WO2015101918A1 - Antileishmanial compositions containing fullerol and use thereof - Google Patents

Antileishmanial compositions containing fullerol and use thereof Download PDF

Info

Publication number
WO2015101918A1
WO2015101918A1 PCT/IB2014/067384 IB2014067384W WO2015101918A1 WO 2015101918 A1 WO2015101918 A1 WO 2015101918A1 IB 2014067384 W IB2014067384 W IB 2014067384W WO 2015101918 A1 WO2015101918 A1 WO 2015101918A1
Authority
WO
WIPO (PCT)
Prior art keywords
fullerol
pharmaceutical compositions
liposomes
leishmaniasis
glucantime
Prior art date
Application number
PCT/IB2014/067384
Other languages
French (fr)
Portuguese (pt)
Inventor
Frederic Jean Georges Frezard
Maurício VELOSO BRANT PINHEIRO
Maria NORMA MELO
Paula PEIXOTO CAMPOS
Marina DE SOUZA LADEIRA
Luiz Orlando Ladeira
Guilherme SANTOS RAMOS
Silvia CAROLINA GUATIMOSIN FONSECA
Kelly CRISTINA KATO
Ricardo TOSHIO FUJIWARA
Priscila GOMES DOS REIS
Original Assignee
Universidade Federal De Minas Gerais - Ufmg
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
Priority claimed from BR102013033866A external-priority patent/BR102013033866A2/en
Priority claimed from BR102014032447-0A external-priority patent/BR102014032447B1/en
Application filed by Universidade Federal De Minas Gerais - Ufmg filed Critical Universidade Federal De Minas Gerais - Ufmg
Publication of WO2015101918A1 publication Critical patent/WO2015101918A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/045Hydroxy compounds, e.g. alcohols; Salts thereof, e.g. alcoholates
    • 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
    • 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

  • fullerene-containing pharmaceutical compositions preferably fullerol, which are spherical carbon nanostructures, as novel tools for the treatment of visceral and cutaneous leishmaniasis.
  • Fullerol-containing antileishmania compositions may comprise yet another leishmanicidal or immunomodulatory drug, either in free form or in association with a nanocarrier such as liposomes, which confers pharmacokinetic properties more favorable to the active ingredients.
  • Fullerol has surprisingly demonstrated an antileishmania activity as well as an ability to reduce meglumine antimoniate hepatotoxicity when administered in combination with an animal model of visceral leishmaniasis.
  • fullerol was the first drug to demonstrate the dual ability to exert antileishmania action without side effects and to reduce the toxicity of another leishmanicidal drug without interfering with its pharmacological activity.
  • the invention comprises the use of pharmaceutical compositions containing fullerenes for the treatment of leishmaniasis.
  • Leishmaniasis is among the most neglected tropical diseases in the world, according to the resolution of the 60th World Health Assembly in 2007. Epidemiologically, over 12 million people are infected worldwide, with 1, 3 million new people. cases reported each year, with about 20 to 30,000 deaths annually, whose illness affects the poorest populations in 88 countries (developing countries) (World Health Organization - WHO. Report of the Fifth Consultative Meeting on Leishmania / HIV Coinfection). Addis Ababa, Ethiopia, March 20 - 22, 2007; WHO Leishmaniasis, Feb. 201 3. Available at: http://www.who.int/mediacenter/factsheets/fs375/en/# Accessed: December 16, 2013).
  • Leishmaniasis are parasitic diseases caused by more than 20 species of protozoa of the genus Leishmania spp., Of the Trypanosomatidae family, hemoflagellate parasites showing preference for phagolysosomes of the mononuclear phagocytic system. Are transmitted to humans by the bite of the vector, females of the genera Phlebotomo sp. and Lutzomyia sp. infected. The disease presents with three different clinical manifestations: visceral, or commonly known as calazar; the cutaneous; and the mucocutaneous. (RICCIARDI, LV Antileishmania Therapy: Reviewing the Past, the Present, and the Future. Gac. Med. Caracas, v.1 17, no. 2, p.93-111, 2009).
  • Cutaneous leishmaniasis is the most common form of leishmaniasis and presents with ulcers distributed on the exposed parts of the body, leaving lifelong scarring and severe disability. About 1/3 of the cases occur in the Americas, the Mediterranean basin, Central Asia and the Middle East. Mucocutaneous leishmaniasis leads to mutilating mucosal lesions, which consist of partial or total destruction of the mucous membranes of the nose, mouth and throat, most of them reported in Cambodia, Brazil and Peru (World Health Organization - WHO. Leishmaniasis. Feb. 2013. Available at: http: //www.who.int/mediacentre/factsheets/fs375/en/ # Accessed: December 16, 2013).
  • the parasite's life cycle is metaxenic, where the agent undergoes transformations in the organism of the phlebotomine vector, the dipterous insect.
  • the flagellated protozoan L. infantum is found in the mobile metacyclic promastigote form in the vector, and when phagocytized by the vertebrate host macrophages it becomes the immovable amastigote form.
  • the cycle begins with the inoculation of infectious promastigote forms in the vertebrate host during blood meal. Upon reaching the bloodstream, promastigote forms use specific mechanisms to survive cell lysis, which will be activated by the complement system.
  • the parasite survives host attack and can still invade macrophages by manipulating cell receptors (CAMPOS-PONCE M, PONCE C, PONCE E, MAINGON RDC.
  • Leishmania chagasi / infantum further investigations on Leishmania tropism in atypical cutaneous and visceral leishmaniasis foci in Central America. Parasitology, New York, v. 109, p. 209-219, 2005).
  • the parasite is protected against the host immune response, residing in the acidic environment of the secondary macrophage lysosome where it undergoes successive multiplications. Once infected, the host becomes the parasite's reservoir.
  • visceral leishmaniasis presents as a chronic, widespread disease characterized by irregular and long-lasting fever, hepatosplenomegaly, lymphadenopathy, anemia with leukopenia, hypergammaglobulinemia, and progressive debilitation leading to cachexia and even death.
  • the evolution of clinical forms is diverse, ranging from spontaneous cure, oligosymptomatic and asymptomatic forms, to severe manifestations, and may reach lethality between 10% and 98% in inadequately treated and untreated cases.
  • Antimonial compounds were first used in the treatment of leishmaniasis in 1912 by Brazilian physician Gaspar Vianna, initially in the trivalent form. Although pentavalent antimonials such as meglumine antimoniate (Glucantime®, Sanofi-Aventis) were recognized as clinically effective in 1947, they are still the drug of choice for all leishmaniasis in most developing countries (BERMAN JD. Human leishmaniasis: clinical, diagnostic, and chemotherapeutic developments in the last 10 years (Lin. Infect. Dis. 24, 684, 1997). Amphotericin B and its various formulations are used as second-choice drugs (Ministry of Health. Visceral Leishmaniasis Surveillance and Control Manual. Bras ⁇ lia, 2003)
  • Glucantime® is contraindicated in patients taking beta-blockers and antiarrhythmic drugs with renal or hepatic impairment in pregnant women in the first two trimesters of pregnancy. There are no absolute contraindications to the use of amphotericin B; however, renal complications limit its use (Ministry of Health. Visceral Leishmaniasis Surveillance and Control Manual. Bras ⁇ lia, 2003).
  • WHO World Health Organization
  • the drugs that may replace pentavalent antimonials include liposomal amphotericin B (AmBisome®), miltefosine (Impávido®) and paromomycin or aminosidine.
  • a successful strategy to increase the therapeutic index of drugs against visceral leishmaniasis refers to their reversible association with a carrier nanosystem, allowing them to extend their half-life in the body and direct them to the sites of infection (FRÉZARD, DEMICHELI, C. New delivery strategies for the old antimonial pentavalent drugs Expert Opinion on Drug Delivery, v. 7, pp. 1343-1358, 2010).
  • liposomes occupy a prominent position. These spherical vesicles, consisting of one or more concentric lipid bilayers, may store in their internal aqueous compartment water-soluble active principles or have lipophilic or amphiphilic active principles incorporated into their membranes.
  • Conventional liposomes are typically formed of a phospholipid, such as phosphatidylcholine, or a nonionic surfactant. They may also include in its composition cholesterol, a negatively charged phospholipid, such as phosphatidylglycerol, phosphatidic acid, diketylphosphate, and / or a positively charged phospholipid such as esterarylamine.
  • Fullerenes were discovered in 1985 (ADAMS, PD, GROSSEKUNSTLEVE, RW, HUNG, L.-W., et al. C60: buckminsferfullerene. Nature, v. 318, p.162-163, 1985) and they represent a large family of spherical carbon nanostructures, having as a classic example the C 6 molecule called fullerene. Due to their chemical and physical properties, these molecules have become attractive targets for use in medicine (REBECCA, M.; HSING-LIN, W .; JUN.G., et al. Impact of physicochemical properties of engineered fullerenes on key. , Toxic. And App. Pharmac, v.234, p.58-67, 2009).
  • fullerene in the biomedical area has been limited by the hydrophobic character of the molecule.
  • a class of polyhydroxylated fullerenes also called fullerenol or fullerol, formed by chemical modification was developed.
  • the solubility of each fullerenol molecule is dependent on the number of hydroxyl groups introduced (KOKUBO, K. Water-Soluble Single-Nano Carbon Particles: Fullerenol and Its Derivatives. The Delivery of Nanoparticles. ⁇ Available at: http://www.intechopen .com / download / get / type / pdfs / id / 36889> Accessed December 18, 2013).
  • a water-soluble polyhydroxylated fullerene derivative, fullerenel has shown antiviral activity in acute-infected human peripheral mononuclear cells (PBMC) and chronically HIV-1 virus-infected H9 cells with an EC50 of 7.3 ⁇ 5, 9 ⁇ and 10.8 ⁇ 8.2 ⁇ , respectively.
  • PBMC peripheral mononuclear cells
  • Fullerol has been shown to act as a neuroprotective drug in oxidative stress-related degenerative diseases (DORDEVIC, A. and BOGDANOVIC, G. Fullerenol - a new nanopharmaceutic? Arch. Oncol., V.16, p.42-5 , 2008).
  • DORDEVIC oxidative stress-related degenerative diseases
  • BOGDANOVIC oxidative stress-related degenerative diseases
  • the mechanism of fullerenol-mediated neuroprotection occurs by inhibiting glutamate receptors, and also by reducing glutamate release via increased intracellular calcium concentrations, a critical mechanism of excitotoxicity in neurons (SILVA, GA). the central nervous system, Surgical Neurology, v. 63, pp. 301-306, 2005).
  • Fullerol is not known to have acute or subacute toxicity in rodents and protects tissues from cellular damage caused by ROS following exposure to pro-oxidant toxic substances including some drugs by absorbing these free radicals that would cause tissue injury. and cellular (BOSI, S .; DA ROS, T .; SPALLUTO, G., et al. Fullerene derivatives: an attractive tool for biological applications. European Journal of Medicinal Chemistry, v. 38, p. 913-23, 2003) .
  • a patent issued entitled "FULLERENE PHARMACEUTICAL COMPOSITIONS FOR PREVENTING OR TREATING DISORDERS” (US 6,777,445 B2) relates to a method for the prevention and treatment of bacterial infections caused by gram-positive and gram-negative bacteria.
  • viral diseases such as dengue and viral encephalitis comprising administering pharmaceutical compositions containing a fullerene, which is administered in vivo in an amount from about 0.001 to about 100 mg / kg body weight of the subject.
  • a single water-soluble fullerene derived from the symmetrical C60 (C3) structural formula carboxylic acid differing primarily from the present invention in that it is a fullerene with addition of 22-24 hydroxyls on the surface, is also not present.
  • fullerol for the treatment of visceral and cutaneous leishmaniasis.
  • this invention claims pharmaceutical compositions containing fullerenes as novel antileishmania drugs and their use in combination with leishmanicidal or immunomodulatory drug (s) to enhance their efficacy and reduce their toxicity.
  • Figure 1 comparatively shows the parasitic burden on the liver of BALB / c mice experimentally infected with Leishmania infantum (murine model of visceral leishmaniasis). The amount of parasites was determined by quantitative real-time PCR after 20 days of intraperitoneal administration with daily doses of Glucantime (120 mg Sb / kg), free fullerol (0.05 mg / kg), liposome-encapsulated fullerol. (0.05 mg / kg) and saline (negative control).
  • Figure 3 shows the efficacy of treatment combining fullerol with Glucantime in a murine model of visceral leishmaniasis, with fullerol being administered in free or liposome-encapsulated form.
  • Figure 3 presents the results of histopathological analysis and apoptosis by light microscopy using the HE liver staining method of Leishmania infantum experimentally infected BALB / c mice after 20 days of intraperitoneal Glucantime treatment. (120 mg Sb / kg / day), Glucantime (120 mg Sb / kg / day) associated with Fullerol (0.05 mg / kg / dose / 4 days), Glucantime (120 mg Sb / kg / day) associated with Fullerol in liposomes (0.05 mg / kg / dose / 4 days) or saline.
  • the histopathological parameters evaluated were hydropic degeneration (A) and apoptotic index (B).
  • Figure 6 shows the antileishmania activity of different doses of liposome-encapsulated fullerol in the liver of L. infantum infected BALB / c mice.
  • A parasitic load in the liver of L. infantum infected BALB / c mice determined by quantitative PCR after 20 days of intraperitoneal treatment with Glucantime (120 mg Sb / kg / day), free fullerol (0.05 mg / day). kg / 4 days) and liposome-encapsulated fullerol (0.05 and 0.2 mg / kg / 4 days) and empty liposomes (same lipid dose as in the groups receiving encapsulated fullerol).
  • Figure 7 shows the antileishmania activity of different doses of liposome-encapsulated fullerol in the spleen of L. infantum infected BALB / c mice.
  • Parasitic burden on liver of L. infantum infected BALB / c mice determined by quantitative PCR after 20 days of intraperitoneal treatment with liposome-encapsulated fullerol (0.05 and 0.2 mg / kg / 4 days), Glucantime ( 120 mg Sb / kg / day), free fullerol (0.05 mg / kg / 4 days), empty liposomes (same dose of lipid as in the groups receiving encapsulated fullerol) and saline.
  • Figure 8 shows the liver cytokine profile of L. infantum infected BALB / c mice following treatment with different doses of liposome-encapsulated fullerol.
  • the present invention relates to fullerene-containing pharmaceutical compositions as novel tools for the treatment of visceral and cutaneous leishmaniasis.
  • Such compositions preferably comprise water-soluble fullerenes of 60 to 120 carbons with the addition of hydroxyl groups such as fullerol C 6 oOH 2 2-24.
