WO2012019772A1 - Method for treating protozoan parasitic diseases - Google Patents

Method for treating protozoan parasitic diseases Download PDF

Info

Publication number
WO2012019772A1
WO2012019772A1 PCT/EP2011/004055 EP2011004055W WO2012019772A1 WO 2012019772 A1 WO2012019772 A1 WO 2012019772A1 EP 2011004055 W EP2011004055 W EP 2011004055W WO 2012019772 A1 WO2012019772 A1 WO 2012019772A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
leishmania
compound
nhr
nhs0
Prior art date
Application number
PCT/EP2011/004055
Other languages
French (fr)
Inventor
Denis Sereno
Elodie Gazanion
Baptiste Vergnes
Jean-François GUICHOU
Gilles Labesse
Original Assignee
Institut De Recherche Pour Le Developpement (I.R.D)
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 Institut De Recherche Pour Le Developpement (I.R.D) filed Critical Institut De Recherche Pour Le Developpement (I.R.D)
Publication of WO2012019772A1 publication Critical patent/WO2012019772A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/12Ketones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/06Aluminium, calcium or magnesium; Compounds thereof, e.g. clay
    • A61K33/12Magnesium silicate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/02Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C259/00Compounds containing carboxyl groups, an oxygen atom of a carboxyl group being replaced by a nitrogen atom, this nitrogen atom being further bound to an oxygen atom and not being part of nitro or nitroso groups
    • C07C259/04Compounds containing carboxyl groups, an oxygen atom of a carboxyl group being replaced by a nitrogen atom, this nitrogen atom being further bound to an oxygen atom and not being part of nitro or nitroso groups without replacement of the other oxygen atom of the carboxyl group, e.g. hydroxamic acids
    • C07C259/10Compounds containing carboxyl groups, an oxygen atom of a carboxyl group being replaced by a nitrogen atom, this nitrogen atom being further bound to an oxygen atom and not being part of nitro or nitroso groups without replacement of the other oxygen atom of the carboxyl group, e.g. hydroxamic acids having carbon atoms of hydroxamic groups bound to carbon atoms of six-membered aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C49/00Ketones; Ketenes; Dimeric ketenes; Ketonic chelates
    • C07C49/04Saturated compounds containing keto groups bound to acyclic carbon atoms
    • C07C49/17Saturated compounds containing keto groups bound to acyclic carbon atoms containing hydroxy groups
    • C07C49/172Saturated compounds containing keto groups bound to acyclic carbon atoms containing hydroxy groups containing rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C49/00Ketones; Ketenes; Dimeric ketenes; Ketonic chelates
    • C07C49/76Ketones containing a keto group bound to a six-membered aromatic ring
    • C07C49/82Ketones containing a keto group bound to a six-membered aromatic ring containing hydroxy groups
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates to a method of treating, preventing or inhibiting parasitic disease, in particular leishmaniasis or trypanosomiasis in a subject.
  • Leishmania parasite is the causative agent of a wide spectrum of diseases termed leishmaniasis that concern more that 12 millions people, over 88 countries around the world.
  • Leishmaniasis is caused by several species of Leishmania, such as L. aethiopica, L. donovani, L. infantum, L. major, L. mexicana or L. tropica.
  • leishmaniasis is essentially caused by L. infantum.
  • two different hosts lodge the parasite.
  • Promastigotes develop in the gut of a phlebotomine sandfly that transmits flagellated infectious metacyclic forms into a vertebrate host during the bloodmeal. They rapidly differentiate into non-flagellated amastigotes, which actively divide within the mononuclear phagocytes. These particular niches constitute specific environments where the parasite has to find essential nutrients required for its development.
  • cutaneous Three main clinical variants of this disease are known: cutaneous, mucocutaneous, and visceral.
  • Cutaneous leishmaniasis can manifest itself as a single skin ulceration at the site of the sandfly bite appearing soon after infection or month later as disseminated lesions.
  • Mucocutaneous syndrome develop as the cutaneous form, but progresses months or years later to lesions of the mouth, nose, or pharynx.
  • Visceral leishmaniasis the most severe form, can prove fatal in the absence of treatment
  • the trypanosomiasis is another current parasitic disease of humans and animals, caused by Trypanosoma.
  • Trypanosoma brucei can cause human African trypanosomiasis, also called sleeping sickness. Without treatment, the disease is invariably fatal, with progressive mental deterioration leading to coma and death. Damage caused in the neurological phase is irreversible.
  • the current antiparasitic treatment need an intensive drug administration and is not always efficient.
  • Trypanosoma cruzi can cause American trypanosomiasis, also called Chagas disease.
  • the infection of parasite can affect nervous system, digestive system and hear.
  • the current antiparasitic treatment can only delay in poor level the development of disease symptoms during the chronic phase of the disease.
  • the currently available antiparasitic treatments can cause side effects in many patients including skin disorders, brain toxicity, and digestive system irritation.
  • - A represents a group chosen from a phenyl, a triazolyl, and a thienyl
  • - Ri represents -CO-CH 2 OH or -CO-NHOH
  • R 2 is chosen from H, triazolyl, methyltriazolyl, -CONHR 3 , -CH 2 -CONHR 3 , - NHS0 2 NHR 3 , -CH 2 -NHS0 2 NHR 3 , wherein R 3 represents H, a (C]-C 8 ) alkyl group, an aryl group, a (C 3 -C 8 ) cycloalkyl group, a heterocyclic group, or a heteroaryl group,
  • Cj-Cs alkyl denotes a straight or branched chain hydrocarbon group with 1 to 8 carbon atoms, especially with 1 to 6 carbon atoms, more especially with 1 to 4 carbon atoms.
  • Examples of such groups are methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec- butyl, tert-butyl, pentyl, isopentyl, neopentyl, tert-pentyl, hexyl, isohexyl, 1,1-dimethylbutyl, 2,2-dimethylbutyl, -3,3-dimethylbutyl and 2-ethylbutyl.
  • (C3-C 8 )cycloalkyl group represents a saturated cyclic hydrocarbon such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and cyclooctyl.
  • aryl refers to a 6- to 18-membered monocyclic, bicyclic, tricyclic, or polycyclic aromatic hydrocarbon ring system.
  • Examples of an aryl group include phenyl, naphthyl, pyrenyl, anthracyl, quinolyl, and isoquinolyl.
  • heterocyclic group represents 3- to 7-membered non-aromatic ring system containing 1 or 2 hetero atoms selected from nitrogen, oxygen and sulfur or sulfur oxidized to sulfones or sulfoxides.
  • heterocyclic groups are oxiranyl, azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, tetrahydrofuryl, thiolanyl, piperidyl, tetrahydropyranyl, mo holinyl, thiomorpholinyl and piperazinyl.
  • heteroaryl group represents 5- or 6- membered aromatic mono- or bicyclic heterocydic group having 1 to 4 heteroatoms chosen from N, O and S.
  • aromatic heterocydic groups include pyridyl, pyrimidinyl, pyridazinyl, pyrrolyl, furanyl, thiophenyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, thiazolyl, oxazolyl, 1,2,4- oxadiazolyl, 1,2,3-oxadiazolyl, 1,3,4-oxadiazolyl, 1,2,3-thiadiazolyl, 1 ,2,4-thiadiazolyl, 1,3,4- thiadiazolyl, 1,2,4-triazolyl, 1,3,4-triazolyl, 1,2,3-triazolyl and tetrazolyl; examples of bicyclic heteroaryl groups are benzofuranyl, isobenzo
