure imgf000046_0001
wherein the symbol 1 represents the attachment point of the R group to the N(H) group;
X is selected from OH, SH, COOH, SO3H;
n and m independently from each other are an integer from 1 to 10, preferably from 2 to 4; with the proviso that when X is OH or SH, the minimum value of n or m is 2;
their enantiomers and salts acceptable for pharmaceutical or cosmetic use, for use as antibacterial and wound healing agents.
2. Compound for use according to claim 1, wherein R is a residue selected from:
- H
- (CH2)2SH
- (CH2)2SO3H
- CH2COOH (CH2)3COOH
(CH2)4OH
CH2)3C(O)NH(CH2)3COOH
Figure imgf000047_0001
3. Compound of general formula (II)
Figure imgf000048_0001
(II) wherein:
represents a single or double bond;
R' is a residue selected from:
- (CH2)nX'
- (CH2)nC(O)NH(CH2)mX
Figure imgf000048_0002
Figure imgf000049_0001
wherein the symbol represents the attachment point of the R' group to the N(H) group;
X is selected from OH, SH, COOH, SO3H;
X' is selected from OH, SH, COOH;
n and m independently from each other are an integer from 1 to 10, preferably from 2 to 4;
with the following provisos:
when X or X' is OH or SH, the minimum value of n or m is 2;
when X' is COOH, n is different from 1 or 2;
when X' is OH, n is different from 4;
their enantiomers and salts acceptable for pharmaceutical or cosmetic use.
4. Compound according to claim 3 wherein R' is selected in the group of:
- H
- (CH2)2SH
- CH2COOH
- (CH2)3COOH
- (CH2)4OH
- (CH2)3C(O)NH(CH2)3COOH
Figure imgf000049_0002
Figure imgf000050_0001
5. Compound according to claim 3 or 4 for use as a medicament.
6. Compound according to claim 3 or 4 for use as an antibacterial or wound healing agent.
7. Dermatological or cosmetic composition containing a compound according to claims 1-4 together with excipients compatible for pharmaceutical or cosmetic use.
8. Compound according to claims 1-4 or composition according to claim 7 for use in the regenerative or anti-aging treatment of skin tissues.
9. Compound or composition according to claim 8 wherein the regenerative or anti-aging treatment of skin tissues comprises the treatment of topical infections selected from infected burns, topical otitis, haemorrhoids, vaginal lesions and mouth infections.
PCT/EP2013/062664 2012-06-20 2013-06-18 Dibenzofuran derivatives with antibacterial and wound-healing activity WO2013189950A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
ITMI2012A001082 2012-06-20
IT001082A ITMI20121082A1 (en) 2012-06-20 2012-06-20 DIBENZOFURANIC DERIVATIVES WITH ANTIBACTERIAL AND CICATRIZING ACTIVITIES

Publications (1)

Publication Number Publication Date
WO2013189950A1 true WO2013189950A1 (en) 2013-12-27

Family

ID=46582946

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2013/062664 WO2013189950A1 (en) 2012-06-20 2013-06-18 Dibenzofuran derivatives with antibacterial and wound-healing activity

Country Status (2)

Country Link
IT (1) ITMI20121082A1 (en)
WO (1) WO2013189950A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB800114A (en) 1956-02-21 1958-08-20 Laeaeke Ag Method of preparing derivatives from usnic acid
WO2010034512A1 (en) * 2008-09-29 2010-04-01 Universita' Degli Studi Di Milano Compounds with antimalarial activity

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB800114A (en) 1956-02-21 1958-08-20 Laeaeke Ag Method of preparing derivatives from usnic acid
WO2010034512A1 (en) * 2008-09-29 2010-04-01 Universita' Degli Studi Di Milano Compounds with antimalarial activity

Non-Patent Citations (37)