  • Antileishmania compositions containing fullerol may comprise the drug in combination with other drug (s). leishmanicide (s) or immunomodulator (s), either in free form or in association with a nanocarrier, such as liposomes, which gives the active principles more favorable pharmacokinetic properties.
  • the invention comprises the use of pharmaceutical compositions containing fullerenes for the treatment of leishmaniasis.
  • Fullerol was synthesized according to the process already described in the filed patent BR 0404543-2 A2, entitled "LARGE-SCALE CONTINUOUS PRODUCTION PROCESS AND SYSTEM OF CONSTITUTED DEVICES TO PERFORM THE PROCESS".
  • Fullerol was also encapsulated in liposomes formed of phosphatidylcholine, cholesterol and phosphatidylglycerol in a 5: 4: 1 molar ratio using the dehydration-rehydration method (FRÉZARD, F. and SCHETTINI, DA.
  • Liposomes physicochemical and pharmacological properties, chemotherapy applications. based on antimony Quim Nova, V. 28, No. 3, p.51 1 -518, 2005). However, these may be formed of cholesterol and a negatively or positively charged lipid in the presence or absence and polymers and / or ligands coupled to their surfaces.
  • Encapsulation was followed by calibration of vesicle size by extrusion through 200 nm pore polycarbonate membrane (NAYAR, R., HOPE, MJ, CULLIS, PR. Generation of large unilamellar vesicles from long-chain saturated phosphatidylcholines. by extrusion technique, Biochim, Biophys (Acta, v. 986, pp. 200-206, 1989). in the presence or absence of coupled polymers and / or binders on their surfaces.
  • fullerol may be associated with carrier nanosystems and / or controlled release drug systems, aiming at increased stability, increased bioavailability, longer action and / or targeting to the target tissue.
  • carrier or controlled release systems include, but are not limited to, cyclodextrins, polymers, mucoadhesive polymers, lipid vesicles, liposomes, polymersomes, solid lipid nanoparticles, polymeric micro- and nanoparticles, micro- and nanocapsules, micro- - and nanoemulsions, dendrimers, micelles, polymer micelles, inorganic nanoparticles, carbon nanoparticles, transdermal patches, implantable polymer matrices and implantable devices.
  • fullerol has antioxidant properties
  • the murine visceral leishmaniasis model was used to evaluate, by quantitative real-time PCR, the parasitic load on the liver of the animals after the treatments. A significant reduction in hepatic parasitic load was observed in the groups treated with Glucantime and Glucantime associated with liposome-encapsulated Fullerol when compared to the control group. However, the association between fullerol and Glucantime appeared to be more effective in treating leishmaniasis compared to the effect of Fullerol or Glucantime alone.
  • the present invention also relates to pharmaceutical compositions comprising fullerenes associated with one or more leishmanicidal drugs and these are: pentavalent antimonials, preferably meglumine antimoniate; amphotericin B; allopurinol; ketoconazole; itraconazole; fluconazole; posaconazole; tucaresol; paramomycin; pentamidine; miltefosine, sitamaquine, iminoquimod; azithromycin; buparvaquone; tamoxifen; terbinafine; furazolidone; fluoroquinolone; domperidone; alkyl lysophospholipid or alkyl phospholipid derivative; aza-sterol; licocalcona A; maesabalid III; trichothecenes; n-acetyl cysteine; 3-substituted quinoline, or one or more immunomodulatory substances, preferably Leishmania-derived immunogenic peptides or proteins
  • Livers from animals with visceral leishmaniasis submitted to antimonial chemotherapy were also used for the analysis of histopathological changes and apoptosis using the HE staining method by optical mischroscopy. Histopathological changes characteristic of hydropic degeneration were observed in the livers of animals that received Glucantime alone, showing the hepatotoxicity of the drug that can be attributed to the induction of oxidative stress. However, these changes were not observed in the group that received the combination Glucantime + liposome-encapsulated fullerol, which demonstrates the hepatoprotective action of co-treatment with fullerol in nanocarrier system.
  • fullerol has surprisingly demonstrated an antileishmania activity as well as an ability to reduce the hepatotoxicity caused by Glucantime when given in combination in animal model for visceral leishmaniasis. Therefore, fullerol was the first drug to demonstrate the dual ability to exert antileishmania action without side effects and to reduce the toxicity of another leishmanicidal drug without interfering with its pharmacological activity.
  • the set of test results allowed us to anticipate that pharmaceutical compositions containing the antimonial drug and fullerol, especially those based on liposomes, have higher antileishmania activity and reduced toxicity when compared to the commercial drug Glucantime.
  • compositions containing pure water soluble fullerenes or associated with leishmanicidal or immunomodulatory drugs may further be administered parenterally, preferably intravenously, intramuscularly, subcutaneously, intralesionally and intraperitoneally; orally, preferably in the form of capsules, dragees, tablets, syrups or elixir; topical, preferably in the form of ointment, lotion, paste or gel.
  • compositions particularly comprise fullerol associated with liposome-encapsulated meglumine antimoniate.
  • Figure 1 shows a significant reduction in hepatic parasitic burden of animals treated with liposome-encapsulated Glucantime, Fulerol and Fullerol at a dose of 0.05 mg / kg ( * P ⁇ 0.05 compared to the control group; Kruskal-Wallis test with Dunn's post-test's). Therefore, we established for the first time the anti-leishmania activity of fullerol in an experimental leishmaniasis model, which was equivalent to that of the commercial drug Glucantime.
  • Figure 2 shows a significant reduction in hepatic parasite burden only in the groups treated with Glucantime and Glucantime associated with liposome-encapsulated fullerol when compared to the control group (* P ⁇ 0.05 with respect to the saline group; Kruskal-Wallis test with post-test Dunn's).
  • the association between Fullerol and Glucantime appeared to be more effective in treating leishmaniasis compared to Fullerol or Glucantime alone. Therefore, this example establishes the benefit of combining Glucantime with liposome-encapsulated fullerol in terms of improved efficacy in the treatment of visceral leishmaniasis.
  • Example 3 Chemoprotective effect of fullerol in a visceral leishmaniasis model submitted to antimonial chemotherapy
  • Histopathological and apoptotic analysis was performed by light microscopy using the HE liver staining method of experimentally infected Leishmania infantum infected BALB / c mice after 20 days of intraperitoneal Glucantime treatment (120 mg Sb / kg / kg).
  • Glucantime 120 mg Sb / kg / day
  • fullerol 0.05 mg / kg / dose / 4 days
  • Glucantime 120 mg Sb / kg / day associated with fullerol liposomes (0.05 mg / kg / dose / 4 days) or saline.
  • the histopathological parameters evaluated were hydropic degeneration (A) and apoptotic index (B).
  • Figure 3 shows characteristic pathological changes of hydropic degeneration and increased apoptotic index in the liver of animals receiving Glucantime showing the hepatotoxicity of the drug. These changes were also observed in the group that received the Glucantime + fullerol combination, but not in the group that received the liposome-encapsulated Glucantime + fullerol combination, demonstrating the hepatoprotective action of co-treatment with liposome-encapsulated fullerol. That is, besides exerting antileishmania action, fullerol in liposomes showed a chemoprotective action, evidenced by the reduction of Glucantime hepatotoxicity in the presence of fullerol in liposomes.
  • peritoneal macrophages were isolated from BALB / c mice 3 days after intraperitoneal injection of thioglycolate 3% by peritoneal lavage with cold RPMI medium. . The cells were washed, counted and a resulting suspension of 1x10 6 cells / ml density was obtained. Cells were maintained at 37 ° C in RPMI 1640 medium supplemented with 10% fetal bovine serum, streptomycin (50 pg / mL) and penicillin (1000 U / mL).
  • the MTT assay is based on the reduction of tetrazolium by the action of viable mitochondrial monoxygenases resulting in formazan formation (Mosman, 1983). After formation of formazan crystals, they were solubilized in DMSO and read on an Elisa reader at 570 nm. Results were shown as absorbance values and controls (not exposed to antimonial and / or fullerol).
  • Leishmania infantum promastigote cells (strain BH46) were maintained in minimal essential culture medium (a-MEM, Gibco) supplemented with 10% inactivated fetal bovine serum, 100 ⁇ g / mL kanamycin and 50pg / mL ampicillin and incubated at 24 ⁇ 1 ° C and at pH 7.0 in an oven BOD the promastigotes in logarithmic growth phase were placed into 96 - well plates at a density of 10 6 cells / ml, exposed to increasing concentrations of 0.005 fulerol at 0.2 mg / ml. Replicates in the absence of the drug were established as controls. After 72 hours, growth was evaluated by measuring the absorbance of the cultures at 600nm using an automated microplate reading system.
  • fullerol antileishmania activity was used at a maximum non-cytotoxic concentration (0.12 mg / ml) in the Leishmania infantum-infected macrophage model (strain BH46). As shown in Figure 5, fullerol alone reduced the parasite burden by about 75% compared to the control group (dark bar). The effect was equivalent to that of Glucantime given at a concentration of Sb 0.75 mg / ml.
  • Group 1 treated with glucantime (120mg Sb / kg / day - 20 doses);
  • Group 2 treated with free fullerol in 0.15 M NaCl solution (0.05mg / kg / 4 days
  • Group 3 treated with 0.15 M NaCl solution containing liposome-encapsulated fullerol (0.05mg / kg / 4 days - 5 doses);
  • Group 4 treated with 0.15 M NaCI solution containing liposome encapsulated fullerol (0.2mg / kg / 4 days - 5 doses);
  • Group 5 treated with empty liposome solution (same lipid dose as in groups 4 and 5);
  • cytokines IL-4, IL-10, TNF-alpha and IFN-gamma
  • Figure 6 (A and B) and Figure 7 show the results of parasitic burden on the liver and spleen, respectively. Significant reduction in parasite load in both liver and spleen was observed in the liposome-encapsulated fullerol (0.05 and 0.2 mg / kg) and Glucantime treated groups compared to the saline group. It is also interesting to note that free fullerol administered every 4 days did not significantly reduce parasite load and that the encapsulated form was significantly more effective than free form.
  • liposomal fullerol treatment was significantly more effective than conventional Glucantime treatment.
  • TNF-alpha is a cytokine known to stimulate hepatocytes to synthesize and secrete acute inflammatory phase proteins (mannose-linked protein, C-reactive protein) that serve to activate phagocytic complement and chemotaxis system (dendritic cells, macrophages and neutrophils).
  • Liposomal fullerol administered at a dose of 0.2 mg / kg / 4 days promotes a marked reduction in parasite burden in the liver and spleen in a murine model of visceral leishmaniasis, and complete elimination of the parasite in the liver;
  • the antileishmania activity of the liposomal formulation is much higher than that of the standard drug (Glucantime);
  • the efficacy of the liposomal formulation in the liver seems to result from the induction of proinflammatory immune profile;
  • the liposomal formulation of fullerol may find applications in the treatment of other conditions affecting the liver and for which a proinflammatory immune profile would be beneficial.

Landscapes

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

Abstract

The present invention relates to pharmaceutical compositions containing fullerenes, preferably fullerol, which are spherical carbon nanostructures, as novel tools for the treatment of visceral and cutaneous leishmaniasis. The antileishmanial compositions containing fullerol can further comprise another leishmanicidal drug or immunomodulator, in free form or in combination with a nanocarrier, such as liposomes, which improves the pharmacokinetic proprieties of the active principles. Surprisingly, fullerol had antileishmanial activity and was able to reduce hepatotoxicity of meglumine antimoniate when administered in combination therewith in an animal model of visceral leishmaniasis. Fullerol therefore proved to be the first drug capable both of exerting antileishmanial activity without side effects and reducing the toxicity of another leishmanicidal drug without interfering in the pharmacological activity thereof. Finally, the invention comprises the use of pharmaceutical compositions containing fullerenes for the treatment of leishmaniasis.

Description

COMPOSIÇÕES ANTILEISHMANIA CONTENDO O FULEROL E USO  ANTILEISHMANY COMPOSITIONS CONTAINING FULEROL AND USE
[001 ] A presente invenção refere-se às composições farmacêuticas contendo fulerenos, preferencialmente o fulerol, que são nanoestruturas esféricas de carbono, como novas ferramentas para o tratamento das leishmanioses visceral e cutânea. As composições antileishmania contendo o fulerol podem compreender ainda um outro fármaco leishmanicida ou imunomodulador, estando na forma livre ou associada a um nanocarreador, tal como lipossomas, que confere propriedades farmacocinéticas mais favoráveis aos princípios ativos. O fulerol demonstrou de forma surpreendente uma atividade antileishmania, assim como uma capacidade de reduzir a hepatotoxicidade do antimoniato de meglumina quando administrado em associação em modelo animal de leishmaniose visceral. Desta forma, o fulerol apresentou-se como o primeiro fármaco a demonstrar a dupla capacidade de exercer ação antileishmania sem efeitos colaterais e de reduzir a toxicidade de outro fármaco leishmanicida sem interferir na sua atividade farmacológica. Por fim, a invenção compreende o uso de composições farmacêuticas contendo fulerenos para o tratamento das leishmanioses.  The present invention relates to fullerene-containing pharmaceutical compositions, preferably fullerol, which are spherical carbon nanostructures, as novel tools for the treatment of visceral and cutaneous leishmaniasis. Fullerol-containing antileishmania compositions may comprise yet another leishmanicidal or immunomodulatory drug, either in free form or in association with a nanocarrier such as liposomes, which confers pharmacokinetic properties more favorable to the active ingredients. Fullerol has surprisingly demonstrated an antileishmania activity as well as an ability to reduce meglumine antimoniate hepatotoxicity when administered in combination with an animal model of visceral leishmaniasis. Thus, fullerol was the first drug to demonstrate the dual ability to exert antileishmania action without side effects and to reduce the toxicity of another leishmanicidal drug without interfering with its pharmacological activity. Finally, the invention comprises the use of pharmaceutical compositions containing fullerenes for the treatment of leishmaniasis.