  • the present invention concerns a compound of formula (1), wherein:
  • - A represents a phenyl group
  • - Ri represents -CO-CH 2 OH or -CO-NHOH
  • R 2 in para-position of R is chosen from H, triazolyl, methyl triazolyl, -CONHR 3 , -CH 2 - CONHR 3 , -NHS0 2 NHR 3 , -CH2-NHS0 2 NHR 3 , wherein R 3 represents H, a (d-C 8 ) alkyl group, an aryl group, a (C 3 -C 8 ) cycloalkyl group, a heterocyclic group, or a heteroaryl group,
  • the present invention concerns more particularly a compound of formula (1), wherein:
  • - A represents a phenyl group
  • - Ri represents -CO-CH 2 OH or -CO-NHOH
  • Rl represents -CO-CH 2 OH and said compound corresponds to the following formula (la):
  • alpha-hydroxyacetophenone alpha-hydroxyacetophenone
  • Rl represents -CO-NHOH and said compound corresponds to the following formula (lb): named also N-hydroxyberizamide.
  • the compound according to the invention may be synthesized by any method known from the art or is commercially available.
  • the compounds of the present invention are useful as a drug for treating, inhibiting or preventing parasitic diseases, in particular protozoan parasitic diseases of a subject.
  • the parasitic disease is caused by a protozoan parasite of the family of the Trypanosomatidae selected from the genus Trypanosoma or the genus Leishmania.
  • the parasite of the genus Trypanosoma can cause various trypanosomiases, such as sleeping sickness caused by Trypanosoma brucei, or Chagas disease caused by Trypanosoma cruzi.
  • the compound of the present invention is for use as drug for treating, inhibiting or preventing parasitic diseases caused by Trypanosoma. . brucei. Gambiense.
  • the parasite of the genus Leishmania is the parasite responsible for the disease leishmaniasis.
  • the compound of the present invention is for use as drug for treating, inhibiting or preventing parasitic diseases caused by Leishmania. aethiopica, Leishmania. donovani, Leishmania. mexicana, Leishmania. infantum, Leishmania. tropica and Leishmania. major, preferably by Leishmania. infantum, Leishmania. tropica and Leishmania. major, more preferably by Leishmania. infantum.
  • the above defined compound is used as drug for treating, inhibiting or preventing a leishmaniasis, in particular cutaneous leishmaniasis, mucocutaneous leishmaniasis, or visceral leishmaniasis, or a trypanosomiasis.
  • the subject is a mammalian including human and dog, more preferably, human.
  • the present invention provides also a composition comprising:
  • - A represents a group chosen from a phenyl, a triazolyl, and a thienyl
  • - Rj represents -COCH 2 OH or -CO-NHOH
  • R 2 is chosen from H, triazolyl, methyltriazolyl, -CONHR 3 , -CH 2 -CONHR 3 , - NHS0 2 NHR 3 , -CH 2 -NHS0 2 NHR 3 , wherein R 3 represents H, a (C C 8 ) alkyl group, an aryl group, a (C 3 -C 8 ) cycloalkyl group, a heterocyclic group, or a heteroaryl group,
  • anti-parasitic compound selected from the group comprising : miltefosin, antimony based drugs, like meglumine antimoniate or sodium stibogluconate, amphotericin B, benznidazol, nifurtimox, paromomycin, pentamidin and its derivatives, arsenic derivatives, melarsoprol and difluoromethylornithin.
  • composition can be used as a drug, in particular a drug for treating, inhibiting or preventing a parasitic disease, in particular a leishmaniasis, more particularly cutaneous leishmaniasis, mucocutaneous leishmaniasis, or visceral leishmaniasis, or a trypanosomiasis.
  • a parasitic disease in particular a leishmaniasis, more particularly cutaneous leishmaniasis, mucocutaneous leishmaniasis, or visceral leishmaniasis, or a trypanosomiasis.
  • the administration of the above defined compound and the composition can be carried out by oral, rectal, nasal, topical (including buccal and sublingual), parenteral (including subcutaneous, intramuscular, intravenous and intradermal) or intralesional route.
  • parenteral including subcutaneous, intramuscular, intravenous and intradermal
  • intralesional means that the administration of the compound of formula (1) is carried out at the sites of parasite-caused skin lesions of patients, in particular in case of Leishmania infections.
  • the above defined compound or the composition is administrated to a subject in need thereof in a therapeutically effective amount.
  • therapeutically effective amount refers to an amount which prevents, inhibits, suppresses or reduces the amount of Leishmania or Trypanosoma amastigotes, promastigotes, or both in a subject as compared to a control.
  • the therapeutically effective amount may be determined by conventional methods known in the art.
  • a therapeutically effective amount of a compound of formula (I) is about 5 mg/m 2 /day to about 5 g/m 2 /day, in particular of about 500 mg/m 2 /day to about 3 g/m /day.
  • this standard amount could be influenced by certain factors, such as the severity of the disease or disorder, previous treatment, the general health and/or age of the subject accepting the treatment, and other disease present.
  • the present invention provides a method for reducing, suppressing or inhibiting a protozoan parasite of the family of the Trypanosomatidae in a target comprising contacting the target with an effective amount of an above defined composition or compound.
  • the target is a mammal tissue or cells derived therefrom.
  • the mammal tissue is a human tissue, a dog tissue.
  • the protozoan parasite is chosen from Leishmania parasite, in particular Leishmania. aethiopica, Leishmania. donovani, Leishmania. mexicana, Leishmania. infantum, Leishmania, tropica and Leishmania. major, preferably from Leishmania. infantum, Leishmania. tropica and Leishmania. major, more preferably from Leishmania. Infantum, or trypanosome parasite, in . particular Trypanosoma, brucei. gambiense.
  • the aboved defined compound or composition reduces, suppresses, or inhibits the Leishmania or Trypanosoma parasite growth, infection or proliferation by about 50% at a concentration of about 350 ⁇ or less, about 200 ⁇ or less, about 100 ⁇ or less, about 70 ⁇ or less, about 50 ⁇ or less.
  • the present invention provides a pharmaceutical composition comprising as active substance at least a therapeutically effective amount of a compound of formula (1):
  • - A represents a group chosen from a phenyl, a triazolyl, and a thienyl
  • - Ri represents -CO-CH 2 OH or -CO-NHOH
  • R 2 is chosen from H, triazolyl, methyltriazolyl, -CONHR 3 , -CH 2 -CONHR 3 , - NHS0 2 NHR 3 , -CH 2 -NHS0 2 NHR 3 , wherein R 3 represents H, a (Ci-C 8 ) alkyl group, an aryl group, a (C 3 -C 8 ) cycloalkyl group, a heterocyclic group, or a heteroaryl group,
  • a parasitic disease in particular of protozoan parasitic diseases, more particularly of leishmaniasis or trypanosomiasis, in a subject in need thereof.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising as active substance at least a therapeutically effective amount of a compound of formula (1) and at least one anti -parasitic compound, such as a compound selected from: miltefosin, antimony based drugs, like meglumine antimoniate or sodium stibogluconate, amphotericin B, benzimidazol, nifurtimox, paromomycin, pentamidin and its derivatives, arsenic derivatives, melarsoprol and difluoromethylornithin.