* Cited by examiner, † Cited by third party
Title
ARONOVITZ G.H.: "Antimicrobial therapy of acute otitis media: review of treatment recommendations", CLIN. THER., vol. 22, no. 1, 2000, pages 29 - 39
BARNES, M.; CORRELL, R.; STEVENS, D.: "A simple spreadsheet for estimating low-effect concentrations and associated logistic dose response curves. Solutions to pollution: Program abstract book. The Society of Environmental Toxicology and Chemistry Asia/Pacific - Australasian Society of Ecotoxicology", SETAC ASE ASIA PACIFIC, 2003
BORENFREUND, E.; BABICH, H.; MARTIN-ALGUACIL, N.: "Effect of methylazoxymethanol acetate on bluegill sunfish cell cultures in vitro", ECOTOXICOL. ENVIRON. SAF, vol. 17, 1989, pages 297 - 307
BURLANDO B.; RANZATO E.; VOLANTE A.; APPENDINO G.; POLLASTRO F.; VEROTTA L.: "Antiproliferative Effects on Tumor Cells and Promotion of Keratinocytes Wound Healing by Different Lichen Compounds", PLANTA MED., vol. 75, no. 6, 2009, pages 607 - 613
CHANGON SEO ET AL: "Usimines A-C, Bioactive Usnic Acid Derivatives from the Antarctic Lichen Stereocaulon alpinum", JOURNAL OF NATURAL PRODUCTS, vol. 71, no. 4, 1 April 2008 (2008-04-01), pages 710 - 712, XP055039737, ISSN: 0163-3864, DOI: 10.1021/np070464b *
COSENTINO S.; BARRA A.; PISANO B.; ABIZZA M.; PIRISI FM.; PALMAS F: "Composition and antimicrobial properties of Sardinian Juniperus essential oils against foodbome pathogens and spoilage microorganisms", J. FOOD PROT., vol. 66, no. 7, 2003, pages 1288 - 9L
ELO H.; MATIKAINEN J.; PELTTARI E.: "Potent activity of the lichen antibiotic (+)-usnic acid against clinical isolates of vancomycin-resistant enterococci and methicillin-resistant Staphylococcus aureus", NATURWISSENSCHAFTEN, vol. 94, no. 6, 2007, pages 465 - 8
GUO L.; SHI Q.; FANG J.-L.; MEI N.; AFSHAN A.A.; LEWIS S.M.; LEAKEY J.E.A.; FRANKOS V.H.: "Review of usnic acid and usnea barbata toxicity", J. ENVIRON. SCI & HEALTH, PART C: ENVIRON. CARCIN. ECO. R., vol. 26, no. 4, 2008, pages 317 - 338
HAUSEN B.M.; EMDE L.; MARKS V.: "An investigation of the allergenic constituents of Cladonia stellaris (Opiz) Pous & Vezda ('silver moss', 'reindeer moss' or 'reindeer lichen", CONTACT DERMATITIS, vol. 28, no. 2, 1993, pages 70 - 6
INGÓLFSDÓTTIR K.: "Usnic acid", PHYTOCHEMISTRY, vol. 61, no. 7, 2002, pages 729 - 736
JAMES P. KUTNEY ET AL: "Studies in the usnic acid series. I. The condensation of (+)-usnic acid with aliphatic and aromatic amines", CANADIAN JOURNAL OF CHEMISTRY, vol. 54, no. 17, 1 September 1976 (1976-09-01), pages 2795 - 2803, XP055039733, ISSN: 0008-4042, DOI: 10.1139/v76-395 *
JIN J.; DONG Y.; HE L.: "The study on skin wound healing promoting action of sodium usnic acid. Zhong Yao Cai", JOURNAL OF CHINESE MEDICINAL MATERIALS, vol. 28, no. 2, 2005, pages 109 - 111
KOKUBUN T.; SHIU W.K.P.; GIBBONS S.: "Inhibitory activities of lichen-derived compounds against methicillin- and multidrug-resistant Staphylococcus aureus", PLANTA MED., vol. 73, no. 2, 2007, pages 176 - 179
LAUTERWEIN M.; OETHINGER M.; BELSNER K.; PETERS T.; MARRE R.: "In vitro activities of the lichen secondary metabolites vulpinic acid, (+)-usnic acid and (-)-usnic acid against aerobic and anaerobic microorganisms", ANTIMICROB. AGENTS CHEMOTHER., vol. 39, 1995, pages 2541 - 2543
LODHI, S.; PAWAR, R.S.; JAIN, A.P.; SINGHAI, A.K.: "Wound healing potential of Tephrosia purpurea (Linn.) Pers. in rats", JOURNAL OF ETHNOPHARMACOLOGY, vol. 108, 2006, pages 204 - 210
LUZINA O A ET AL: "Chemical modification of usnic acid 2. Reactions of (+)-usnic acid with amino acids", RUSSIAN CHEMICAL BULLETIN, KLUWER ACADEMIC PUBLISHERS-PLENUM PUBLISHERS, NE, vol. 56, no. 6, 1 June 2007 (2007-06-01), pages 1249 - 1251, XP019556738, ISSN: 1573-9171, DOI: 10.1007/S11172-007-0189-7 *
MARC-ANTOINE BAZIN ET AL: "Synthesis and cytotoxic activities of usnic acid derivatives", BIOORGANIC & MEDICINAL CHEMISTRY, vol. 16, no. 14, 1 July 2008 (2008-07-01), pages 6860 - 6866, XP055039732, ISSN: 0968-0896, DOI: 10.1016/j.bmc.2008.05.069 *