[002] A leishmaniose está entre as doenças tropicais mais negligenciadas no mundo, segundo a resolução da Sexagésima Assembleia Mundial da Saúde, em 2007. Epidemiologicamente, mais de 12 milhões de pessoas encontram-se infectadas no mundo, sendo 1 ,3 milhões de novos casos registrados a cada ano, com cerca de 20 a 30 mil mortes anuais, cuja doença afeta as populações mais pobres de 88 países (países em desenvolvimento) (World Health Organization - WHO. Report of the Fifth Consultative Meeting on Leishmania/HIV Coinfection . Addis Ababa, Ethiopia, 20 a 22 march 2007; WHO. Leishmaniasis. Fev. 201 3. Disponível em: http://www.who. int/mediacentre/factsheets/fs375/en/#. Acessado em: 16 de dezembro de 2013).  [002] Leishmaniasis is among the most neglected tropical diseases in the world, according to the resolution of the 60th World Health Assembly in 2007. Epidemiologically, over 12 million people are infected worldwide, with 1, 3 million new people. cases reported each year, with about 20 to 30,000 deaths annually, whose illness affects the poorest populations in 88 countries (developing countries) (World Health Organization - WHO. Report of the Fifth Consultative Meeting on Leishmania / HIV Coinfection). Addis Ababa, Ethiopia, March 20 - 22, 2007; WHO Leishmaniasis, Feb. 201 3. Available at: http://www.who.int/mediacenter/factsheets/fs375/en/# Accessed: December 16, 2013).
[003] As leishmanioses são doenças parasitárias causadas por mais de 20 espécies de protozoários do género Leishmania spp., da família dos Trypanosomatidae, parasites hemoflagelados que mostram preferência pelos fagolisossomos do sistema fagocítico mononuclear. São transmitidas para os humanos pela picada do vetor, as fêmeas dos géneros Phlebotomo sp. e Lutzomyia sp. infectadas. A doença se apresenta com três manifestações clínicas diferentes: visceral, ou comumente conhecida como calazar; a cutânea; e a muco-cutânea. (RICCIARDI, L.V. Terapêutica antileishmania: revisando el pasado, el presente y el futuro. Gac. Méd. Caracas, v.1 17, n. 2, p.93-1 1 1 , 2009). [003] Leishmaniasis are parasitic diseases caused by more than 20 species of protozoa of the genus Leishmania spp., Of the Trypanosomatidae family, hemoflagellate parasites showing preference for phagolysosomes of the mononuclear phagocytic system. Are transmitted to humans by the bite of the vector, females of the genera Phlebotomo sp. and Lutzomyia sp. infected. The disease presents with three different clinical manifestations: visceral, or commonly known as calazar; the cutaneous; and the mucocutaneous. (RICCIARDI, LV Antileishmania Therapy: Reviewing the Past, the Present, and the Future. Gac. Med. Caracas, v.1 17, no. 2, p.93-111, 2009).
[004] A leishmaniose cutânea é a forma mais comum de leishmaniose e apresenta- se por úlceras distribuídas nas partes expostas do corpo, deixando cicatrizes ao longo da vida e incapacidade grave. Cerca de 1/3 dos casos ocorrem nas Américas, na bacia do Mediterrâneo, Ásia Central e Oriente Médio. A leishmaniose mucocutânea leva às lesões mucosas mutilantes, que consistem na destruição parcial ou total das membranas mucosas do nariz, boca e garganta, sendo a maior parte dos casos relatados na Bolívia, Brasil e Peru (World Health Organization - WHO. Leishmaniasis. Fev. 2013. Disponível em: http://www.who. int/mediacentre/factsheets/fs375/en/#. Acessado em: 16 de dezembro de 2013).  Cutaneous leishmaniasis is the most common form of leishmaniasis and presents with ulcers distributed on the exposed parts of the body, leaving lifelong scarring and severe disability. About 1/3 of the cases occur in the Americas, the Mediterranean basin, Central Asia and the Middle East. Mucocutaneous leishmaniasis leads to mutilating mucosal lesions, which consist of partial or total destruction of the mucous membranes of the nose, mouth and throat, most of them reported in Bolivia, Brazil and Peru (World Health Organization - WHO. Leishmaniasis. Feb. 2013. Available at: http: //www.who.int/mediacentre/factsheets/fs375/en/ # Accessed: December 16, 2013).
[005] Contudo, a forma mais grave de leishmaniose compreende a leishmaniose visceral. 90% dos novos casos de calazar ocorrem em seis países: Bangladesh, Brasil, Etiópia, índia, Sudão do Sul e Sudão. A forma zoonótica, para a qual os cães são o principal reservatório e fonte de infecção da doença nas áreas endémicas, está presente na Bacia do Mediterrâneo, China, Oriente Médio e América do Sul, e é causada pela Leishmania infantum infantum e Leishmania infantum chagasi, sendo esta última a espécie causadora da leishmaniose visceral no Brasil (GRIENSVEN, J.; BALASEGARAM, M.; MEHEUS, F. et al. Combination therapy for visceral leishmaniasis. Lancet Infect Dis., v.10, p.184-94, 2010).  However, the most severe form of leishmaniasis comprises visceral leishmaniasis. 90% of new cases of kala-azar occur in six countries: Bangladesh, Brazil, Ethiopia, India, South Sudan and Sudan. The zoonotic form, for which dogs are the main reservoir and source of disease infection in endemic areas, is It is present in the Mediterranean Basin, China, Middle East and South America, and is caused by Leishmania infantum infantum and Leishmania infantum chagasi, the latter being the causative species of visceral leishmaniasis in Brazil (GRIENSVEN, J .; BALASEGARAM, M .; MEHEUS , F. et al., Combination therapy for visceral leishmaniasis (Lancet Infect Dis., V.10, p.184-94, 2010).
[006] O ciclo de vida do parasito é metaxênico, onde o agente passa por transformações no organismo do vetor flebotomíneo, inseto díptero. O protozoário flagelado L. infantum é encontrado na forma móvel promastigota metacíclica no vetor, e quando fagocitado pelos macrófagos do hospedeiro vertebrado se transforma na forma imóvel amastigota. O ciclo tem início com a inoculação de formas promastigotas infectantes no hospedeiro vertebrado durante o repasto sanguíneo. Ao atingirem a circulação sanguínea, as formas promastigotas utilizam de mecanismos específicos para sobreviver à lise celular, que será ativada pelo sistema complemento. Devido a este mecanismo protetor, o parasito sobrevive ao ataque do hospedeiro e ainda consegue invadir macrófagos através da manipulação de receptores celulares (CAMPOS-PONCE M, PONCE C, PONCE E, MAINGON RDC. Leishmania chagasi/infantum: further investigations on Leishmania tropism in atypical cutaneous and visceral leishmaniasis foci in Central America. Experimental Parasitology, New York, v. 109, p. 209-219, 2005). No interior destes, o parasito encontra-se protegido contra a resposta imune do hospedeiro, residindo no ambiente ácido do lisossomo secundário do macrófago onde sofre sucessivas multiplicações. Uma vez infectado, o hospedeiro torna-se reservatório do parasito. [006] The parasite's life cycle is metaxenic, where the agent undergoes transformations in the organism of the phlebotomine vector, the dipterous insect. The flagellated protozoan L. infantum is found in the mobile metacyclic promastigote form in the vector, and when phagocytized by the vertebrate host macrophages it becomes the immovable amastigote form. The cycle begins with the inoculation of infectious promastigote forms in the vertebrate host during blood meal. Upon reaching the bloodstream, promastigote forms use specific mechanisms to survive cell lysis, which will be activated by the complement system. Due to this protective mechanism, the parasite survives host attack and can still invade macrophages by manipulating cell receptors (CAMPOS-PONCE M, PONCE C, PONCE E, MAINGON RDC. Leishmania chagasi / infantum: further investigations on Leishmania tropism in atypical cutaneous and visceral leishmaniasis foci in Central America. Parasitology, New York, v. 109, p. 209-219, 2005). Within them, the parasite is protected against the host immune response, residing in the acidic environment of the secondary macrophage lysosome where it undergoes successive multiplications. Once infected, the host becomes the parasite's reservoir.
[007] Em humanos, a leishmaniose visceral apresenta-se como uma enfermidade generalizada, crónica, caracterizada por febre irregular e de longa duração, hepatoesplenomegalia, linfadenopatia, anemia com leucopenia, hipergamaglobulinemia e hipoalbuminemia, emagrecimento, edema e estado de debilidade progressivo, levando à caquexia e, até mesmo, ao óbito. A evolução das formas clínicas é diversa, podendo o indivíduo apresentar desde cura espontânea, formas oligossintomáticas e assintomáticas, até manifestações graves, podendo alcançar letalidade entre 10% e 98% em casos tratados inadequadamente e não tratados. Nos últimos anos, a letalidade da leishmaniose visceral vem aumentando gradativamente, passando de 3,6% em 1 994 para 6,7% em 2003, com um incremento de 85%, e para 8,4% em 2004 (ALVARENGA, D.G.; ESCALDA, P.M.F.; COSTA, A.S.V. et al. Leishmaniose visceral: estudo retrospectivo de fatores associados à letalidade. Rev. Soe. Bras. Med. Trop., v. 43, n. 2, p. 194-97, mar-abr 2010). In humans, visceral leishmaniasis presents as a chronic, widespread disease characterized by irregular and long-lasting fever, hepatosplenomegaly, lymphadenopathy, anemia with leukopenia, hypergammaglobulinemia, and progressive debilitation leading to cachexia and even death. The evolution of clinical forms is diverse, ranging from spontaneous cure, oligosymptomatic and asymptomatic forms, to severe manifestations, and may reach lethality between 10% and 98% in inadequately treated and untreated cases. In recent years, the mortality of visceral leishmaniasis has been gradually increasing, from 3.6% in 1 994 to 6.7% in 2003, with an increase of 85%, and to 8.4% in 2004 (ALVARENGA, DG; ESCALDA, PMF; COSTA, ASV Visceral leishmaniasis: a retrospective study of factors associated with lethality Rev. Soe, Bras. Med. Trop., V. 43, no 2, pp. 194-97, Mar-Apr 2010 ).
[008] Compostos antimoniais foram utilizados no tratamento da leishmaniose pela primeira vez em 1912 pelo médico brasileiro Gaspar Vianna, inicialmente na forma trivalente. Embora os antimoniais pentavalentes, como o antimoniato de meglumina (Glucantime®, Sanofi-Aventis), tenham sido reconhecidos como clinicamente eficazes em 1947, atualmente ainda são os fármacos de primeira escolha para todas as leishmanioses na maioria dos países em desenvolvimento (BERMAN JD. Human leishmaniasis: clinicai, diagnostic, and chemotherapeutic developments in the last 10 years.Clin. Infect. Dis. 24, 684, 1997). A anfotericina B suas várias formulações são utilizados como medicamentos de segunda escolha (Ministério da Saúde. Manual de vigilância e controle da leishmaniose visceral. Brasília, 2003)  Antimonial compounds were first used in the treatment of leishmaniasis in 1912 by Brazilian physician Gaspar Vianna, initially in the trivalent form. Although pentavalent antimonials such as meglumine antimoniate (Glucantime®, Sanofi-Aventis) were recognized as clinically effective in 1947, they are still the drug of choice for all leishmaniasis in most developing countries (BERMAN JD. Human leishmaniasis: clinical, diagnostic, and chemotherapeutic developments in the last 10 years (Lin. Infect. Dis. 24, 684, 1997). Amphotericin B and its various formulations are used as second-choice drugs (Ministry of Health. Visceral Leishmaniasis Surveillance and Control Manual. Brasília, 2003)
[009] No Brasil, a terapêutica das leishmanioses consiste no uso do antimoniato de meglumina (RICCIARDI, L.V. Terapêutica antileishmania: revisando el pasado, el presente y el futuro. Gac. Méd. Caracas, v.1 17, n. 2, p.93-1 1 1 , 2009). Todavia, alguns fatores relacionados à farmacocinética deste fármaco desfavorecem o seu uso. A baixa absorção oral e elevada depuração plasmática determinam que seja administrado diariamente por via parenteral (endovenosa ou intramuscular), por um período de 20 a 40 dias. Além disso, os efeitos colaterais representam um problema importante no uso dos antimoniais pentavalentes. Estes aparecem principalmente ao final do tratamento e incluem náuseas, vómitos, diarréia, atralgias, mialgias, anorexia, elevação dos níveis de enzimas hepáticas, anomalias eletrocardiográficas, convulsões, pancreatite química e nefrotoxicidade. Reclamações de pacientes com sensação de dor no local e no momento da aplicação desses medicamentos são também comuns (FRÉZARD et al. Pentavalent Antimonials: New Perspectives for Old Drugs. Molecules 14, 231 7, 2009). In Brazil, the therapy of leishmaniasis consists of the use of meglumine antimoniate (RICCIARDI, LV Antileishmania therapy: reviewing the past, present and future. Gac. Méd. Caracas, v.1 17, n. 2, p .93-111, 2009). However, some factors related to the pharmacokinetics of this drug disadvantage its use. Low oral absorption and high plasma clearance mean that it is administered daily parenterally (intravenously or intramuscularly), through a 20 to 40 days. In addition, side effects represent a major problem in the use of pentavalent antimonials. These appear mainly at the end of treatment and include nausea, vomiting, diarrhea, atralgias, myalgias, anorexia, elevated liver enzyme levels, electrocardiographic abnormalities, seizures, chemical pancreatitis and nephrotoxicity. Complaints from patients with pain sensation at the site and at the time of application of these medications are also common (FRÉZARD et al. Pentavalent Antimonials: New Perspectives for Old Drugs. Molecules 14, 231 7, 2009).
[010] O modelo geralmente aceito para o mecanismo de ação dos antimoniais pentavalentes (SbV) é que estes seriam um pró-fármaco, sendo reduzido à forma ativa e tóxica de antimônio trivalente (Sblll), que destrói as formas amastigotas dentro do fagolisossomos dos macrófagos. Neste contexto, Sblll promoveria desbalanço no metabolismo dos tióis, resultando em estresse oxidativo e apoptose (FRÉZARD et al. Pentavalent Antimonials: New Perspectives for Old Drugs. Molecules 14, 2317, 2009).  [010] The generally accepted model for the mechanism of action of pentavalent antimonials (SbV) is that they would be a prodrug, being reduced to the active and toxic form of trivalent antimony (Sblll), which destroys amastigote forms within the phagolysosomes. macrophages. In this context, Sblll would promote imbalance in thiol metabolism resulting in oxidative stress and apoptosis (FRÉZARD et al. Pentavalent Antimonials: New Perspectives for Old Drugs. Molecules 14, 2317, 2009).