  • the present invention provides a product containing:
  • anti-parasitic compound such as a compound selected from: miltefosin, antimony based drugs, like meglumine antimoniate or sodium stibogluconate, amphotericin B, benzimidazol, nifurtimox, paromomycin, pentamidin and its derivatives, arsenic derivatives, melarsoprol and difluoromethylornithin,
  • a compound selected from: miltefosin, antimony based drugs, like meglumine antimoniate or sodium stibogluconate, amphotericin B, benzimidazol, nifurtimox, paromomycin, pentamidin and its derivatives, arsenic derivatives, melarsoprol and difluoromethylornithin such as a compound selected from: miltefosin, antimony based drugs, like meglumine antimoniate or sodium stibogluconate, amphotericin B, benzimi
  • compositions or separate parts of the above defined products could include pharmaceutically acceptable salts of the compound of formula (1) and/or pharmaceutically acceptable carriers.
  • pharmaceutically acceptable salt refers to salt forms that are pharmacologically acceptable and substantially non-toxic to the subject being treated with the compound of the invention.
  • the pharmaceutically acceptable salt forms of the compound of formula (la) or formula (lb) may be prepared according to methods well known in the art.
  • pharmaceutically acceptable carriers refers to non-API substances such as disintegrators, binders, fillers, and lubricants used in formulating pharmaceutical products. They are generally non-toxic for administering to humans.
  • compositions or separate parts of the above defined products could be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries or suspensions.
  • compositions or products could be carried out by oral, rectal, nasal, topical (including buccal and sublingual), parenteral (including subcutaneous, intramuscular, intravenous and intradermal) or intralesional route.
  • the present invention provides also a method for treating, inhibiting, or preventing parasitic diseases, in particular protozoan parasitic diseases, more particularly leishmaniasis, or trypanosomiasis, in a subject in need thereof comprising administering to said subject at least a therapeutically effective amount of a compound of formula (1):
  • - A represents a group chosen from a phenyl, a triazolyl, and a thienyl
  • - R represents -CO-CH 2 OH or -CO-NHOH
  • R 2 is chosen from H, triazolyl, methyltriazolyl, -CONHR 3 , -CH 2 -CONHR 3 , - NHS0 2 NHR 3 , -CH 2 -NHS0 2 NHR 3 , wherein R 3 represents H, a (C C 4 ) alkyl group, an aryl group, a (C 3 -C 8 ) cycloalkyl group, a heterocyclic group, or a heteroaryl group,
  • Figure 1 illustrates the growth curves of promastigote of L. infantum parasites in presence of lb (represented by ⁇ ), la (represented by A) and nicotinamide (represented by ⁇ ). Results are the mean of two to four experiments, each performed in triplicate. The mean percentage of survival promastigotes respectively treated by compounds la, lb or nicotinamide is marked on ordinate axis. The concentration of compounds la or lb or nicotinamide in different cell cultures is marked on abscissa axis.
  • Alpha-hydroxyacetophenone (compound la) and N-hydroxybenzamide (compound lb) were dissolved in 100 % DMSO at a concentration of 100 mM and stored at +4°C. For in vitro tests, compounds were diluted in culture medium and used immediately.
  • Promastigotes of Leishmania infantum (MHOM/MA/67/ITMAP-263), L. major (MHOM/SU/73/5-ASKH), L. tropica (MHOM/SU/74/SAF-K27), L. amazonensis (MHOM/BR/73/M2269) and L. braziliensis (MHOM/BR/75/M2904) were maintained at 26°C in SDM-79 medium (Brun and Schonenberger, Acta Tropica 1979;36(3):289-292) supplemented with 10% heat-inactivated foetal calf serum (FCS; Lonza), 100 IU penicillin/ml, 100 ⁇ g streptomycin/ml and 5 mg/1 hemin.
  • FCS heat-inactivated foetal calf serum
  • the L. infantum RSb80 and RSM60 strains were selected from a wild-type population using a stepwise selection until they were resistant to 80 ⁇ g/ml and 160 ⁇ g/ml trivalent antimony (Sblll), respectively.
  • the sensitivity towards Sblll was determined after 3 days of incubation in presence of growing concentrations of Sblll.
  • the IC 5 o values for Sblll are 132.3 ⁇ 14.-6 ⁇ g/ml and 163.2 ⁇ 3.7 ⁇ g/ml for RSb80 and RSM60 strains, respectively. Both strains are 10- fold less sensitive to Sblll than wild-type parental strain.
  • Epimastigotes of Trypanosoma brucei gambiense were maintained at 26°C in SM medium (Cunningham, J Protozool 1977;21(2):325-329) supplemented with 10% FCS, 2 mM glutamine, 100 IU penicillin/ml and 100 ⁇ g streptomycin/ml.
  • Human leukemia monocyte cell line (THP-1) was cultured in RPMI 1640 medium (Lonza) supplemented with 10% FCS, 2 mM glutamine, 100 IU penicillin/ml and 100 ⁇ g streptomycin/ml, and incubated at 37°C with 5% C0 2 .
  • Late-log phase parasites were seeded at 1 x 10 6 cells/ml in 96-well plates and grown at 26°C in the presence of serial dilutions of each compound. Each concentration point was tested in triplicate and all experiments were performed two times. The final concentration of DMSO did not exceed 0.5%, which was not toxic for parasites. After 3 days of incubation for Leishmania strains and 5 days for T. b. gambiense, viability and growth of parasites were determined by flow cytometry analysis. Briefly, a sample of cells (5 ⁇ ) was taken from each well and diluted into 500 ⁇ of PBS (pH 7.2).
  • Propidium iodide was added at a final concentration of 1 ⁇ / ⁇ 1 and cells were analyzed on a flow cytometer (FacsaliburTM; Becton Dickinson) for 52 s.
  • the parasite concentration was determined using standard curves where parasite concentrations were plotted as a function of mean number of cells counted by the cytometer after 52 s.
  • THP-1 cells were cultured in RPMI 1640 medium supplemented with 10% FCS, 2 mM glutamine, 100 IU of penicillin/ml, and 100 ⁇ g of streptomycin/ml.
  • THP-1 cells in the log phase of growth were differentiated in macrophages by incubation for 2 days in medium containing 20 ng/ml of phorbol myristate acetate in 96-well plates, at 37°C with 5% C0 2 . Cells were washed once and serial dilutions of compounds up to 1 mM were added in culture medium. Each concentration point was tested in triplicate. Cells were incubated for 4 days at 37°C with 5% C0 2 . The cytotoxic effect of each compound on non-infected macrophages was ascertained using trypan blue coloration.
  • THP-1 cells in the log phase of growth were differentiated in Labtek chamber slides at a concentration of 5 ⁇ 10 4 cells/well by incubation for 2 days in medium containing 20 ng of phorbol myristate acetate/ml.
  • Five-day-old promastigote cultures were transferred to Schneider's medium (Lonza) at pH 5.4 supplemented with 20% FCS and incubated 24 hours at 26°C.
  • PI parasitic index
  • alpha-hydroxyacetophenone (compound la) and N- hydroxybenzamide (compound lb) are able to inhibit the in vitro proliferation of trypanosomatid parasites, such as Leishmania and Trypanosoma brucei species.
  • the compounds exhibited micromolar inhibitory activities against promastigote and intracellular amastigote forms of Leishmania parasites with IC50 values ranging from 195.8 ⁇ to 384.2 ⁇ for compound la and from 19.6 ⁇ to 174.1 ⁇ for compound lb.
  • these two compounds are not toxic on non-infected differentiated monocytes at antileishmanial doses.
  • T. b. gambiense parasites are highly sensitive to both compounds with IC 50 values as low as 40.7 ⁇ .