MATSUURA K; KURATANI T; GONDO T ET AL.: "Promotion of skin epithelial cell migration and wound healing by a 2-benzazepine derivative", EUR J PHARMACOL., vol. 563, 2007, pages 83 - 87
MERTZ P.; OVINGTON L.: "Wound healing microbiology", DERMATOL. CLIN., vol. 11, no. 4, 1993, pages 739 - 747
NATIC M.; TESIC Z.; ANDELKOVIC K.; BRCESKI I.; RADULOVIC S.; MANIC S.; SLADIC D.: "Synthesis and Biological Activity of Pd(II) and Cu(II) Complexes with Acylhydrazones of Usnic Acid.", SYN. REACT. INORG. MET., vol. 34, no. 1, 2004, pages 101 - 113
NUNES P.S.; ALBUQUERQUE-JUNIOR R.L.C.; CAVALCANTE D.R.R.; DANTAS M.D.M.; CARDOSO, J.C.; BEZERRA M.S.; SOUZA J.C.C.; SERAFINI M.R.;: "Collagen-based films containing liposome-loaded usnic acid as dressing for dermal bum healing.", J BIOMED. BIOTECH., 2011, pages 1 - 9
PETRUSSEVSKA RT; BREITKREUTZ D ET AL.: "Normal keratinization in a spontaneously immortalized aneuploid human keratinocyte cell line", J. CELL BIOL., vol. 106, 1988, pages 761 - 771
RANZATO ET AL., BR J DERMATOL, vol. 159, September 2008 (2008-09-01), pages 537 - 45
RANZATO ET AL.: "Advances in Medicine and Biology", vol. 11, 2009, NOVA SCIENCE PUBLISHERS, INC., article "Platelet Derivatives: A New Horizon in Regenerative Medicine."
RANZATO, E.; MAZZUCCO, L.; PATRONE, M.; BURLANDO, B.: "Platelet lysate promotes in vitro wound scratch closure of human dermal fibroblasts: different roles of cell calcium, P38, ERK and PI3K/AKT", J CELL MOL MED, vol. 13, 2009, pages 2030 - 2038
REYIM M.; ABDULLA A.: "Extraction technology of usnic acid from Cladonia comuta (L.) Hoffin and content determination", ZHONGGUO NIANGZAO, vol. 11, 2010, pages 122 - 124
SADAF, F.; SALEEM, R.; AHMED, M.; AHMAD, S.I.; NAVAID-UL, Z.: "Healing potential of cream containing extract of Sphaeranthus indicius on dermal wounds in guinea pigs", JOURNAL OFETHNOPHARMACOLOGY, vol. 107, 2006, pages 161 - 163
SCHOOP VM; MIRANCEA N; FUSENIG NE: "Epidermal organization and differentiation of HaCaT keratinocytes in organotypic coculture with human dermal fibroblasts", J.INVEST DERMATOL., vol. 112, 1999, pages 343 - 353
SEGATORE B; BELLIO P; SETACCI D; BRISDELLI F; PIOVANO M; GARBARINO JA; NICOLETTI M; AMICOSANTE G; PERILLI M; CELENZA G: "In vitro interaction of usnic acid in combination with antimicrobial agents against methicillin-resistant Staphylococcus aureus clinical isolates determined by FICI and AE model methods", PHYTOMEDICINE, vol. 19, no. 3-4, 25 November 2011 (2011-11-25), pages 341 - 7
SEIFERT P. ET AL., COSMETIC NEWS, 1995
SEIFERT P.; BERTRAND C.: "Usnic acid: a natural preservative from lichens", COSMETIC NEWS, vol. 18, no. 102, 1995, pages 169 - 72
SHETTY, B.S.; UDUPA, S.L.; UDUPA, A.L.; SOMAYAJI, S.N.: "Effect of Centella asiatica L (Umbelliferae) on normal and dexamethasone-suppressed wound healing in wistar albino rats", THE INTERNATIONAL JOURNAL OF LOWER EXTREMITY WOUNDS, vol. 5, 2006, pages 137 - 143
SUGUNA, L.; SINGH, S.; SIVAKUMAR, P.; SAMPATH, P.; CHANDRAKASAN, G.: "Influence of Terminalia chebula on dermal wound healing in rats", PHYTOTHERAPY RESEARCH, vol. 16, 2002, pages 227 - 231
TOMASI S ET AL: "Solid-Phase Synthesis of Polyfunctionalized Natural Products:", JOURNAL OF COMBINATORIAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY, WASHINGTON, US, vol. 8, no. 1, 1 January 2006 (2006-01-01), pages 11 - 14, XP009163315, ISSN: 1520-4766 *
TOMASI S. ET AL.: "Solid-Phase Synthesis of Polyfunctionalized Natural Products: Application to Usnic Acid, a Bioactive Lichen Compound.", J. COMB. CHEM., vol. 8, 2006, pages 11 - 14
TRAMONTINA, V.A.; MACHADO, M.A.; NOGUEIRA FILHO GDA R.; KIM, S.H.; VIZZIOLI, M.R.; TOLEDO, S.: "Effect of bismuth subgallate (local hemostatic agent) on wound healing in rats. Histological and histometric findings", BRAZILIAN DENTAL JOURNAL, vol. 13, 2002, pages 11 - 16
VARIATI R.: "Usnic acid derivatives, a family of natural derived bacteriostatic substances and their use in cosmetic products", OLAJ, SZAPPAN, KOZMETIKA, vol. 59, no. 1, 2010, pages 11 - 16