[01 1 ] Contudo, o Glucantime® é contra indicado em pacientes que fazem uso de beta-bloqueadores e drogas antiarrítmicas, com insuficiência renal ou hepática, em mulheres grávidas nos dois primeiros trimestres de gestação. Não há contraindicações absolutas ao uso da anfotericina B; entretanto, complicações renais limitam o seu uso (Ministério da Saúde. Manual de vigilância e controle da leishmaniose visceral. Brasília, 2003).  [01 1] However, Glucantime® is contraindicated in patients taking beta-blockers and antiarrhythmic drugs with renal or hepatic impairment in pregnant women in the first two trimesters of pregnancy. There are no absolute contraindications to the use of amphotericin B; however, renal complications limit its use (Ministry of Health. Visceral Leishmaniasis Surveillance and Control Manual. Brasília, 2003).
[012] Uma estratégia atualmente recomenda pela Organização Mundial da Saúde (OMS) refere-se ao tratamento com associação de fármacos leishmanicidas diferentes, tendo como principais benefícios, um tratamento mais eficaz e seguro e com menor risco de desenvolvimento de resistência (Ministério da Saúde. Manual de vigilância e controle da leishmaniose visceral. Brasília, 2003) . Dentre os medicamentos que podem vir a substituir os antimoniais pentavalentes destacam-se: anfotericina B lipossomal (AmBisome®), miltefosina (Impávido®) e paromomicina ou aminosidina. Entretanto, o uso desses medicamentos até hoje encontra-se limitado, seja pelo elevado custo e por problema de estabilidade (Ambisome®), pela baixa eficácia (paromomicina), por efeitos tóxicos indesejáveis (miltefosina, paromomicina) ou pelo aparecimento de resistência (miltefosina) (FRÉZARD et al. Pentavalent Antimonials: New Perspectives for Old Drugs. Molecules 14, 2317, 2009). [013] Outro problema na terapêutica dessa doença no Brasil é sua grande ocorrência em zonas rurais, o que dificulta a assistência aos pacientes que têm que se deslocar até um centro de saúde para receber o tratamento sob controle médico. Nesse contexto, os casos de interrupção de tratamento se tomam frequentes, o que pode resultar no aparecimento de resistência ao medicamento. [012] One strategy currently recommended by the World Health Organization (WHO) refers to combination treatment of different leishmanicidal drugs, with the main benefits being more effective and safer treatment and with lower risk of developing resistance (Ministry of Health Visceral Leishmaniasis Surveillance and Control Manual (Brasília, 2003). The drugs that may replace pentavalent antimonials include liposomal amphotericin B (AmBisome®), miltefosine (Impávido®) and paromomycin or aminosidine. However, the use of these drugs to date is limited, either due to the high cost and stability problem (Ambisome®), low efficacy (paromomycin), undesirable toxic effects (miltefosine, paromomycin) or the appearance of resistance (miltefosine). ) (FRÉZARD et al. Pentavalent Antimonials: New Perspectives for Old Drugs. Molecules 14, 2317, 2009). [013] Another problem in the treatment of this disease in Brazil is its high occurrence in rural areas, which makes it difficult to assist patients who have to travel to a health center to receive treatment under medical control. In this context, cases of treatment discontinuation are frequent, which may result in the emergence of drug resistance.
[014] No cão, o principal reservatório doméstico de L. infantum chagasi, é recomendada a utilização de uma combinação de antimoniato de meglumina com alopurinol ou a combinação de miltefosina e alopurinol. O prolongamento do tratamento pode ser necessário e o prognóstico pode variar de favorável a desfavorável, isso dependerá do estado clínico e da resposta imunológica do animal (DANTAS-TORRES, F. ; SOLANO-GALLEGO, L ; BANETH, G.; RIBEIRO, V. M. et al. Canine leishmaniosis in the Old and New Worlds: unveiled similarities and differences. Trends in Parasitology, Oxford, v. 28, n. 12, p. 531 -538, 2012). Todavia, independente do fármaco de escolha, a cura parasitológica está muito longe de ser obtida, uma vez que mesmo que o tratamento permita redução da transmissão do agente através dos flebotomíneos, isto ocorre por um curto período. O que vai determinar a eficiência do tratamento está relacionado com o estado imune do cão, a farmacocinética e a sensibilidade de cada isolado de Leishmania, ou a resistência destes aos medicamentos (AIT-OUDHIA, K.; et al. In vitro susceptibility to antimonials and amphotericin B of Leishmania infantum strains isolated from dogs in a region lacking drug selection pressure. Veterinary Parasitology, Amsterdam, v. 187, p. 386-393, 2012). [014] In dogs, the main domestic reservoir of L. infantum chagasi, the use of a combination of meglumine antimoniate with allopurinol or the combination of miltefosine and allopurinol is recommended. Prolongation of treatment may be necessary and prognosis may vary from favorable to unfavorable, this will depend on the clinical condition and the immune response of the animal (DANTAS-TORRES, F.; SOLANO-GALLEGO, L; BANETH, G .; RIBEIRO, VM et al., Canine Leishmaniasis in the Old and New Worlds: Unveiled Similarities and Differences Trends in Parasitology, Oxford, v. 28, No. 12, pp. 531-538, 2012). However, regardless of the drug of choice, parasitological cure is very far from being achieved, since even if the treatment permits a reduction in the transmission of the agent through sand flies, this occurs for a short period. What will determine the effectiveness of treatment is related to the dog's immune status, pharmacokinetics and sensitivity of each Leishmania isolate, or drug resistance (AIT-OUDHIA, K .; et al. In vitro susceptibility to antimonials and amphotericin B of Leishmania infantum strains isolated from dogs in a region lacking drug selection pressure Veterinary Parasitology, Amsterdam, v 187, pp 386-393, 2012).
[015] As evidências sobre a ineficácia do tratamento farmacológico de cães infectados são cumulativas; elas demonstram que o animal apesar de apresentar melhoras clínicas não apresenta reversão do estado infectado, o que mantém o cão como fator de risco para a população e ainda aumenta a possibilidade de gerar cepas resistentes aos medicamentos de uso humano. Por não existir estratégias suficientes para evitar a transmissão da doença aos homens e animais, a conduta atual de maior segurança indicada é o sacrifício dos cães infectados (ORGANIZAÇÃO PANAM ERICANA DE SAÚDE - OPAS. Encuentro sobre vigilância, prevención y control de leishmaniasis visceral (LV) en el Cono Sur de Sudamérica. Foz do Iguazú, Brasil. 2009. Disponível em: http://new.paho. org/hq/index.php?option=com_docman&task=doc_view&gid=1 6961 & ltemid=, Acesso em: 17 de dezembro de 2013). [016] Nesse contexto, a inexistência de tratamento efetivo da leishmaniose visceral canina representa uma importante limitação da terapêutica atual. O pequeno número de medicamentos disponíveis associado às limitações encontradas no seu uso aponta para a necessidade urgente de desenvolver novos fármacos ou formulações. [015] Evidence on the ineffective pharmacological treatment of infected dogs is cumulative; They demonstrate that the animal, despite presenting clinical improvements, does not present a reversal of the infected state, which keeps the dog as a risk factor for the population and also increases the possibility of generating strains resistant to human medicines. Because there are not enough strategies to prevent transmission of the disease to humans and animals, the current most safe course of action is to sacrifice infected dogs (PANAM ERICAN HEALTH ORGANIZATION - PAHO. Surveillance, Prevention, and Control Survey of Visceral Leishmaniasis (LV)). ) in the South Cono of South America Foz do Iguazú, Brazil 2009. Available at: http://new.paho.org/hq/index.php? option = com_docman & task = doc_view & gid = 1 6961 & ltemid =, Accessed at: December 17, 2013). [016] In this context, the lack of effective treatment for canine visceral leishmaniasis represents an important limitation of current therapy. The small number of drugs available associated with the limitations found in their use points to the urgent need to develop new drugs or formulations.
[017] Uma estratégia bem sucedida para aumentar o índice terapêutico de fármacos contra a leishmaniose visceral refere-se à sua associação reversível a um nanossistema carreador, permitindo prolongar sua meia-vida no organismo e direcioná-los para os sítios de infeção (FRÉZARD, F.; DEMICHELI, C. New delivery strategies for the old pentavalent antimonial drugs. Expert Opinion on Drug Delivery, v. 7, p. 1343-1358, 2010). [017] A successful strategy to increase the therapeutic index of drugs against visceral leishmaniasis refers to their reversible association with a carrier nanosystem, allowing them to extend their half-life in the body and direct them to the sites of infection (FRÉZARD, DEMICHELI, C. New delivery strategies for the old antimonial pentavalent drugs Expert Opinion on Drug Delivery, v. 7, pp. 1343-1358, 2010).
[018] Entre os nanossistemas carreadores de medicamentos atualmente disponíveis, os lipossomas ocupam uma posição de destaque. Essas vesículas esféricas, constituídas por uma ou várias bicamadas concêntricas de lipídeos, podem armazenar em seu compartimento aquoso interno princípios ativos hidrossolúveis ou terem princípios ativos lipofílicos ou anfifílicos incorporados em suas membranas. Lipossomas convencionais são tipicamente formados de um fosfolipídeo, tal como a fosfatidilcolina, ou de um surfactante não iônico. Podem também incluir na sua composição o colesterol, um fosfolipídeo com carga negativa, como por exemplo, fosfatidilglicerol, ácido fosfatidico, dicetilfosfato, e/ou um fosfolipídeo com carga positiva, tal como esterarilamina.  [018] Among the currently available drug-bearing nanosystems, liposomes occupy a prominent position. These spherical vesicles, consisting of one or more concentric lipid bilayers, may store in their internal aqueous compartment water-soluble active principles or have lipophilic or amphiphilic active principles incorporated into their membranes. Conventional liposomes are typically formed of a phospholipid, such as phosphatidylcholine, or a nonionic surfactant. They may also include in its composition cholesterol, a negatively charged phospholipid, such as phosphatidylglycerol, phosphatidic acid, diketylphosphate, and / or a positively charged phospholipid such as esterarylamine.
[019] O medicamento administrado por via parenteral na forma encapsulada em lipossomas é liberado lentamente, evitando, por consequência, sua rápida eliminação pelo organismo. O resultado é um aumento da biodisponibilidade do medicamento, com potencialização da ação e redução da toxicidade.  [019] The parenterally administered drug in liposome-encapsulated form is slowly released, thus preventing its rapid elimination by the body. The result is increased bioavailability of the drug, with potentiation of action and reduction of toxicity.
[020] Por serem rapidamente clareados da circulação sanguínea pelos macrófagos do sistema fagocítico mononuclear, principalmente o fígado, o baço e a medula óssea, os lipossomas convencionais carregam a droga para os principais sítios de infecção da leishmaniose visceral, o que disponibiliza uma maior quantidade do fármaco para interagir com o parasito. Nesse contexto, foi desenvolvida uma formulação de anfotericina B em lipossomas convencionais (AmBisome) que foi usada com sucesso no tratamento de pacientes não responsivos aos medicamentos antimoniais, o mesmo ocorrendo no tratamento de pacientes com a forma PKDL, sem relato de efeitos colaterais. A eficácia na faixa de 100% em pacientes imunocompetentes rendeu-lhe a aprovação pela U.S. Food and Drug Administration (FDA) como a primeira apresentação à base de lipossomas a ser reconhecida para tratamento do calazar (MEYERHOFF A. U.S. Food and Drug Administration approval of AmBisome (liposomal amphotericin B) for treatment of visceral leishmaniasis. Clinicai Infectious Disease, v. 28, p. 42-48, 1999). Because they are rapidly cleared from the bloodstream by macrophages of the mononuclear phagocytic system, especially the liver, spleen and bone marrow, conventional liposomes carry the drug to major sites of visceral leishmaniasis infection, which provides a greater amount of the drug to interact with the parasite. In this context, a formulation of amphotericin B in conventional liposomes (AmBisome) was developed that was successfully used to treat patients not responsive to antimonial drugs, as well as to treat patients with the PKDL form without reporting side effects. Efficacy in the 100% range in immunocompetent patients earned it US Food and Drug Administration approval (FDA) as the first liposome-based presentation to be recognized for treatment of kala azar (MEYERHOFF AUS Food and Drug Administration of AmBisome (liposomal amphotericin B) for treatment of visceral leishmaniasis. Clinical Infectious Disease, v. 28, p. 42 -48, 1999).
[021 ] Encontra-se também no estado da técnica, pedido de patente intitulado "COMPOSIÇÃO FARMACÊUTICA COMPREENDENDO LIPOSSOMAS CONVENCIONAIS E LIPOSSOMAS DE CIRCULAÇÃO PROLONGADA COMO SISTEMA ENTREGADOR DE FÁRMACOS LEISHMANICIDAS" (BR102012005265) que reivindica composições farmacêuticas para o tratamento da leishmaniose visceral caracterizada por compreender a associação de lipossomas convencionais e de lipossomas de circulação prolongada como sistema entregador de fármacos leishmanicidas. Essa composição permite que o fármaco atinja com eficiência todos os sítios de infecção pelo parasito, sendo que os lipossomas de ciculação prolongada promovem o direcionamento mais efetivo de fármacos leishmanicidas para a medula óssea e o baço, enquanto lipossomas convencionais atuam no direcionamento do fármaco para o fígado.  [021] Also in the prior art is a patent application entitled "PHARMACEUTICAL COMPOSITION UNDERSTANDING CONVENTIONAL LIPOSOMES AND PROLONGED CIRCULATION LIPOSOMES" (BR1020120052ish) for claiming the pharmaceutical composition for claiming the treatment of pharmaceutical compositions. the combination of conventional liposomes and prolonged circulation liposomes as leishmanicidal drug delivery system. This composition enables the drug to efficiently reach all sites of parasite infection, and long-cycle liposomes promote more effective targeting of leishmanicidal drugs to the bone marrow and spleen, while conventional liposomes act to target the drug to the parasite. liver.
[022] Os fulerenos foram descobertos em 1985 (ADAMS, P.D., GROSSE- KUNSTLEVE, R.W., HUNG, L.-W., et aí. C60: buckminsferfullerene. Nature, v. 318, p.162-163, 1985) e representam uma família extensa de nanoestruturas esféricas de carbono, tendo como exemplo clássico a molécula de C6o denominada fulereno. Devido ás suas propriedades químicas e físicas, estas moléculas tornaram-se alvos atraentes para o uso na medicina (REBECCA, M.; HSING-LIN, W.; JUN.G., et al. Impact of physicochemical properties of engineered fullerenes on key, Toxic. and App. Pharmac, v.234, p.58-67, 2009). Fullerenes were discovered in 1985 (ADAMS, PD, GROSSEKUNSTLEVE, RW, HUNG, L.-W., et al. C60: buckminsferfullerene. Nature, v. 318, p.162-163, 1985) and they represent a large family of spherical carbon nanostructures, having as a classic example the C 6 molecule called fullerene. Due to their chemical and physical properties, these molecules have become attractive targets for use in medicine (REBECCA, M.; HSING-LIN, W .; JUN.G., et al. Impact of physicochemical properties of engineered fullerenes on key. , Toxic. And App. Pharmac, v.234, p.58-67, 2009).