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Inorganic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

The present invention relates to a method of treating, preventing or inhibiting parasitic disease, in particular leishmaniasis or trypanosomiasis in a subject, by a compound of formula (1): R2-A-R1 wherein: - A represents a group chosen from a phenyl, a triazolyl, and a thienyl, - R1 represents -CO-CH2OH or -CO-NHOH, - R2 is chosen from H, triazolyl, methyltriazolyl, -CONHR3, -CH2-CONHR3, - 'NHS02NHR3, -CH2-NHSO2NHR3, wherein R3 represents H, a (C1-C8) alkyl group, an aryl group, a (C3-C8) cycloalkyl group, a heterocyclic group, or a heteroaryl group. Or a pharmaceutically acceptable salt thereof.

Description

METHOD FOR TREATING PROTOZOAN PARASITIC DISEASES
The present invention relates to a method of treating, preventing or inhibiting parasitic disease, in particular leishmaniasis or trypanosomiasis in a subject.
The protozoan Leishmania parasite is the causative agent of a wide spectrum of diseases termed leishmaniasis that concern more that 12 millions people, over 88 countries around the world. Leishmaniasis is caused by several species of Leishmania, such as L. aethiopica, L. donovani, L. infantum, L. major, L. mexicana or L. tropica. Around the Mediterranean and in France in particular, leishmaniasis is essentially caused by L. infantum. During the ife cycle of Leishmania parasites, two different hosts lodge the parasite. Promastigotes develop in the gut of a phlebotomine sandfly that transmits flagellated infectious metacyclic forms into a vertebrate host during the bloodmeal. They rapidly differentiate into non-flagellated amastigotes, which actively divide within the mononuclear phagocytes. These particular niches constitute specific environments where the parasite has to find essential nutrients required for its development.
Three main clinical variants of this disease are known: cutaneous, mucocutaneous, and visceral. Cutaneous leishmaniasis can manifest itself as a single skin ulceration at the site of the sandfly bite appearing soon after infection or month later as disseminated lesions. Mucocutaneous syndrome develop as the cutaneous form, but progresses months or years later to lesions of the mouth, nose, or pharynx. Visceral leishmaniasis, the most severe form, can prove fatal in the absence of treatment
Currently, basic treatment for all forms of leishmaniases consists of the administration of pentavalent antimony SbV (sodium stibogluconate Pentostam or meglumine antimoniate Glucantime) or more recently the liposomal formulation of amphotericin B in endemic mediterranean countries and for immunocompromised patients. Alternative drug options include pentamidine, miltefosine, paromomycin and allopurinol but their use is limited due to their toxicity or declined efficacy. These drugs are difficult to handle and have serious side effects. By the way, some compounds, such as amphotericin B, are only efficient for visceral leishmaniasis, but not for nonvisceral disease.
The trypanosomiasis is another current parasitic disease of humans and animals, caused by Trypanosoma. For example, Trypanosoma brucei can cause human African trypanosomiasis, also called sleeping sickness. Without treatment, the disease is invariably fatal, with progressive mental deterioration leading to coma and death. Damage caused in the neurological phase is irreversible. The current antiparasitic treatment need an intensive drug administration and is not always efficient.
Trypanosoma cruzi can cause American trypanosomiasis, also called Chagas disease. The infection of parasite can affect nervous system, digestive system and hear. The current antiparasitic treatment can only delay in poor level the development of disease symptoms during the chronic phase of the disease. By the way, the currently available antiparasitic treatments can cause side effects in many patients including skin disorders, brain toxicity, and digestive system irritation.
Thus, there is a need for providing new medicaments without the drawbacks of the currently used medicaments, for the treatment of protozoan parasitic diseases, such as leishmaniasis or trypanosomiasis.
Consequently, the present invention provides a compound of formula (1):
R2-A-R1,
wherein:
- A represents a group chosen from a phenyl, a triazolyl, and a thienyl,
- Ri represents -CO-CH2OH or -CO-NHOH,
- R2 is chosen from H, triazolyl, methyltriazolyl, -CONHR3, -CH2-CONHR3, - NHS02NHR3, -CH2-NHS02NHR3, wherein R3 represents H, a (C]-C8) alkyl group, an aryl group, a (C3-C8) cycloalkyl group, a heterocyclic group, or a heteroaryl group,
or a pharmaceutically acceptable salt thereof, for use as a drug.
The term "Cj-Cs alkyl" denotes a straight or branched chain hydrocarbon group with 1 to 8 carbon atoms, especially with 1 to 6 carbon atoms, more especially with 1 to 4 carbon atoms. Examples of such groups are methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec- butyl, tert-butyl, pentyl, isopentyl, neopentyl, tert-pentyl, hexyl, isohexyl, 1,1-dimethylbutyl, 2,2-dimethylbutyl, -3,3-dimethylbutyl and 2-ethylbutyl.
The term (C3-C8)cycloalkyl group represents a saturated cyclic hydrocarbon such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and cyclooctyl.
The term "aryl" refers to a 6- to 18-membered monocyclic, bicyclic, tricyclic, or polycyclic aromatic hydrocarbon ring system. Examples of an aryl group include phenyl, naphthyl, pyrenyl, anthracyl, quinolyl, and isoquinolyl.
The term "heterocyclic group" represents 3- to 7-membered non-aromatic ring system containing 1 or 2 hetero atoms selected from nitrogen, oxygen and sulfur or sulfur oxidized to sulfones or sulfoxides. Examples of such groups are oxiranyl, azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, tetrahydrofuryl, thiolanyl, piperidyl, tetrahydropyranyl, mo holinyl, thiomorpholinyl and piperazinyl.
The term "heteroaryl group" represents 5- or 6- membered aromatic mono- or bicyclic heterocydic group having 1 to 4 heteroatoms chosen from N, O and S. Examples of such aromatic heterocydic groups include pyridyl, pyrimidinyl, pyridazinyl, pyrrolyl, furanyl, thiophenyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, thiazolyl, oxazolyl, 1,2,4- oxadiazolyl, 1,2,3-oxadiazolyl, 1,3,4-oxadiazolyl, 1,2,3-thiadiazolyl, 1 ,2,4-thiadiazolyl, 1,3,4- thiadiazolyl, 1,2,4-triazolyl, 1,3,4-triazolyl, 1,2,3-triazolyl and tetrazolyl; examples of bicyclic heteroaryl groups are benzofuranyl, isobenzofuranyl, benzo[b]thienyl, indolyl, isoindolyl, 1H- indazolyl, benzimidazolyl, benzoxazolyl, benzisoxazolyl, benzothiazolyl, benzisothiazolyl, benzodioxolyl, IH-benzotriazolyl, quinolyl, isoquinolyl, cinnolinyl, quinazolinyl; quinoxalinyl or phthalazinyl.
In a particular embodiment, the present invention concerns a compound of formula (1), wherein:
- A represents a phenyl group,
- Ri represents -CO-CH2OH or -CO-NHOH,
- R2 in para-position of R is chosen from H, triazolyl, methyl triazolyl, -CONHR3, -CH2- CONHR3, -NHS02NHR3, -CH2-NHS02NHR3, wherein R3 represents H, a (d-C8) alkyl group, an aryl group, a (C3-C8) cycloalkyl group, a heterocyclic group, or a heteroaryl group,
said compound corresponding to formula (la) :
Figure imgf000004_0001
The present invention concerns more particularly a compound of formula (1), wherein:
- A represents a phenyl group,
- R2 represents H,
- Ri represents -CO-CH2OH or -CO-NHOH,
said compound corresponding to formula I
Figure imgf000004_0002
In a preferred embodiment, Rl represents -CO-CH2OH and said compound corresponds to the following formula (la):
Figure imgf000005_0001
named alpha-hydroxyacetophenone.
In another preferred embodiment, Rl represents -CO-NHOH and said compound corresponds to the following formula (lb):
Figure imgf000005_0002
named also N-hydroxyberizamide.
The compound according to the invention may be synthesized by any method known from the art or is commercially available.
Particularly, the compounds of the present invention are useful as a drug for treating, inhibiting or preventing parasitic diseases, in particular protozoan parasitic diseases of a subject.
More particularly, the parasitic disease is caused by a protozoan parasite of the family of the Trypanosomatidae selected from the genus Trypanosoma or the genus Leishmania.