Also Published As

Publication number Publication date
ITMI20121082A1 (en) 2013-12-21

Similar Documents

Publication Publication Date Title
US20230365491A1 (en) Method for inhibiting or disrupting biofilm formation, or reducing biofilm
Saurav et al. Cytotoxicity and antioxidant activity of 5-(2, 4-dimethylbenzyl) pyrrolidin-2-one extracted from marine Streptomyces VITSVK5 spp.
US10064897B2 (en) Treating a bacterial skin infection with a whole, leech saliva extract
KR101159699B1 (en) Antimicrobial agent and antimicrobial composition
Sepčić et al. Biological activities of aqueous extracts from marine sponges and cytotoxic effects of 3-alkylpyridinium polymers from Reniera sarai
CN112341522A (en) Antibacterial peptide and application thereof
KR20180024002A (en) Novel bicyclic lipotropic peptide, preparation and use as antimicrobial agent
WO2013189950A1 (en) Dibenzofuran derivatives with antibacterial and wound-healing activity
Luisella et al. Dibenzofuran derivatives with antibacterial and wound-healing activity
JP3403780B2 (en) Cosmetics
Manivannan et al. Isolation, identification and antibacterial and wound healing studies of quercetin-3-O-α-L-rhamnopyranoside-2"-gallate
CN100522992C (en) Novel ring-shape small-peptide BA and its use
JP2013166732A (en) Method for producing endothelin activity inhibitor and method for producing usnea longissima extract
KR101313724B1 (en) The hair growth solution mainly comprised of Ecklonia cava extract, and the hair treatment composition
EP2752416B1 (en) Novel mangromicin compound and production method therefor
JP2004345971A (en) Anti-influenza virus agent using chlorogenate derivative
US20110033566A1 (en) Anti-bacterial composition comprising extract from barks of Alnus pendula Matsum
Bsharat et al. Synthesis and biological evaluation of several new heterocyclic carvacrol esters
JPWO2020170388A1 (en) Type I collagen or elastin production promoter
WO2023276786A1 (en) Copper chelator, anticancer agent and prophylactic or therapeutic agent for wilson&#39;s disease
Munawar et al. Antibacterial, antioxidant and anti-inflammatory potential of the different extracts of Holoptelia integrifolia
Akkol et al. Michela Bruno, Beatrice Trucchi, Bruno Burlando c, Elia Ranzato c, Simona Martinotti c
EP4070798A1 (en) Chronic wound healing composition and application thereof
WO2016016582A1 (en) Novel strombine salts and cosmetic use thereof
KR101123366B1 (en) Complex of antioxidant and chitosan having improved stability

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: 13736501

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: 13736501

Country of ref document: EP

Kind code of ref document: A1