[023] Essas nanoestruturas destacam-se pela alta estabilidade química e sua capacidade de complexar radicais livres e promover a expressão gênica de enzimas antioxidantes, inibindo os danos oxidativos (JENSEN, A.W.; WILSON, S.R., SCHUSTER, D.l. Biological Applications of Fullerenes . Bioorg. Med. Chem. v. 4, 767-79, 1996). Os fulerenos possuem alta reatividade com espécies reativas de oxigénio (ROS), tais como superóxido, radicais hidroxilados e óxido nítrico, os quais atacam macromoléculas biológicas como proteínas, lipídeos e DNA (BOSI, S. ; ROS T.D. ; SPALLUTO, G. et al. Fullerene derivatives: an attractive tool for biological applications, v. 38, p. 913-23, 2003). Entretanto, a aplicação tecnológica do fulereno na área biomédica tem sido limitada pelo caráter hidrofóbico da molécula. Neste contexto, foi desenvolvida uma classe de fulerenos polihidroxilados, também chamados de fulerenol ou fulerol, formados por modificação química. A solubilidade de cada molécula de fulerenol é dependente do número de grupos hidroxila introduzidos (KOKUBO, K. Water-Soluble Single-Nano Carbon Particles: Fullerenol and Its Derivatives. The Delivery of Nanoparticles. <Disponível em: http://www.intechopen.com/download/get/type/pdfs/id/36889>. Acessado em: 18 de dezembro de 2013). Dentre estas moléculas, foi feita a adição de 22-24 hidroxilas na sua superfície, gerando um composto hidrofílico denominado fulerol, o que aumenta sua solubilidade em solução aquosa e reduz a sua citotoxicidade (LI, J., TAKEUCHI, A., OZAWA, M., LI, X., et al. Fullerol formation catalyzed by quaternary ammonium hydroxides. J. Chem. Soe, Chem. Commun, p.1784-1785, 1993). Desde então, estas moléculas vem sendo estudadas pelo seu potencial terapêutico em diferentes patologias, incluindo doenças infectocontagiantes, que envolvem o estresse oxidativo, oncóticas. [023] These nanostructures stand out for their high chemical stability and their ability to complex free radicals and promote gene expression of antioxidant enzymes, inhibiting oxidative damage (JENSEN, AW; WILSON, SR, SCHUSTER, DL Biological Applications of Fullerenes. Bioorg (Chem. V. 4, 767-79, 1996). Fullerenes have high reactivity with reactive oxygen species (ROS) such as superoxide, hydroxylated radicals and nitric oxide which attack biological macromolecules such as proteins, lipids and DNA (BOSI, S.; ROS TD; SPALLUTO, G. et al. Fullerene derivatives: an attractive tool for biological applications, v. 38, pp. 913-23, 2003). However, the technological application of fullerene in the biomedical area has been limited by the hydrophobic character of the molecule. In this In this context, a class of polyhydroxylated fullerenes, also called fullerenol or fullerol, formed by chemical modification was developed. The solubility of each fullerenol molecule is dependent on the number of hydroxyl groups introduced (KOKUBO, K. Water-Soluble Single-Nano Carbon Particles: Fullerenol and Its Derivatives. The Delivery of Nanoparticles. <Available at: http://www.intechopen .com / download / get / type / pdfs / id / 36889> Accessed December 18, 2013). Among these molecules, 22-24 hydroxyls were added to their surface, generating a hydrophilic compound called fullerol, which increases its solubility in aqueous solution and reduces its cytotoxicity (LI, J., TAKEUCHI, A., OZAWA, M., L., X., et al., Fullerol formation catalyzed by quaternary ammonium hydroxides (J. Chem. Soc., Chem. Commun, p. 1784-1785, 1993). Since then, these molecules have been studied for their therapeutic potential in different pathologies, including infectious diseases, involving oncotic oxidative stress.
[024] Um derivado polihidroxilado de fulereno solúvel em água, o fulerenel, demonstrou atividade antiviral em células mononucleares periféricas de humanos (PBMC) com infecção aguda e células H9 cronicamente infectadas com vírus HIV-1 , apresentando EC50 de 7,3±5,9 μΜ e 10,8±8,2 μΜ, respectivamente. Já em células PBMC infectadas de forma aguda com HIV-2ROD, cepa resistente aos fármacos anti-retrovirais, o fulerenel apresentou uma EC50= 0,003±0,004 μΜ, mostrando-se portanto altamente eficaz como fármaco antiviral no tratamento desta síndrome (Schinazi, R. F.; Sijbesma, R.; Srdanov, G.; Hill, C. L.; Wudl, F. Synthesis and Virucidal Activity of a Water-Soluble, Configurationally Stable, Derivatized C60 Fullerenel. Antimicrob. Agents Chemother, v. 37, p. 1707-10. 1993).  [024] A water-soluble polyhydroxylated fullerene derivative, fullerenel, has shown antiviral activity in acute-infected human peripheral mononuclear cells (PBMC) and chronically HIV-1 virus-infected H9 cells with an EC50 of 7.3 ± 5, 9 μΜ and 10.8 ± 8.2 μΜ, respectively. In PBMC cells infected with HIV-2ROD, a strain resistant to antiretroviral drugs, the fullerenel had an EC50 = 0.003 ± 0.004 μΜ, thus proving to be highly effective as an antiviral drug in the treatment of this syndrome (Schinazi, RF; Sijbesma, R.; Srdanov, G.; Hill, CL; Wudl, F. Synthesis and Virucidal Activity of a Water-Soluble, Configurationally Stable, Derivatized C60 Fullerenel, Antimicrob., Agents Chemother, v. 37, pp. 1707-10. 1993).
[025] Foi comprovado que o fulerol pode agir como fármaco neuroprotetor em doenças degenerativas relacionadas ao estresse oxidativo (DORDEVIC, A. e BOGDANOVIC, G. Fullerenol - a new nanopharmaceutic? Arch. Oncol., v.16, p.42-5, 2008). O mecanismo de neuroproteção mediada pelo fulerenol ocorre pela inibição dos receptores para glutamato, e também pela redução da liberação de glutamato via aumento nas concentrações intracelulares de cálcio, um mecanismo crítico de excitotoxicidade em neurónios (SILVA, G.A. Nanotechnology approaches for the regeneration and neuroprotection of the central nervous system. Surgical Neurology , v. 63, p. 301 - 306, 2005). [026] Já se sabe que o fulerol não apresenta toxicidade aguda ou sub-aguda em roedores e protege os tecidos de danos celulares causados por ROS após exposição a substâncias tóxicas pró-oxidantes incluindo alguns fármacos, por absorver estes radicais livres que causariam injúrias teciduais e celulares (BOSI, S.; DA ROS, T.; SPALLUTO, G., et al. Fullerene derivatives: an attractive tool for biological applications. European Journal of Medicinal Chemistry, v. 38, p. 913-23, 2003). Fullerol has been shown to act as a neuroprotective drug in oxidative stress-related degenerative diseases (DORDEVIC, A. and BOGDANOVIC, G. Fullerenol - a new nanopharmaceutic? Arch. Oncol., V.16, p.42-5 , 2008). The mechanism of fullerenol-mediated neuroprotection occurs by inhibiting glutamate receptors, and also by reducing glutamate release via increased intracellular calcium concentrations, a critical mechanism of excitotoxicity in neurons (SILVA, GA). the central nervous system, Surgical Neurology, v. 63, pp. 301-306, 2005). [026] Fullerol is not known to have acute or subacute toxicity in rodents and protects tissues from cellular damage caused by ROS following exposure to pro-oxidant toxic substances including some drugs by absorbing these free radicals that would cause tissue injury. and cellular (BOSI, S .; DA ROS, T .; SPALLUTO, G., et al. Fullerene derivatives: an attractive tool for biological applications. European Journal of Medicinal Chemistry, v. 38, p. 913-23, 2003) .
[027] Uma patente concedida intitulada "FULLERENE PHARMACEUTICAL COMPOSITIONS FOR PREVENTING OR TREATING DISORDERS" (US 6.777.445 B2), refere-se a um método para a prevenção e tratamento de infecções bacterianas causadas por bactérias gram-positivas e gram-negativas e doenças virais, tais como dengue e encefalite virai compreendendo a administração de composições farmacêuticas contendo um fulereno, sendo este administrado in vivo numa quantidade de cerca de 0,001 a cerca de 1 00 mg/kg de peso corporal do indivíduo. Entretanto, nesta invenção, foi testado um único fulereno solúvel em água derivado de ácido carboxílico de fórmula estrutural C6o(C3) simétrico, se diferenciando primordialmente da presente invenção por esta ser um fulereno com adição de 22-24 hidroxilas na superfície, além desta não reivindicar a administração parenteral de formulações com nanossistemas carreadores. Ademais, distingui-se por se tratar do uso do fulerol para tratamento das leishmanioses visceral e cutânea.  [027] A patent issued entitled "FULLERENE PHARMACEUTICAL COMPOSITIONS FOR PREVENTING OR TREATING DISORDERS" (US 6,777,445 B2) relates to a method for the prevention and treatment of bacterial infections caused by gram-positive and gram-negative bacteria. viral diseases such as dengue and viral encephalitis comprising administering pharmaceutical compositions containing a fullerene, which is administered in vivo in an amount from about 0.001 to about 100 mg / kg body weight of the subject. However, in this invention, a single water-soluble fullerene derived from the symmetrical C60 (C3) structural formula carboxylic acid, differing primarily from the present invention in that it is a fullerene with addition of 22-24 hydroxyls on the surface, is also not present. claim parenteral administration of formulations with carrier nanosystems. Moreover, it is distinguished by the use of fullerol for the treatment of visceral and cutaneous leishmaniasis.
[028] Outra patente concedida, a US 7.947.262 B2, intitulada "USE OF FULLERENES FOR THE TREATMENT OF MAST CELL AND BASOPHIL- MEDIATED DISEASE" que compreende o uso de derivados fulerenos solúveis em água, preferencialmente C40-C8o para o tratamento de distúrbios envolvendo células do sistema imune como mastócitos e basófilos periféricos sanguíneos relacionados com processos alérgicos e inflamatórios, como artrite. Essa invenção refere-se ainda a moléculas quiméricas de fulerenos solúveis em água funcionalizadas com compostos capazes de se ligar a receptores presentes em mastócitos ou basófilos, mediando a liberação de citocinas e imunoglobulinas, como por exemplo peptídeos derivados de fatores de crescimento de células-tronco. A patente apesar de reivindicar composições farmacêuticas de fulerenos C40-C80 encapsulados em lipossomas, não avalia pré-clinica e clinicamente, e tampouco especifica as formulações lipossomais. Além de apresentar uma aplicabilidade distinta da tecnologia aqui descrita. [029] A patente intitulada "FINELY PARTICULATE COMPOSITE CONTAINING CARBON COMPOUND ENCAPSULATED THEREIN" (US 2007/0077432 A1 ), que reivindica composições contendo nanocompostos a base de polímeros fechados de carbono C30-C2000, preferencialmente entre 60-120 átomos de carbono, como fulerenos ou nanotubos podendo conter um metal associado, sendo estes empregados na medicina para a terapêutica ou usados para diagnósticos, distingui- se da presente tecnologia por esta compreender composições antileishmania contendo fulerenos, preferencialmente o fulerol, estando estes em associação a fármaco leishmanicida ou imunomodulador, livre ou encapsulado em nanossistema carreador coloidal. [028] Another patent granted, US 7,947,262 B2, entitled "USE OF FULLERENES FOR THE TREATMENT OF MAST CELL AND BASOPHIL-MEDIATED DISEASE" which comprises the use of water soluble fullerene derivatives, preferably C 40 -C 8 o for the treatment of disorders involving immune system cells such as mast cells and peripheral blood basophils related to allergic and inflammatory processes such as arthritis. This invention further relates to water-soluble chimeric fullerene molecules functionalized with compounds capable of binding to receptors present on mast cells or basophils, mediating the release of cytokines and immunoglobulins, such as peptides derived from stem cell growth factors. . The patent, despite claiming liposome-encapsulated C40-C80 fullerene pharmaceutical compositions, does not preclinically and clinically evaluate, nor does it specify liposomal formulations. In addition to having a different applicability of the technology described herein. [029] The patent entitled "FINELY PARTICULATE COMPOSITE CONTAINING CARBON COMPOUND ENCAPSULATED THEREIN" (US 2007/0077432 A1), which claims compositions containing nanocomposites based on C30-C2000 closed carbon polymers, preferably between 60-120 carbon atoms, such as Fullerenes or nanotubes which may contain an associated metal, whether employed in medicine for therapy or used for diagnosis, are distinguished from the present technology in that it comprises fullerenes-containing antileishmania compositions, preferably fullerol, which are in combination with leishmanicidal or immunomodulatory drug. free or encapsulated in a colloidal carrier nanosystem.