The parasite of the genus Trypanosoma can cause various trypanosomiases, such as sleeping sickness caused by Trypanosoma brucei, or Chagas disease caused by Trypanosoma cruzi.
In a particular embodiment, the compound of the present invention is for use as drug for treating, inhibiting or preventing parasitic diseases caused by Trypanosoma. . brucei. Gambiense.
The parasite of the genus Leishmania is the parasite responsible for the disease leishmaniasis.
In a particular embodiment, the compound of the present invention is for use as drug for treating, inhibiting or preventing parasitic diseases caused by Leishmania. aethiopica, Leishmania. donovani, Leishmania. mexicana, Leishmania. infantum, Leishmania. tropica and Leishmania. major, preferably by Leishmania. infantum, Leishmania. tropica and Leishmania. major, more preferably by Leishmania. infantum.
In another particular embodiment, the above defined compound is used as drug for treating, inhibiting or preventing a leishmaniasis, in particular cutaneous leishmaniasis, mucocutaneous leishmaniasis, or visceral leishmaniasis, or a trypanosomiasis.
In a more particular embodiment of the above defined compound, the subject is a mammalian including human and dog, more preferably, human.
The present invention provides also a composition comprising:
- a compound of formula (1):
Figure imgf000006_0001
wherein:
- A represents a group chosen from a phenyl, a triazolyl, and a thienyl,
- Rj represents -COCH2OH or -CO-NHOH,
- R2 is chosen from H, triazolyl, methyltriazolyl, -CONHR3, -CH2-CONHR3, - NHS02NHR3, -CH2-NHS02NHR3, wherein R3 represents H, a (C C8) alkyl group, an aryl group, a (C3-C8) cycloalkyl group, a heterocyclic group, or a heteroaryl group,
or a pharmaceutically acceptable salt thereof, and
- at least one anti-parasitic compound, selected from the group comprising : miltefosin, antimony based drugs, like meglumine antimoniate or sodium stibogluconate, amphotericin B, benznidazol, nifurtimox, paromomycin, pentamidin and its derivatives, arsenic derivatives, melarsoprol and difluoromethylornithin.
The above defined composition can be used as a drug, in particular a drug for treating, inhibiting or preventing a parasitic disease, in particular a leishmaniasis, more particularly cutaneous leishmaniasis, mucocutaneous leishmaniasis, or visceral leishmaniasis, or a trypanosomiasis.
The administration of the above defined compound and the composition can be carried out by oral, rectal, nasal, topical (including buccal and sublingual), parenteral (including subcutaneous, intramuscular, intravenous and intradermal) or intralesional route. As intended herein "intralesional" means that the administration of the compound of formula (1) is carried out at the sites of parasite-caused skin lesions of patients, in particular in case of Leishmania infections.
The above defined compound or the composition is administrated to a subject in need thereof in a therapeutically effective amount.
The term "therapeutically effective amount" refers to an amount which prevents, inhibits, suppresses or reduces the amount of Leishmania or Trypanosoma amastigotes, promastigotes, or both in a subject as compared to a control. The therapeutically effective amount may be determined by conventional methods known in the art.
In a preferred embodiment, a therapeutically effective amount of a compound of formula (I) is about 5 mg/m2/day to about 5 g/m2/day, in particular of about 500 mg/m2/day to about 3 g/m /day. However, one skilled in the art will know that this standard amount could be influenced by certain factors, such as the severity of the disease or disorder, previous treatment, the general health and/or age of the subject accepting the treatment, and other disease present.
In other embodiments, the present invention provides a method for reducing, suppressing or inhibiting a protozoan parasite of the family of the Trypanosomatidae in a target comprising contacting the target with an effective amount of an above defined composition or compound.
The target is a mammal tissue or cells derived therefrom. Preferably, the mammal tissue is a human tissue, a dog tissue.
In a preferred embodiment, the protozoan parasite is chosen from Leishmania parasite, in particular Leishmania. aethiopica, Leishmania. donovani, Leishmania. mexicana, Leishmania. infantum, Leishmania, tropica and Leishmania. major, preferably from Leishmania. infantum, Leishmania. tropica and Leishmania. major, more preferably from Leishmania. Infantum, or trypanosome parasite, in . particular Trypanosoma, brucei. gambiense.
According to a preferred embodiment of said method, the aboved defined compound or composition reduces, suppresses, or inhibits the Leishmania or Trypanosoma parasite growth, infection or proliferation by about 50% at a concentration of about 350 μΜ or less, about 200 μΜ or less, about 100 μΜ or less, about 70 μΜ or less, about 50 μΜ or less. In other embodiments, the present invention provides a pharmaceutical composition comprising as active substance at least a therapeutically effective amount of a compound of formula (1):
Figure imgf000008_0001
wherein:
- A represents a group chosen from a phenyl, a triazolyl, and a thienyl,
- Ri represents -CO-CH2OH or -CO-NHOH,
- R2 is chosen from H, triazolyl, methyltriazolyl, -CONHR3, -CH2-CONHR3, - NHS02NHR3, -CH2-NHS02NHR3, wherein R3 represents H, a (Ci-C8) alkyl group, an aryl group, a (C3-C8) cycloalkyl group, a heterocyclic group, or a heteroaryl group,
or a pharmaceutically acceptable salt thereof,
for its use in the treatment or prevention or inhibition of a parasitic disease, in particular of protozoan parasitic diseases, more particularly of leishmaniasis or trypanosomiasis, in a subject in need thereof.
In other embodiments, the present invention provides a pharmaceutical composition comprising as active substance at least a therapeutically effective amount of a compound of formula (1) and at least one anti -parasitic compound, such as a compound selected from: miltefosin, antimony based drugs, like meglumine antimoniate or sodium stibogluconate, amphotericin B, benzimidazol, nifurtimox, paromomycin, pentamidin and its derivatives, arsenic derivatives, melarsoprol and difluoromethylornithin.
In other embodiments, the present invention provides a product containing:
- at least a therapeutically effective amount of a compound of formula (1), and
- at least one anti-parasitic compound, such as a compound selected from: miltefosin, antimony based drugs, like meglumine antimoniate or sodium stibogluconate, amphotericin B, benzimidazol, nifurtimox, paromomycin, pentamidin and its derivatives, arsenic derivatives, melarsoprol and difluoromethylornithin,
as a combined preparation for simultaneous, separate or sequential use in the prevention or the treatment of parasitic diseases, in particular of protozoan parasitic diseases, more particularly of leishmaniasis or trypanosomiasis. The above defined pharmaceutical compositions or separate parts of the above defined products could include pharmaceutically acceptable salts of the compound of formula (1) and/or pharmaceutically acceptable carriers.
The term "pharmaceutically acceptable salt" refers to salt forms that are pharmacologically acceptable and substantially non-toxic to the subject being treated with the compound of the invention. The pharmaceutically acceptable salt forms of the compound of formula (la) or formula (lb) may be prepared according to methods well known in the art.
The term "pharmaceutically acceptable carriers" refers to non-API substances such as disintegrators, binders, fillers, and lubricants used in formulating pharmaceutical products. They are generally non-toxic for administering to humans.
The above defined pharmaceutical compositions or separate parts of the above defined products could be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries or suspensions.
The administration of the above defined pharmaceutical compositions or products could be carried out by oral, rectal, nasal, topical (including buccal and sublingual), parenteral (including subcutaneous, intramuscular, intravenous and intradermal) or intralesional route.
The present invention provides also a method for treating, inhibiting, or preventing parasitic diseases, in particular protozoan parasitic diseases, more particularly leishmaniasis, or trypanosomiasis, in a subject in need thereof comprising administering to said subject at least a therapeutically effective amount of a compound of formula (1):
- R2-A-R1,
wherein:
- A represents a group chosen from a phenyl, a triazolyl, and a thienyl,
- R, represents -CO-CH2OH or -CO-NHOH,
- R2 is chosen from H, triazolyl, methyltriazolyl, -CONHR3, -CH2-CONHR3, - NHS02NHR3, -CH2-NHS02NHR3, wherein R3 represents H, a (C C4) alkyl group, an aryl group, a (C3-C8) cycloalkyl group, a heterocyclic group, or a heteroaryl group,
or a pharmaceutically acceptable salt thereof.
The present invention is illustrated by the following figures and examples.
Figure 1 illustrates the growth curves of promastigote of L. infantum parasites in presence of lb (represented by■), la (represented by A) and nicotinamide (represented by ·). Results are the mean of two to four experiments, each performed in triplicate. The mean percentage of survival promastigotes respectively treated by compounds la, lb or nicotinamide is marked on ordinate axis. The concentration of compounds la or lb or nicotinamide in different cell cultures is marked on abscissa axis.
Examples
1.1. Compounds
Alpha-hydroxyacetophenone (compound la) and N-hydroxybenzamide (compound lb) were dissolved in 100 % DMSO at a concentration of 100 mM and stored at +4°C. For in vitro tests, compounds were diluted in culture medium and used immediately.
1.2. Strains and culture conditions
Promastigotes of Leishmania infantum (MHOM/MA/67/ITMAP-263), L. major (MHOM/SU/73/5-ASKH), L. tropica (MHOM/SU/74/SAF-K27), L. amazonensis (MHOM/BR/73/M2269) and L. braziliensis (MHOM/BR/75/M2904) were maintained at 26°C in SDM-79 medium (Brun and Schonenberger, Acta Tropica 1979;36(3):289-292) supplemented with 10% heat-inactivated foetal calf serum (FCS; Lonza), 100 IU penicillin/ml, 100 μg streptomycin/ml and 5 mg/1 hemin.
The L. infantum RSb80 and RSM60 strains were selected from a wild-type population using a stepwise selection until they were resistant to 80 μg/ml and 160 μg/ml trivalent antimony (Sblll), respectively. The sensitivity towards Sblll was determined after 3 days of incubation in presence of growing concentrations of Sblll. The IC5o values for Sblll are 132.3 ± 14.-6 μg/ml and 163.2 ± 3.7 μg/ml for RSb80 and RSM60 strains, respectively. Both strains are 10- fold less sensitive to Sblll than wild-type parental strain.
Epimastigotes of Trypanosoma brucei gambiense (MHOM/CI/86/DAL967) were maintained at 26°C in SM medium (Cunningham, J Protozool 1977;21(2):325-329) supplemented with 10% FCS, 2 mM glutamine, 100 IU penicillin/ml and 100 μg streptomycin/ml.
Human leukemia monocyte cell line (THP-1) was cultured in RPMI 1640 medium (Lonza) supplemented with 10% FCS, 2 mM glutamine, 100 IU penicillin/ml and 100 μg streptomycin/ml, and incubated at 37°C with 5% C02.
1.3. IC50 determination of selected compounds on parasites
Late-log phase parasites were seeded at 1 x 106 cells/ml in 96-well plates and grown at 26°C in the presence of serial dilutions of each compound. Each concentration point was tested in triplicate and all experiments were performed two times. The final concentration of DMSO did not exceed 0.5%, which was not toxic for parasites. After 3 days of incubation for Leishmania strains and 5 days for T. b. gambiense, viability and growth of parasites were determined by flow cytometry analysis. Briefly, a sample of cells (5 μΐ) was taken from each well and diluted into 500 μΐ of PBS (pH 7.2). Propidium iodide was added at a final concentration of 1 μ /ηι1 and cells were analyzed on a flow cytometer (Facsalibur™; Becton Dickinson) for 52 s. The parasite concentration was determined using standard curves where parasite concentrations were plotted as a function of mean number of cells counted by the cytometer after 52 s.
1.4. Cellular toxicity of compounds against non-infected THP-1 differentiated monocytes
THP-1 cells were cultured in RPMI 1640 medium supplemented with 10% FCS, 2 mM glutamine, 100 IU of penicillin/ml, and 100 μg of streptomycin/ml. THP-1 cells in the log phase of growth were differentiated in macrophages by incubation for 2 days in medium containing 20 ng/ml of phorbol myristate acetate in 96-well plates, at 37°C with 5% C02. Cells were washed once and serial dilutions of compounds up to 1 mM were added in culture medium. Each concentration point was tested in triplicate. Cells were incubated for 4 days at 37°C with 5% C02. The cytotoxic effect of each compound on non-infected macrophages was ascertained using trypan blue coloration.
1.5. IC50 determination of selected compounds on L. infantum intracellular amastigotes
The capacity of parasites to infect and replicate within macrophage host cells was evaluated according to a method adapted from da Luz et al. (da Luz et al, Antimicrob Agents Chemother 2009;53(12):5197-5203). THP-1 cells in the log phase of growth were differentiated in Labtek chamber slides at a concentration of 5 χ 104 cells/well by incubation for 2 days in medium containing 20 ng of phorbol myristate acetate/ml. Five-day-old promastigote cultures were transferred to Schneider's medium (Lonza) at pH 5.4 supplemented with 20% FCS and incubated 24 hours at 26°C. Parasites were then used for the infection of freshly differentiated macrophages at a parasite macrophage ratio of 10:1. Infection was allowed to occur during 24 hours at 37°C with 5% C02, after which non- internalized parasites were removed and compounds were added to the media. After 4 days of incubation, cells were fixed with methanol and stained with Giemsa. The parasitic index (PI) was defined as the percentage of infected macrophages x number of intracellular parasites/ macrophage . 2. Results and discussion
In this study, the in vitro antiprotozoal activity of two compounds is evaluated, the alpha-hydroxyacetophenone, designed as compound la, and a hydroxamate-based compound, N-hydroxybenzamide, designed as compound lb.
The in vitro bioactivity of both compounds is determined on L. infantum promastigotes and intracellular amastigotes. The IC50 values are presented in Table 1. The results show that the two compounds have potent inhibitory activities against both developmental stages with IC50 values ranging from 29.3 μΜ to 348.0 μΜ. The cellular toxicity on non-infected macrophages was checked and it is founded that these cells were at least 10-fold less sensitive to compounds la and lb, as compared to wild-type promastigotes (Table 1). The efficacy of the two compounds is further tested on Sblll-resistant L. infantum parasites, namely RSb80 and RSM60 strains (see material and method section), and it is founded that these parasites are as sensitive as wild-type strain (Table 1).
Figure imgf000012_0001
Table 1
Comparison of survival of promastigote of L. infantum in presence of compounds lb, la and nicotinamide was also determined as described in materials and methods section. Growth curve experiments showed that compounds la and lb are 40 to 400 fold more active than nicotinamide, respectively (Figure 1).
The antiparasitic activity of compounds la and lb was then determined on a large panel of Leishmania species, responsible for various clinical symptoms, and on Trypanosoma brucei gambiense that causes Human African Trypanosomiasis. The IC50 values of both compounds on promastigote (Leishmania) or epimastigote (Trypanosoma) forms are listed in Table 2. The results show some discrepancies of sensibility to compounds la and lb, depending on the species tested. L. braziliensis appears to be the most sensitive species, among the Leishmania strains tested. Interestingly, compounds la and lb showed similar and high efficacy against T. b. gambiense parasites.
Figure imgf000013_0001
Table 2
In conclusion, it is demonstrated that alpha-hydroxyacetophenone (compound la) and N- hydroxybenzamide (compound lb) are able to inhibit the in vitro proliferation of trypanosomatid parasites, such as Leishmania and Trypanosoma brucei species. The compounds exhibited micromolar inhibitory activities against promastigote and intracellular amastigote forms of Leishmania parasites with IC50 values ranging from 195.8 μΜ to 384.2 μΜ for compound la and from 19.6 μΜ to 174.1 μΜ for compound lb. In addition, these two compounds are not toxic on non-infected differentiated monocytes at antileishmanial doses. T. b. gambiense parasites are highly sensitive to both compounds with IC50 values as low as 40.7 μΜ.