[030] Assim, diante do que foi encontrado no estado da técnica, frente à problemática do alto índice mundial de casos de infecção por Leishmania spp., dos graves efeitos colaterais ocasionados pelo medicamento atual antimoniato de meglumina e do número escasso de outros medicamentos disponíveis para o tratamento das leishmanioses, conclui-se que as composições antileishmania contendo o fulerol e demais composições contendo um fulereno solúvel em água mostram-se ferramentas promissoras para serem produzidas e utilizadas na terapêutica contra as leishmanioses. A presente invenção relata de forma surpreendente a atividade antileishmania do fulerol, assim como sua dupla capacidade de exercer ação antileishmania sem efeitos colaterais e de reduzir os efeitos colaterais do antimoniato de meglumina sem interferir nas propriedades farmacológicas deste, quando administrado em associação via parenteral. O tratamento de camundongos infectados experimentalmente com Leishmania infantum chagasi com fulerol livre ou encapsulado reduziu a carga parasitária hepática de modo equivalente ao observado pelo antimoniato de meglumina. O fulerol apresentou uma maior atividade antileishmania na forma encapsulada em lipossomas que na forma livre. Todavia, quando comparada a posologia deste (5 doses de 0,05 mg/kg a cada 4 dias) com a do medicamento de primeira escolha (20 doses de Glucantime 120 mg Sb/kg/dia), o fulerol mostrou-se muito mais eficaz considerando a baixa dose administrada e o longo intervalo de tempo entre as doses. Ademais, quando avaliado o tratamento com fulerol encapsulado em lipossomas em associação ao Glucantime, não foram observadas as alterações histopatológicas no fígado dos animais ocasionadas com o uso do fármaco de primeira escolha e que caracterizam a hepatotoxicidade. O que demonstra a ação hepatoprotetora do co-tratamento com o fulerol encapsulado em lipossomas. Portanto, essa invenção reivindica composições farmacêuticas contendo os fulerenos como novos medicamentos antileishmania e o uso destes em associação com fárrmaco(s) leishmanicida(s) ou irmunomodulador(es) para potenciar a eficácia e reduzir a toxicidade dos mesmos. [030] Thus, in light of what has been found in the prior art, in view of the problem of the high worldwide rate of cases of Leishmania spp. Infection, the serious side effects caused by the current meglumine antimoniate drug and the scarce number of other available drugs. For the treatment of leishmaniasis, it is concluded that the antileishmania compositions containing fullerol and other compositions containing a water soluble fullerene are promising tools to be produced and used in therapy against leishmaniasis. The present invention surprisingly reports the fullerol antileishmania activity as well as its dual ability to exert antileishmania action without side effects and to reduce the side effects of meglumine antimoniate without interfering with the pharmacological properties of meglumine when administered in combination via parenteral route. Treatment of experimentally infected mice with Leishmania infantum chagasi with free or encapsulated fullerol reduced hepatic parasitic burden in a manner equivalent to that observed by meglumine antimoniate. Fullerol showed higher antileishmania activity in the liposome encapsulated form than in the free form. However, when compared to its dosage (5 doses of 0.05 mg / kg every 4 days) with that of the first-choice medicine (20 doses of Glucantime 120 mg Sb / kg / day), fullerol was much higher. effective considering the low dose administered and the long time interval between doses. Moreover, when the treatment with liposome-encapsulated fullerol in combination with Glucantime was evaluated, the histopathological changes in the liver of the animals caused by the use of the first choice drug that characterize the hepatotoxicity were not observed. What the action demonstrates co-treatment with liposome-encapsulated fullerol. Therefore, this invention claims pharmaceutical compositions containing fullerenes as novel antileishmania drugs and their use in combination with leishmanicidal or immunomodulatory drug (s) to enhance their efficacy and reduce their toxicity.
BREVE DESCRIÇÃO DAS FIGURAS  BRIEF DESCRIPTION OF THE FIGURES
[031 ] Figura 1 : A figura 1 mostra de forma comparativa a carga parasitária no fígado de camundongos BALB/c infectados experimentalmente com Leishmania infantum (modelo murino de leishmaniose visceral). A quantidade de parasitos foi determinada por PCR em tempo real quantitativa, após 20 dias de tratamento administrados por via intraperitoneal com doses diárias de Glucantime (120 mg Sb/kg), fulerol livre (0,05 mg/kg), fulerol encapsulado em lipossomas (0,05 mg/kg) e salina (controle negativo).  [031] Figure 1: Figure 1 comparatively shows the parasitic burden on the liver of BALB / c mice experimentally infected with Leishmania infantum (murine model of visceral leishmaniasis). The amount of parasites was determined by quantitative real-time PCR after 20 days of intraperitoneal administration with daily doses of Glucantime (120 mg Sb / kg), free fullerol (0.05 mg / kg), liposome-encapsulated fullerol. (0.05 mg / kg) and saline (negative control).
[032] Figura 2: A figura 3 mostra a eficácia do tratamento associando o fulerol com Glucantime em modelo murino de leishmaniose visceral, sendo o fulerol administrado na forma livre ou encapsulada em lipossomas. Avaliou-se a carga parasitária no fígado de camundongos BALB/c infectados com Leishmania infantum chagasi, determinada por PCR em tempo real quantitativa, após 20 dias de tratamento por via intraperitoneal com fulerol encapsulado em lipossomas (0,05 mg/kg/dose com intervalo de 4 dias), Glucantime (120 mg Sb/kg/dia), Glucantime (120 mg Sb/kg/dia) + Fulerol (0,05 mg/kg/dose/4 dias), Glucantime (120 mg Sb/kg/dia) + Fulerol em lipossomas (0,05 mg/kg/dose/4 dias) ou salina.  [032] Figure 2: Figure 3 shows the efficacy of treatment combining fullerol with Glucantime in a murine model of visceral leishmaniasis, with fullerol being administered in free or liposome-encapsulated form. The liver parasite burden of Leishmania infantum chagasi infected BALB / c mice, determined by quantitative real-time PCR, was evaluated after 20 days of intraperitoneal treatment with liposome-encapsulated fullerol (0.05 mg / kg / dose with 4 days interval), Glucantime (120 mg Sb / kg / day), Glucantime (120 mg Sb / kg / day) + Fullerol (0.05 mg / kg / dose / 4 days), Glucantime (120 mg Sb / kg) / day) + Liposome fullerol (0.05 mg / kg / dose / 4 days) or saline.
[033] Figura 3: A figura 3 apresenta os resultados de análise histopatológica e de apoptose por microscopia óptica utilizando o método de coloração HE do fígado de camundongos BALB/c infectados experimentalmente com Leishmania infantum, após 20 dias de tratamento por via intraperitoneal com Glucantime (120 mg Sb/kg/dia), Glucantime (120 mg Sb/kg/dia) associado ao Fulerol (0,05 mg/kg/dose/4 dias), Glucantime (120 mg Sb/kg/dia) associado ao Fulerol em lipossomas (0,05 mg/kg/dose/4 dias) ou salina. Os parâmetros histopatológicos avaliados foram a degeneração hidrópica (A) e o índice apoptótico (B). [033] Figure 3: Figure 3 presents the results of histopathological analysis and apoptosis by light microscopy using the HE liver staining method of Leishmania infantum experimentally infected BALB / c mice after 20 days of intraperitoneal Glucantime treatment. (120 mg Sb / kg / day), Glucantime (120 mg Sb / kg / day) associated with Fullerol (0.05 mg / kg / dose / 4 days), Glucantime (120 mg Sb / kg / day) associated with Fullerol in liposomes (0.05 mg / kg / dose / 4 days) or saline. The histopathological parameters evaluated were hydropic degeneration (A) and apoptotic index (B).
[034] Figura 4: A figura 4 apresenta a viabilidade de macrófagos peritoniais expostos ao fulerol. A viabilidade celular foi representada através de valores de absorbância pós 72 horas de exposição a concentrações crescentes de fulerol. *p <0,05 = diferença significativa em relação ao grupo controle (0). Teste de Kruskal- Wallis com pós-teste de Dunn's. Figure 4: Figure 4 shows the viability of peritoneal macrophages exposed to fullerol. Cell viability was represented by absorbance values after 72 hours of exposure to increasing concentrations of fullerol. * p <0.05 = significant difference compared to the control group (0). Kruskal-Wallis test with Dunn's post-test.
[035] Figura 5: A figura 5 apresenta a atividade antileishmania in vitro do fulerol e glucantime. índice de infecção de macrófagos peritoneais com amastigotas de L. Infantum (cepa BH 46), 72 horas após exposição ao Glucantime (0,05 mg/ml, 0,75mg/ml ou 1 mg/ml), fulerol 0,12mg/ml. Os dados são mostrados como medias ± EP. *p < 0,05 = diferença significativa em relação ao grupo não tratado. Análise com One-Way ANOVA e pós teste Bonferroni.  [035] Figure 5: Figure 5 shows the in vitro antileishmania activity of fullerol and glucantime. peritoneal macrophage infection rate with L. Infantum amastigotes (BH 46 strain), 72 hours after exposure to Glucantime (0.05 mg / ml, 0.75 mg / ml or 1 mg / ml), fullerol 0.12 mg / ml . Data are shown as means ± SE. * p <0.05 = significant difference from untreated group. One-way ANOVA analysis and Bonferroni post test.
[036] Figura 6: A figura 6 apresenta a atividade antileishmania de diferentes doses do fulerol encapsulado em lipossomas no fígado de camundongos BALB/c infectados com L. infantum. Em A, carga parasitária no fígado de camundongos BALB/c infectados com L. infantum determinada por PCR quantitativa, após 20 dias de tratamento por via intraperitoneal com Glucantime (120 mg Sb/kg/dia), fulerol livre (0,05 mg/kg/4dias) e fulerol encapsulado em lipossomas (0,05 e 0,2 mg/kg/4dias) e lipossomas vazios (mesma dose de lipídeo que nos grupos recebendo o fulerol encapsulado). As barras representam as medianas da carga parasitaria (n = 6). *P< 0,05 e *** p<0,001 teste de Kruskal-Wallis com pós-teste de Dunn's. Em B, proporção de camundongos com fígado positivo para Leishmania por PCR. ** p<0,05 no teste exato de Fischer. [036] Figure 6: Figure 6 shows the antileishmania activity of different doses of liposome-encapsulated fullerol in the liver of L. infantum infected BALB / c mice. In A, parasitic load in the liver of L. infantum infected BALB / c mice determined by quantitative PCR after 20 days of intraperitoneal treatment with Glucantime (120 mg Sb / kg / day), free fullerol (0.05 mg / day). kg / 4 days) and liposome-encapsulated fullerol (0.05 and 0.2 mg / kg / 4 days) and empty liposomes (same lipid dose as in the groups receiving encapsulated fullerol). The bars represent the parasite load medians (n = 6). * P <0.05 and *** p <0.001 Kruskal-Wallis test with Dunn 's post-test's. In B, proportion of mice with liver positive for Leishmania by PCR. ** p <0.05 in Fischer's exact test.
[037] Figura 7: A figura 7 apresenta a atividade antileishmania de diferentes doses do fulerol encapsulado em lipossomas no baço de camundongos BALB/c infectados com L. infantum. Carga parasitária no fígado de camundongos BALB/c infectados com L. infantum determinada por PCR quantitativa, após 20 dias de tratamento por via intraperitoneal com fulerol encapsulado em lipossomas (0,05 e 0,2 mg/kg/4 dias), Glucantime (120 mg Sb/kg/dia), fulerol livre (0,05 mg/kg/4 dias), lipossomas vazios (mesma dose de lipídeo que nos grupos recebendo o fulerol encapsulado) e salina. As barras representam as medianas da carga parasitaria (n = 6). *P< 0,05 e *** p<0,001 , teste de Kruskal-Wallis com pós-teste de Dunn's. *P<0,05 Mann Whitney test para comparação entre os grupos Glucantime e Fui Lipo 0,2. [037] Figure 7: Figure 7 shows the antileishmania activity of different doses of liposome-encapsulated fullerol in the spleen of L. infantum infected BALB / c mice. Parasitic burden on liver of L. infantum infected BALB / c mice determined by quantitative PCR after 20 days of intraperitoneal treatment with liposome-encapsulated fullerol (0.05 and 0.2 mg / kg / 4 days), Glucantime ( 120 mg Sb / kg / day), free fullerol (0.05 mg / kg / 4 days), empty liposomes (same dose of lipid as in the groups receiving encapsulated fullerol) and saline. The bars represent the parasite load medians (n = 6). * P <0.05 and *** p <0.001, Kruskal-Wallis test with Dunn 's post-test's. * P <0.05 Mann Whitney test for comparison between Glucantime and Fui Lipo 0.2 groups.
[038] Figura 8: A figura 8 apresenta o perfil de citocinas no fígado de camundongos BALB/c infectados com L. infantum, após tratamento com diferentes doses de fulerol encapsulado em lipossomas. Perfil de citocinas determinadas no fígado de camundongos BALB/c infectados com L. infantum, após 20 dias de tratamento por via intraperitoneal com fulerol encapsulado em lipossomas (0,05 e 0,2 mg/kg/4dias), fulerol livre (0,05 mg/kg/4 dias), Glucantime (120 mg Sb/kg/dia), lipossomas vazios (mesma dose de lipídeo que nos grupos recebendo o fulerol encapsulado) e salina. As barras representam as medias da concentração de citocina (n = 6). *P< 0,05, teste de One Way ANOVA com pós-teste de Bonferoni. [038] Figure 8: Figure 8 shows the liver cytokine profile of L. infantum infected BALB / c mice following treatment with different doses of liposome-encapsulated fullerol. Profile of liver cytokines determined from L. infantum infected BALB / c mice after 20 days of intraperitoneal treatment with liposome-encapsulated fullerol (0.05 and 0.2 mg / kg / 4 days), free fullerol (0.05 mg / kg / 4 days), Glucantime (120 mg Sb / kg / day), empty liposomes (same lipid dose as in the groups receiving encapsulated fullerol) and saline. Bars represent cytokine concentration averages (n = 6). * P <0.05, One Way ANOVA test with Bonferoni posttest.
DESCRIÇÃO DETALHADA DA INVENÇÃO DETAILED DESCRIPTION OF THE INVENTION
[039] A presente invenção refere-se às composições farmacêuticas contendo fulerenos, como novas ferramentas para o tratamento das leishmanioses visceral e cutânea. Essas composições compreende preferencialmente os fulerenos solúveis em água de 60 a 120 carbonos com adição de grupamentos hidroxila, tal como o fulerol C6oOH22-24- As composições antileishmania contendo o fulerol podem compreender o fármaco em associação com outro(s) fármaco(s) leishmanicida(s) ou imunomodulador(es), estando na forma livre ou associada a um nanocarreador, como lipossomass, que confere aos princípios ativos propriedades farmacocinéticas mais favoráveis. Por fim, a invenção compreende o uso de composições farmacêuticas contendo fulerenos para o tratamento das leishmanioses. [039] The present invention relates to fullerene-containing pharmaceutical compositions as novel tools for the treatment of visceral and cutaneous leishmaniasis. Such compositions preferably comprise water-soluble fullerenes of 60 to 120 carbons with the addition of hydroxyl groups such as fullerol C 6 oOH 2 2-24. Antileishmania compositions containing fullerol may comprise the drug in combination with other drug (s). leishmanicide (s) or immunomodulator (s), either in free form or in association with a nanocarrier, such as liposomes, which gives the active principles more favorable pharmacokinetic properties. Finally, the invention comprises the use of pharmaceutical compositions containing fullerenes for the treatment of leishmaniasis.