Claims

1. Compound of formula (1):
R2-A-R,,
wherein:
- A represents a group chosen from a phenyl, a triazolyl, and a thienyl,
- Ri represents -CO-CH2OH or -CO-NHOH,
- R2 is chosen from H, triazolyl, methyltriazolyl, -CONHR3, -CH2-CONHR3, - NHS02NHR3, -CH2-NHS02NHR3, wherein R3 represents H, a (C,-C8) alkyl group, an aryl group, a (C3-C ) cycloalkyl group, a heterocyclic group, or a heteroaryl group.
or a pharmaceutically acceptable salt thereof, for its use as a drug.
2. Compound according to claim 1, wherein:
- A represents a phenyl group,
- Ri represents -CO-CH2OH or -CO-NHOH,
- R2 in para-position of Ri is chosen from H, triazolyl, methyltriazolyl, -CONHR3, -CH2- CONHR3, -NHS02NHR3, -CH2-NHS02NHR3, wherein R3 represents H, or a (C,-C8) alkyl group, an aryl group, a (C3-C8) cycloalkyl group, a heterocyclic group, or a heteroaryl group,
said compound corresponding to formula (l a) :
Figure imgf000014_0001
3. Compound according to claim 2, wherein:
- A represents a phenyl group,
- R2 represents H,
- Ri represents -CO-CH2OH or -CO-NHOH,
said compound corresponding to formula I
Figure imgf000014_0002
4. Compound according to claim 1 to 3, for its use as a drug for treating, inhibiting or preventing parasitic diseases, in particular protozoan parasitic diseases of a subject.
5. Compound according to claim 4, wherein the parasitic disease is caused by a protozoan parasite of the family of the Trypanosomatidae selected from the genus Trypanosoma or the genus Leishmania.
6. Compound according to claim 5, wherein the protozoan parasite of the genus Leishmania is selected from: Leishmania. infantum, Leishmania. tropica and Leishmania. Major.
7. Compound according to claim 5, wherein the protozoan parasite of the genus Trypanosoma is selected from Trypanosoma, brucei. gambiense.
8. Compound according to claim 4, wherein the parasitic diseases is a leishmaniasis, in particular cutaneous leishmaniasis, mucocutaneous leishmaniasis, or visceral leishmaniasis, or a trypanosomiasis.
9. Compound according to any one of claims 4 to 8, wherein the subject is a mammalian including human and dog.
10. Composition comprising:
- a compound of formula (1):
R2-A-Rl 5
wherein:
- A represents a group chosen from a phenyl, a triazolyl, and a thienyl,
- R, represents -CO-CH2OH or -CO-NHOH,
- R2 is chosen from H, triazolyl, methyltriazolyl, -CONHR3, -CH2-CONHR3, - NHS02NHR3, -CH2-NHS02NHR3, wherein R3 represents H, a (C,-C8) alkyl group, an aryl group, a (C3-C8) cycloalkyl group, a heterocyclic group, or a heteroaryl group,
or a pharmaceutically acceptable salt thereof, and
- at least one anti-parasitic compound, selected from the group comprising : miltefosin, antimony based drugs, like meglumine antimoniate or sodium stibogluconate, amphotericin B, benzimidazol, nifurtimox, paromomycin, pentamidin and its derivatives, arsenic derivatives, melarsoprol and difluoromethylornithin.
1 1. Method for reducing, suppressing or inhibiting a protozoan parasite of the family of the Trypanosomatidae in a target comprising administering to the target an effective amount of a compound of formula (1):
R2-A-R,,
wherein:
- A represents a group chosen from a phenyl, a triazolyl, and a thienyl,
- R, represents -CO-CH2OH or -CO-NHOH,
- R2 is chosen from H, triazolyl, methyltriazolyl, -CONHR3, -CH2-CONHR3, - NHS02NHR3, -CH2-NHS02NHR3, wherein R3 represents H, a (CrC8) alkyl group, an aryl group, a (C3-C8) cycloalkyl group, a heterocyclic group, or a heteroaryl group, or a pharmaceutically acceptable salt thereof.
12. Method according to claim 1 1, wherein the protozoan parasite of the family of the Trypanosomatidae is leishmania parasite, in particular Leishmania. infantum, Leishmania. tropica or Leishmania. Major, or trypanosoma parasite, in particular Trypanosoma, brucei. gambiense.
PCT/EP2011/004055 2010-08-12 2011-08-12 Method for treating protozoan parasitic diseases WO2012019772A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US37294510P 2010-08-12 2010-08-12
US61/372945 2010-08-12

Publications (1)

Publication Number Publication Date
WO2012019772A1 true WO2012019772A1 (en) 2012-02-16

Family

ID=44512783

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2011/004055 WO2012019772A1 (en) 2010-08-12 2011-08-12 Method for treating protozoan parasitic diseases

Country Status (1)

Country Link
WO (1) WO2012019772A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2826769A1 (en) * 2013-07-18 2015-01-21 Institut de Recherche pour le Développement ( IRD) Compounds for the treatment and/or prevention of parasitic diseases and method of production thereof
WO2017030728A1 (en) 2015-08-17 2017-02-23 Southwestern Oklahoma State University Compositions comprising macrocycle derivatives incorporating bridged macrocycles and methods of producing and using same

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1993007148A1 (en) * 1991-10-04 1993-04-15 Sloan-Kettering Institute For Cancer Research Novel potent inducers of terminal differentiation and methods of use thereof
US20040122079A1 (en) * 2002-12-16 2004-06-24 Adelbert Grossmann Thiophene hydroxamic acid derivatives
EP1541549A1 (en) * 2003-12-12 2005-06-15 Exonhit Therapeutics S.A. Tricyclic hydroxamate and benzaminde derivatives, compositions and methods
US20060252834A1 (en) * 2003-08-26 2006-11-09 Amorepacific Corporation Hydroxamic acid derivatives and the method for preparing thereof
US20080254130A1 (en) * 2003-08-29 2008-10-16 Bioderm Research Skin Antiaging & Brightening via Multi-function Treatment of Enzyme Dysfunction

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1993007148A1 (en) * 1991-10-04 1993-04-15 Sloan-Kettering Institute For Cancer Research Novel potent inducers of terminal differentiation and methods of use thereof
US20040122079A1 (en) * 2002-12-16 2004-06-24 Adelbert Grossmann Thiophene hydroxamic acid derivatives
US20060252834A1 (en) * 2003-08-26 2006-11-09 Amorepacific Corporation Hydroxamic acid derivatives and the method for preparing thereof
US20080254130A1 (en) * 2003-08-29 2008-10-16 Bioderm Research Skin Antiaging & Brightening via Multi-function Treatment of Enzyme Dysfunction
EP1541549A1 (en) * 2003-12-12 2005-06-15 Exonhit Therapeutics S.A. Tricyclic hydroxamate and benzaminde derivatives, compositions and methods

Non-Patent Citations (16)