1 . O fulerol foi sintetizado de acordo com o processo já descrito na patente depositada BR 0404543-2 A2, intitulada "PROCESSO E SISTEMA DE PRODUÇÃO CONTÍNUA EM LARGA ESCALA DE NANOESTRUTURAS DE CARBONO CONSTITUÍDO DE DISPOSITIVOS PARA EFETUAR O PROCESSO". O fulerol foi também encapsulado em lipossomas formados de fosfatidilcolina, colesterol e fosfatidilglicerol na relação molar 5:4:1 usando o método de desidratação-reidratação (FRÉZARD, F. e SCHETTINI, D. A. Lipossomas: propriedades físico-químicas e farmacológicas, aplicações na Quimioterapia à base de antimônio. Quim. Nova, V. 28, n. 3, p.51 1 -518, 2005). No entanto esses podem ser formados de colesterol e um lipídeo com carga negativa ou positiva, na presença ou ausência e de polímeros e/ou ligantes acoplados em suas superfícies.  1 . Fullerol was synthesized according to the process already described in the filed patent BR 0404543-2 A2, entitled "LARGE-SCALE CONTINUOUS PRODUCTION PROCESS AND SYSTEM OF CONSTITUTED DEVICES TO PERFORM THE PROCESS". Fullerol was also encapsulated in liposomes formed of phosphatidylcholine, cholesterol and phosphatidylglycerol in a 5: 4: 1 molar ratio using the dehydration-rehydration method (FRÉZARD, F. and SCHETTINI, DA. Liposomes: physicochemical and pharmacological properties, chemotherapy applications. based on antimony Quim Nova, V. 28, No. 3, p.51 1 -518, 2005). However, these may be formed of cholesterol and a negatively or positively charged lipid in the presence or absence and polymers and / or ligands coupled to their surfaces.
2. A encapsulação foi seguida da calibração do tamanho das vesículas por extrusão através de membrana de policarbonato de poro de diâmetro de 200 nm (NAYAR, R., HOPE, M.J., CULLIS, P.R. Generation of large unilamellar vesicles from long-chain saturated phosphatidylcholines by extrusion technique. Biochim. Biophys. Acta, v. 986, p. 200-206, 1989). na presença ou ausência e de polímeros e/ou ligantes acoplados em suas superfícies. [040] A atividade antileishmania e a ação quimioprotetora do fulerol foram avaliadas em camundongos BALB/c infectados com Leishmania iníantum, como modelo murino de leishmaniose visceral tratado ou não com Glucantime® (antimoniato de meglumina). No primeiro teste, foram utilizados quatro grupos (n = 9-10) que receberam doses diárias por via intraperitoneal durante 20 dias dos seguintes tratamentos: 1 ) Glucantime na dose de 120 mg Sb/kg de peso corporal; 2) Fulerol na dose de 0,05 mg/kg de peso corporal; 3) Fulerol encapsulado em lipossomas na dose de 0,05 mg/kg; 4) salina (controle negativo). Depois dos tratamentos, os animais foram sacrificados para a determinação da carga parasitária no fígado pela técnica de PCR em tempo real quantitativa. 2. Encapsulation was followed by calibration of vesicle size by extrusion through 200 nm pore polycarbonate membrane (NAYAR, R., HOPE, MJ, CULLIS, PR. Generation of large unilamellar vesicles from long-chain saturated phosphatidylcholines. by extrusion technique, Biochim, Biophys (Acta, v. 986, pp. 200-206, 1989). in the presence or absence of coupled polymers and / or binders on their surfaces. [040] Antileishmania activity and fullerol chemoprotective action were evaluated in Leishmania iníantum-infected BALB / c mice as a murine model of Glucantime®-treated or non-Glucantime-treated visceral leishmaniasis (meglumine antimoniate). In the first test, four groups (n = 9-10) were administered daily intraperitoneal doses for 20 days of the following treatments: 1) Glucantime at a dose of 120 mg Sb / kg body weight; 2) Fullerol at a dose of 0.05 mg / kg body weight; 3) Liposome-encapsulated fullerol at a dose of 0.05 mg / kg; 4) saline (negative control). After the treatments, the animals were sacrificed for the determination of the parasite load in the liver by the quantitative real time PCR technique.
[041 ] Também foi avaliada a eficácia do Fulerol livre e encapsulado em lipossomas após a administração de apenas cinco doses (0,05 mg/kg/dose), com intervalo de 4 dias (diferentemente das 20 doses administradas no primeiro teste) em comparação à eficácia do Glucantime (120 mg kg/dia) administrado diariamente durante 20 dias. O resultado obtido estabeleceu que o uso do fulerol encapsulado em lipossomas apresentou uma atividade antileishmania similar a demonstrada pelo Glucantime, porém com posologia mais aceitável.  [041] The efficacy of free and liposome-encapsulated fullerol was also evaluated after administration of only five doses (0.05 mg / kg / dose), 4 days apart (unlike the 20 doses given in the first test) compared efficacy of Glucantime (120 mg kg / day) administered daily for 20 days. The obtained result established that the use of liposome-encapsulated fullerol presented an antileishmania activity similar to that demonstrated by Glucantime, but with a more acceptable dosage.
[042] Dessa forma, de acordo com a presente invenção, o fulerol pode ser associado a nanossistemas carreadores e/ou sistemas de liberação controlada de fármaco, visando um aumento da sua estabilidade, uma maior biodisponibilidade, uma ação mais prolongada e/ou um direcionamento para o tecido alvo. Esses sistemas carreadores ou de liberação controlada classicamente usados no estado da técnica incluem, de forma não limitante, ciclodextrinas, polímeros, polímeros mucoadesivos, vesículas lipídicas, lipossomas, polimersomas, nanopartículas lipídicas sólidas, micro- e nanopartículas poliméricas, micro- e nanocapsulas, micro- e nanoemulsões, dendrímeros, micelas, micelas poliméricas, nanopartículas inorgânicas, nanopartículas de carbono, adesivos transdérmicos, matrices poliméricas implantáveis e dispositivos que possam ser implantados.  Thus, according to the present invention, fullerol may be associated with carrier nanosystems and / or controlled release drug systems, aiming at increased stability, increased bioavailability, longer action and / or targeting to the target tissue. Such classically known carrier or controlled release systems include, but are not limited to, cyclodextrins, polymers, mucoadhesive polymers, lipid vesicles, liposomes, polymersomes, solid lipid nanoparticles, polymeric micro- and nanoparticles, micro- and nanocapsules, micro- - and nanoemulsions, dendrimers, micelles, polymer micelles, inorganic nanoparticles, carbon nanoparticles, transdermal patches, implantable polymer matrices and implantable devices.
[043] Sabendo que o fulerol apresenta propriedades antioxidantes, o teste seguinte foi para avaliar se haveria algum benefício oriundo da associação do fulerol com o Glucantime, em termos de ganho de eficácia e de redução de toxicidade. Foi utilizado o modelo murino de leishmaniose visceral para avaliar, por PCR em tempo real quantitativa, a carga parasitária no fígado dos animais após os tratamentos. Observou-se uma redução significativa da carga parasitária hepática nos grupos tratados com Glucantime e Glucantime associado ao Fulerol encapsulado em lipossomas, quando comparado ao grupo controle. Todavia, a associação entre o fulerol e o Glucantime pareceu ser mais eficaz no tratamento da leishmaniose, quando comparada ao efeito do Fulerol ou do Glucantime isolado. Knowing that fullerol has antioxidant properties, the next test was to assess whether there would be any benefit from the combination of fullerol with Glucantime in terms of efficiency gains and toxicity reduction. The murine visceral leishmaniasis model was used to evaluate, by quantitative real-time PCR, the parasitic load on the liver of the animals after the treatments. A significant reduction in hepatic parasitic load was observed in the groups treated with Glucantime and Glucantime associated with liposome-encapsulated Fullerol when compared to the control group. However, the association between fullerol and Glucantime appeared to be more effective in treating leishmaniasis compared to the effect of Fullerol or Glucantime alone.
[044] Portanto, a presente invenção refere-se também a composições farmacêuticas que comprendem fulerenos associados a um ou mais fármacos leishmanicidas e estes serem: antimoniais pentavalentes, preferencialmente o antimoniato de meglumina; anfotericina B; alopurinol; cetoconazol; itraconazol; fluconazol; posaconazol; tucaresol; paramomicina; pentamidina; miltefosina, sitamaquina, iminoquimod; azitromicina; buparvaquona; tamoxifeno; terbinafina; furazolidona; fluoroquinolona; domperidona; derivado de alquil-lisofosfolipídeo ou de alquil-fosfolipídeo; aza-esterol; licocalcona A; maesabalid III; tricotecenos; n-acetil- cisteina; quinolina 3-substituída, ou a uma ou mais substâncias imunomoduladoras, preferencialmente peptídeos ou proteínas imunogênicas derivadas de Leishmania, interferon gama, citocinas, lipídeo A, oligonucleotídeos, imunoprotetores e antiinflamatórios esteroidais. Os fígados dos animais com leishmaniose visceral e submetidos à quimioterapia antimonial também foram utilizados para a análise de alterações histopatológicas e de apoptose utilizando o método de coloração HE por miscroscopia óptica. Alterações histopatológicas características de degeneração hidrópica foram observadas nos fígados dos animais que receberam Glucantime isolado, evidenciando a hepatotoxicidade do fármaco que pode ser atribuída à indução de estresse oxidativo. Contudo, essas alterações não foram observadas no grupo que recebeu a associação Glucantime+fulerol encapsulado em lipossomas, o que demonstra a ação hepatoprotetora do co-tratamento com o fulerol em sistema de nanocarreadores. Therefore, the present invention also relates to pharmaceutical compositions comprising fullerenes associated with one or more leishmanicidal drugs and these are: pentavalent antimonials, preferably meglumine antimoniate; amphotericin B; allopurinol; ketoconazole; itraconazole; fluconazole; posaconazole; tucaresol; paramomycin; pentamidine; miltefosine, sitamaquine, iminoquimod; azithromycin; buparvaquone; tamoxifen; terbinafine; furazolidone; fluoroquinolone; domperidone; alkyl lysophospholipid or alkyl phospholipid derivative; aza-sterol; licocalcona A; maesabalid III; trichothecenes; n-acetyl cysteine; 3-substituted quinoline, or one or more immunomodulatory substances, preferably Leishmania-derived immunogenic peptides or proteins, interferon gamma, cytokines, lipid A, oligonucleotides, immunoprotectors and anti-inflammatory steroids. Livers from animals with visceral leishmaniasis submitted to antimonial chemotherapy were also used for the analysis of histopathological changes and apoptosis using the HE staining method by optical mischroscopy. Histopathological changes characteristic of hydropic degeneration were observed in the livers of animals that received Glucantime alone, showing the hepatotoxicity of the drug that can be attributed to the induction of oxidative stress. However, these changes were not observed in the group that received the combination Glucantime + liposome-encapsulated fullerol, which demonstrates the hepatoprotective action of co-treatment with fullerol in nanocarrier system.
[045] Sendo assim, o fulerol demonstrou de forma surpreendente uma atividade antileishmania, assim como uma capacidade de reduzir a hepatotoxicidade ocasionada pelo Glucantime quando administrado em associação em modelo animal para leishmaniose visceral. Logo, o fulerol apresentou-se como sendo o primeiro fármaco a demonstrar a dupla capacidade de exercer ação antileishmania sem efeitos colaterais e de reduzir a toxicidade de outro fármaco leishmanicida sem interferir nas sua atividade farmacológica.  Thus, fullerol has surprisingly demonstrated an antileishmania activity as well as an ability to reduce the hepatotoxicity caused by Glucantime when given in combination in animal model for visceral leishmaniasis. Therefore, fullerol was the first drug to demonstrate the dual ability to exert antileishmania action without side effects and to reduce the toxicity of another leishmanicidal drug without interfering with its pharmacological activity.
[046] Dessa forma, o conjunto de resultados obtidos nos testes permitiu antecipar que composições farmacêuticas contendo o fármaco antimonial e fulerol, principalmente aquelas baseadas em lipossomas, apresentam atividade antileishmania mais elevada e toxicidade reduzida, quando comparado ao medicamento comercial Glucantime. Thus, the set of test results allowed us to anticipate that pharmaceutical compositions containing the antimonial drug and fullerol, especially those based on liposomes, have higher antileishmania activity and reduced toxicity when compared to the commercial drug Glucantime.
[047] Essa composições contendo fulerenos solúveis em água puros ou associados a fármacos leishmanicidas ou imunomoduladores podem ser ainda administradas por via parenteral, preferencialmente endovenosa, intramuscular, subcutânea, intralesional e intraperitoneal; via oral, preferencialmente na forma de cápsulas, drágeas, comprimidos, xaropes ou elixir; tópica, preferencialmente na forma de pomada, loção, pasta ou gel.  Such compositions containing pure water soluble fullerenes or associated with leishmanicidal or immunomodulatory drugs may further be administered parenterally, preferably intravenously, intramuscularly, subcutaneously, intralesionally and intraperitoneally; orally, preferably in the form of capsules, dragees, tablets, syrups or elixir; topical, preferably in the form of ointment, lotion, paste or gel.
[048] Na presente tecnologia as composições compreendem particularmente o fulerol associado com o antimoniato de meglumina encapsulados em lipossomas .  [048] In the present technology the compositions particularly comprise fullerol associated with liposome-encapsulated meglumine antimoniate.
[049] A invenção poderá ser melhor compreendida a partir dos exemplos, não limitantes, que seguem. The invention may be better understood from the following non-limiting examples.
Exemplo 1 : Atividade antileishmania do Fulerol  Example 1: Fulerol antileishmania activity
[050] Para avaliar se o Fulerol apresentaria atividade antileishmania, analisou-se a carga parasitária no fígado de camundongos BALB/c infectados experimentalmente com Leishmania infantum (modelo murino de leishmaniose visceral). Os animais foram divididos em quatro grupos (n = 9-10). Durante 20 dias, foram administradas por via intraperitoneal doses diárias de Glucantime (120 mg Sb/kg), Fulerol livre (0,05 mg/kg), Fulerol encapsulado em lipossomas (0,05 mg/kg) e Salina (controle negativo). Após o período de tratamento, os animais foram sacrificados para determinar a carga parasitária no fígado utilizando a técnica de PCR em tempo real quantitativa. A Figura 1 mostra uma redução significativa da carga parasitária hepática dos animais tratados com Glucantime, Fulerol e Fulerol encapsulado em lipossomas na dose de 0,05 mg/kg (*P < 0,05 com relação ao grupo controle; teste de Kruskal-Wallis com pós-teste de Dunn's). Portanto, estabelecemos pela primeira vez a atividade anti-leishmania do fulerol em modelo experimental de leishmaniose, que mostrou-se equivalente àquela do fármaco comercial Glucantime. [050] To assess whether Fulerol would have antileishmania activity, the parasitic load on the liver of BALB / c mice experimentally infected with Leishmania infantum (murine model of visceral leishmaniasis) was analyzed. The animals were divided into four groups (n = 9-10). Intraperitoneal doses of Glucantime (120 mg Sb / kg), free fullerol (0.05 mg / kg), liposome-encapsulated fullerol (0.05 mg / kg) and saline (negative control) were administered intraperitoneally for 20 days. . After the treatment period, the animals were sacrificed to determine the parasitic load on the liver using the quantitative real time PCR technique. Figure 1 shows a significant reduction in hepatic parasitic burden of animals treated with liposome-encapsulated Glucantime, Fulerol and Fullerol at a dose of 0.05 mg / kg ( * P <0.05 compared to the control group; Kruskal-Wallis test with Dunn's post-test's). Therefore, we established for the first time the anti-leishmania activity of fullerol in an experimental leishmaniasis model, which was equivalent to that of the commercial drug Glucantime.