* Cited by examiner, † Cited by third party
Title
BRUN, SCHONENBERGER, ACTA TROPICA, vol. 36, no. 3, 1979, pages 289 - 292
CLARKSON A B ET AL: "Trypanosoma brucei brucei: A systematic screening for alternatives to the salicylhydroxamic acid-glycerol combination", MOLECULAR AND BIOCHEMICAL PARASITOLOGY, ELSEVIER SCIENCE PUBLISHERS, AMSTERDAM, NL, vol. 3, no. 5, 1 September 1981 (1981-09-01), pages 271 - 291, XP023700200, ISSN: 0166-6851, [retrieved on 19810901], DOI: 10.1016/0166-6851(81)90002-5 *
CUNNINGHAM, J PROTOZOOL, vol. 21, no. 2, 1977, pages 325 - 329
DA LUZ ET AL., ANTIMICROB AGENTS CHEMOTHER, vol. 53, no. 12, 2009, pages 5197 - 5203
DATABASE CA [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; CLARKSON ET AL, XP002666516, Database accession no. 1983:402939 *
DATABASE CA [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; EXONHIT THERAPEUTICS S.A., FR., XP002666519, Database accession no. 2005:516308 *
DATABASE CA [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; GUPTA, SHYAM K.: "Skin antiaging and brightening via multi-function topical treatment of enzyme dysfunction comprising regulating agents for extracellular matrix metalloprotease, intracellular ubiquitin-proteasome, and epidermal melanocytes", XP002666518, retrieved from STN Database accession no. 2008:1251559 *
DATABASE CA [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; SIVAKUMAR, PONNURENGAM MALLIAPPAN ET AL: "Experimental and QSAR of acetophenones as antibacterial agents", XP002666517, retrieved from STN Database accession no. 2008:1284199 *
DATABASE CA CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; PRADO-PRADO ET AL, XP002666515, Database accession no. 2010:304839 *
GRADY R W ET AL: "p-alkyloxybenzhydroxamic acids, effective inhibitors of the trypanosome glycerol-3-phosphate oxidase", MOLECULAR AND BIOCHEMICAL PARASITOLOGY, ELSEVIER SCIENCE PUBLISHERS, AMSTERDAM, NL, vol. 19, no. 3, 1 June 1986 (1986-06-01), pages 231 - 240, XP023700980, ISSN: 0166-6851, [retrieved on 19860601], DOI: 10.1016/0166-6851(86)90005-8 *
HEUNG SOO BAEK ET AL: "The Inhibitory Effect of New Hydroxamic Acid Derivatives on Melanogenesis", BULLETIN OF THE KOREAN CHEMICAL SOCIETY, vol. 29, no. 1, 20 January 2008 (2008-01-20), pages 43 - 46, XP055015612, ISSN: 0253-2964, DOI: 10.5012/bkcs.2008.29.1.043 *
PRADO-PRADO F J ET AL: "Multi-target spectral moment QSAR versus ANN for antiparasitic drugs against different parasite species", BIOORGANIC & MEDICINAL CHEMISTRY, PERGAMON, GB, vol. 18, no. 6, 15 March 2010 (2010-03-15), pages 2225 - 2231, XP026941354, ISSN: 0968-0896, [retrieved on 20100206], DOI: 10.1016/J.BMC.2010.01.068 *
SIVAKUMAR, PONNURENGAM MALLIAPPAN ET AL: "Experimental and QSAR of acetophenones as antibacterial agents", CHEMICAL BIOLOGY & DRUG DESIGN , 72(4), 303-313 CODEN: CBDDAL; ISSN: 1747-0277, 2008 *
TABERNERO E ET AL: "ANTITRYPANOSOMAL AND ANTIMYCOTIC EFFECT OF VARIOUS HYDROXAMIC ACIDS", ACTA CIENTIFICA VENEZOLANA, ASOCIACION VENEZOLANA PARA EL AVANCE DE LA CIENCIA, CARACAS, VE, vol. 32, 1 January 1981 (1981-01-01), pages 411 - 416, XP000874939, ISSN: 0001-5504 *
TORU TANAKA ET AL: "[alpha]-Hydroxyketones as inhibitors of urease", BIOORGANIC & MEDICINAL CHEMISTRY, vol. 12, no. 2, 1 January 2004 (2004-01-01), pages 501 - 505, XP055015610, ISSN: 0968-0896, DOI: 10.1016/j.bmc.2003.10.017 *
WITTER ET AL: "Benzo[b]thiophene-based histone deacetylase inhibitors", BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, PERGAMON, ELSEVIER SCIENCE, GB, vol. 17, no. 16, 17 July 2007 (2007-07-17), pages 4562 - 4567, XP022181867, ISSN: 0960-894X, DOI: 10.1016/J.BMCL.2007.05.091 *

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2826769A1 (en) * 2013-07-18 2015-01-21 Institut de Recherche pour le Développement ( IRD) Compounds for the treatment and/or prevention of parasitic diseases and method of production thereof
WO2015007870A1 (en) * 2013-07-18 2015-01-22 Institut De Recherche Pour Le Developpement (I.R.D.) New compounds for the treatment and/or prevention of parasitic diseases and method of production of thereof
US10106493B2 (en) 2013-07-18 2018-10-23 Institut De Recherche Pour Le Developpement (I.R.D.) N-hydroxybenzamides as HDAC inhibitors for the treatment of parasitic diseases
WO2017030728A1 (en) 2015-08-17 2017-02-23 Southwestern Oklahoma State University Compositions comprising macrocycle derivatives incorporating bridged macrocycles and methods of producing and using same
EP3337519A4 (en) * 2015-08-17 2019-01-09 Southwestern Oklahoma State University Compositions comprising macrocycle derivatives incorporating bridged macrocycles and methods of producing and using same
AU2016307752B2 (en) * 2015-08-17 2020-11-12 Southwestern Oklahoma State University Compositions comprising macrocycle derivatives incorporating bridged macrocycles and methods of producing and using same
US10927108B2 (en) 2015-08-17 2021-02-23 Southwestern Oklahoma State University Compositions comprising macrocycle derivatives incorporating bridged macrocycles and methods of producing and using same

Similar Documents

Publication Publication Date Title
Loo et al. Artemisinin and its derivatives in treating protozoan infections beyond malaria
Monzote Current treatment of leishmaniasis: a review
CA2548313A1 (en) Methods and compositions for the prevention and treatment of inflammatory diseases or conditions
EP0648121B1 (en) Use of rifamycin derivatives for the manufacture of a medicament for the treatment of toxoplasmosis
WO2012019772A1 (en) Method for treating protozoan parasitic diseases
Oliveira-Silva et al. Antileishmanial activity of azithromycin against Leishmania (Leishmania) amazonensis, Leishmania (Viannia) braziliensis, and Leishmania (Leishmania) chagasi
CA3191534A1 (en) Senolytic compounds and compositions
EP1768669B1 (en) Pharmaceutical compositions for the treatment of leishmaniasis
CN114073772A (en) Use of an iron chelator for the treatment or prevention of polyoma viral infections
US20060014723A1 (en) Anti-parasitic uses of borinic acid complexes
CA2657375A1 (en) Combination methods of treating cancer
KR101985087B1 (en) Composition for preventing or treating muscle atrophy or sarcopenia comprising sabinene
KR20200018681A (en) Combination of MCL-1 Inhibitors and Standard Therapeutic Agents for Hematological Cancer, Uses thereof, and Pharmaceutical Compositions
KR20100137442A (en) Combination of a bis-thiazolium salt or a precursor thereof and artemisinin or a derivative thereof for treating acute malaria
CN112020356B (en) Combination of quinoline-4-carboxamides and benzonaphthyridine derivatives as antimalarial drug combinations
EP3884938A1 (en) 1,2,4-trioxane compounds and compositions comprising the same for use in the treatment of covid-19
WO2009063241A1 (en) 3-hydroxyanthranilic acid or salts thereof1 for treating cancer or infections
KR100956576B1 (en) Composition for protecting hearing cells comprising Epicatechin
US10610538B2 (en) Pradimicin derivatives for the treatment of diseases caused by kinetoplastids
KR102498802B1 (en) Pharmaceutical composition for preventing or treating malaria comprising fucoidan
CN117460506A (en) Antimalarial endoperoxides for the treatment of myelodysplastic syndromes
WO1998011883A1 (en) Use of hexahydrolupulones as antibacterial agents
US5610179A (en) Method of treating babesiosis
WO2016038238A1 (en) Use of ester derivatives of pyrazole proton-ionizable compounds and the corresponding salts thereof for the treatment of chagas disease and leishmaniasis
KR100903287B1 (en) Composition comprising 7-ethoxycoumarin for preventing and treating toxoplasmosis

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: 11748895

Country of ref document: EP

Kind code of ref document: A1

DPE1 Request for preliminary examination filed after expiration of 19th month from priority date (pct application filed from 20040101)
NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 11748895

Country of ref document: EP

Kind code of ref document: A1