Exemplo 2: Eficácia da associação do Fulerol com o Glucantime em modelo murino de leishmaniose visceral  Example 2: Effectiveness of Combining Fullerol with Glucantime in a Visceral Leishmaniasis Murine Model
[051 ] Avaliou-se a carga parasitária no fígado de camundongos BALB/c infectados com Leishmania infantum (n = 5-6/grupo) determinada por PCR em tempo real quantitativa, após 20 dias de tratamento por via intraperitoneal com fulerol encapsulado em lipossomas (0,05 mg/kg/dose com intervalo de 4 dias), Glucantime (120 mg Sb/kg/dia), Glucantime (120 mg Sb/kg/dia) associado ao Fulerol (0,05 mg/kg/dose/4 dias), Glucantime (120 mg Sb/kg/dia) associado ao Fulerol em lipossomas (0,05 mg/kg/dose/4 dias) ou salina. A Figura 2 mostra uma redução significativa da carga parasitária hepática apenas nos grupos tratados com Glucantime e Glucantime associado ao Fulerol encapsulado em lipossomas, quando comparado ao grupo controle (*P < 0,05 com relação ao grupo salina; teste de Kruskal-Wallis com pós-teste de Dunn's). Todavia, a associação entre o Fulerol e o Glucantime pareceu ser mais eficaz no tratamento da leishmaniose, quando comparada ao Fulerol ou Glucantime isolado. Portanto, esse exemplo estabelece o benefício da associação do Glucantime com o fulerol encapsulado em lipossomas, em termos de ganho de eficácia no tratamento da leishmaniose visceral. [051] The liver parasite burden of Leishmania infantum-infected BALB / c mice (n = 5-6 / group) determined by quantitative real-time PCR after 20 days of intraperitoneal treatment with fullerol was evaluated. encapsulated in liposomes (0.05 mg / kg / dose 4 days apart), Glucantime (120 mg Sb / kg / day), Glucantime (120 mg Sb / kg / day) plus Fullerol (0.05 mg / kg) / dose / 4 days), Glucantime (120 mg Sb / kg / day) associated with fullerol in liposomes (0.05 mg / kg / dose / 4 days) or saline. Figure 2 shows a significant reduction in hepatic parasite burden only in the groups treated with Glucantime and Glucantime associated with liposome-encapsulated fullerol when compared to the control group (* P <0.05 with respect to the saline group; Kruskal-Wallis test with post-test Dunn's). However, the association between Fullerol and Glucantime appeared to be more effective in treating leishmaniasis compared to Fullerol or Glucantime alone. Therefore, this example establishes the benefit of combining Glucantime with liposome-encapsulated fullerol in terms of improved efficacy in the treatment of visceral leishmaniasis.
Exemplo 3: Efeito quimioprotetor do Fulerol em modelo de leishmaniose visceral submetido à quimioterapia antimonial  Example 3: Chemoprotective effect of fullerol in a visceral leishmaniasis model submitted to antimonial chemotherapy
[052] Foi feita a análise histopatologica e de apoptose por microscopia óptica utilizando o método de coloração HE do fígado de camundongos BALB/c infectados experimentalmente com Leishmania infantum, após 20 dias de tratamento por via intraperitoneal com Glucantime (120 mg Sb/kg/dia), Glucantime (120 mg Sb/kg/dia) associado ao Fulerol (0,05 mg/kg/dose/4 dias), Glucantime (120 mg Sb/kg/dia) associado ao Fulerol em lipossomas (0,05 mg/kg/dose/4 dias) ou salina. Os parâmetros histopatológicos avaliados foram a degeneração hidrópica (A) e o índice apoptótico (B). A Figura 3, mostra alterações h isto pato lógicas características de degeneração hidrópica e aumento do índice apoptótico no fígado dos animais que receberam Glucantime evidenciando a hepatotoxicidade do fármaco. Essas alterações foram observadas também no grupo que recebeu a associação Glucantime + fulerol, mas não no grupo que recebeu a associação do Glucantime + fulerol encapsulado em lipossomas, demonstrando a ação hepatoprotetora do co- tratamento com fulerol encapsulado em lipossomas. Ou seja, além de exercer ação antileishmania, o fulerol em lipossomas mostrou uma ação quimioprotetora, evidenciada pela redução da hepatotoxicidade do Glucantime na presença do fulerol em lipossomas.  Histopathological and apoptotic analysis was performed by light microscopy using the HE liver staining method of experimentally infected Leishmania infantum infected BALB / c mice after 20 days of intraperitoneal Glucantime treatment (120 mg Sb / kg / kg). Glucantime (120 mg Sb / kg / day) associated with fullerol (0.05 mg / kg / dose / 4 days), Glucantime (120 mg Sb / kg / day) associated with fullerol liposomes (0.05 mg / kg / dose / 4 days) or saline. The histopathological parameters evaluated were hydropic degeneration (A) and apoptotic index (B). Figure 3 shows characteristic pathological changes of hydropic degeneration and increased apoptotic index in the liver of animals receiving Glucantime showing the hepatotoxicity of the drug. These changes were also observed in the group that received the Glucantime + fullerol combination, but not in the group that received the liposome-encapsulated Glucantime + fullerol combination, demonstrating the hepatoprotective action of co-treatment with liposome-encapsulated fullerol. That is, besides exerting antileishmania action, fullerol in liposomes showed a chemoprotective action, evidenced by the reduction of Glucantime hepatotoxicity in the presence of fullerol in liposomes.
Exemplo 4: Atividade antileishmania in vitro do fulerol  Example 4: In vitro antileishmania activity of fullerol
[053] Este estudo foi realizado em três etapas. Na primeira etapa, avaliou-se a faixa de concentração citotóxica do fulerol em macrófagos peritoneais. Na segunda etapa, investigou-se a atividade antileishmania do fulerol em concentração não citotóxica para os macrófagos, em modelo de macrófago infectado com Leishmania. Na terceira etapa, avaliou-se a concentração citotóxica do fulerol na forma promatigota de Leishmania. [053] This study was conducted in three steps. In the first stage, the cytotoxic concentration of fullerol in peritoneal macrophages was evaluated. In the second step, the antileishmania activity of fullerol in non-cytotoxic concentration for macrophages was investigated in a Leishmania infected macrophage model. In the third stage, the cytotoxic concentration of fullerol in the promatigote form of Leishmania was evaluated.
[054] Para os testes de citotoxicidade in vitro e de atividade antileishmania em modelo de macrófago infectado com Leishmania, macrófagos peritoneais foram isolados de camundongos BALB/c, 3 dias após injeção intraperitoneal de tioglicolato 3%, por lavagem peritoneal com solução meio RPMI gelado. As células foram lavadas, contadas e uma suspensão resultante de densidade de 1x106 células/ml_ foi obtida. As células foram mantidas a 37°C em meio RPMI 1 640 suplementado com 10% de soro fetal bovino, estreptomicina (50 pg/mL) e penicilina (1000 U/mL). [054] For in vitro cytotoxicity and antileishmania activity tests in a Leishmania-infected macrophage model, peritoneal macrophages were isolated from BALB / c mice 3 days after intraperitoneal injection of thioglycolate 3% by peritoneal lavage with cold RPMI medium. . The cells were washed, counted and a resulting suspension of 1x10 6 cells / ml density was obtained. Cells were maintained at 37 ° C in RPMI 1640 medium supplemented with 10% fetal bovine serum, streptomycin (50 pg / mL) and penicillin (1000 U / mL).
[055] No primeiro experimento, foi avaliada a citotoxicidade do fulerol em macrófagos peritoneais. Para isto, as células foram plaqueadas em placas de 96 poços na densidade de 5x103 células/poço e esperou-se a ocorrência de sua aderência. No dia seguinte, as células foram expostas aos compostos fulerol em concentrações crescentes (0 - 0,15mg/ml_) e foram mantidas a 37 °C em incubadora umidificada e atmosfera contendo 5% de CO2. Após 72 horas de exposição ao fulerol e glucantime, 3-[4,5-dimetiltiazol-2il]-2,5-difeniltetrazolium bromidetiazolil azul (MTT- 5 img/mL) foi adicionado a cada poço e aguardou-se 4 horas. O ensaio de MTT é baseado na redução do tetrazolium pela ação de monoxigenases mitocondriais das células viáveis, resultando na formação de formazan (Mosman, 1983). Após a formação de cristais de formazan, estes foram solubilizados em DMSO e lidos em leitor de Elisa a 570 nm. Os resultados foram mostrados como valores de absorbância e controles (não exposto ao antimonial e/ou fulerol). [055] In the first experiment, the cytotoxicity of fullerol in peritoneal macrophages was evaluated. For this, the cells were plated in 96-well plates at a density of 5x10 3 cells / well and their adherence was expected to occur. The following day, cells were exposed to fullerol compounds at increasing concentrations (0 - 0.15mg / ml) and were maintained at 37 ° C in a humidified incubator and atmosphere containing 5% CO 2. After 72 hours of exposure to fullerol and glucantime, 3- [4,5-dimethylthiazol-2yl] -2,5-diphenyltetrazolium bromidetiazolyl blue (MTT-5 µg / ml) was added to each well and allowed for 4 hours. The MTT assay is based on the reduction of tetrazolium by the action of viable mitochondrial monoxygenases resulting in formazan formation (Mosman, 1983). After formation of formazan crystals, they were solubilized in DMSO and read on an Elisa reader at 570 nm. Results were shown as absorbance values and controls (not exposed to antimonial and / or fullerol).
[056] No segundo experimento, as células foram plaqueadas em placas de 24 poços contendo lamínulas esféricas em cada poço, na densidade de 3x1 05 células/poço, e esperou-se por 1 hora para a ocorrência de sua aderência. Foi feita então a lavagem para a retirada dos macrófagos não aderentes e acrescentou 3x106 de células promastigotas de Leishmania infantum (cepa MHOM/BR/1970/BH46). Esperou-se 4 horas para a infecção e iniciou-se o tratamento expondo as amastigotas intramacrofágicas a uma concentração de fulerol (0,12mg/ml) e a concentrações crescentes de glucantime. Como controle, usou-se amastigotas intramacrofágicas não tratadas. As células foram mantidas em incubadora umidificada a 37SC e atmosfera contendo 5% de C02. Após 72 horas de exposição ao fulerol e/ou glucantime, as lamínulas foram retiradas e coradas em Giemsa, montadas em lâminas e analisadas em microscópio óptico. Os resultados de carga parasitária foram expressos como índice de Infecção obtido pela seguinte fórmula: (% de macrófagos infectados) x (amastigotas/macrófagos infectados)/100. Os dados foram submetidos a análise de variância simples One Way Anova seguido pelo teste de comparação múltipla de Bonferroni. [056] In the second experiment, cells were plated in 24-well plates containing spherical coverslips in each well at a density of 3x10 5 cells / well and allowed to adhere for 1 hour. The non-adherent macrophages were then washed and 3x10 6 of Leishmania infantum promastigote cells (strain MHOM / BR / 1970 / BH46) added. The infection was waited for 4 hours and treatment started by exposing the intramacrophagic amastigotes to fullerol concentration (0.12mg / ml) and increasing glucantime concentrations. As a control, untreated intramacrophagic amastigotes were used. Cells were maintained in a humidified incubator at 37 C and S atmosphere containing 5% C0 2. After 72 hours of exposure At fullerol and / or glucantime, the coverslips were removed and stained in Giemsa, mounted on slides and analyzed under an optical microscope. Parasite load results were expressed as Infection Index obtained by the following formula: (% of infected macrophages) x (amastigotes / infected macrophages) / 100. Data were submitted to One Way Anova simple variance analysis followed by Bonferroni multiple comparison test.
[057] No terceiro experimento, células promastigotas de Leishmania infantum (cepa BH46) foram mantidas em meio essencial mínimo de cultura (a-MEM, Gibco), suplementado com 10% de soro fetal bovino inativado, 100μg/mL de kanamicina e 50pg/ml_ de ampicilina e incubadas a 24±1 aC e pH 7,0 em estufa B.O.D. As promastigotas em fase logarítmica de crescimento foram colocadas em placas de 96 poços na densidade de 106 células/mL e expostas a concentrações crescentes de fulerol de 0,005 a 0,2 mg/ml. Replicatas na ausência da droga foram estabelecidas como controle. Após 72 horas, o crescimento foi avaliado através de medição da absorbância das culturas a 600nm utilizando um sistema automatizado de leitura de microplacas. [057] In the third experiment, Leishmania infantum promastigote cells (strain BH46) were maintained in minimal essential culture medium (a-MEM, Gibco) supplemented with 10% inactivated fetal bovine serum, 100μg / mL kanamycin and 50pg / mL ampicillin and incubated at 24 ± 1 ° C and at pH 7.0 in an oven BOD the promastigotes in logarithmic growth phase were placed into 96 - well plates at a density of 10 6 cells / ml, exposed to increasing concentrations of 0.005 fulerol at 0.2 mg / ml. Replicates in the absence of the drug were established as controls. After 72 hours, growth was evaluated by measuring the absorbance of the cultures at 600nm using an automated microplate reading system.
[058] Como mostrado na Figura 4, até a concentração de 0,12 mg/ml, não há efeito significativo do fulerol na viabilidade celular. Em contrapartida, a partir de 0,15mg/ml o valor de absorbância começa a decrescer significativamente indicando diminuição da viabilidade celular. Este dado nos permite inferir que a concentração máxima de fulerol a ser usada, sem causar dano celular é de 0,12 mg/ml.  [058] As shown in Figure 4, up to the concentration of 0.12 mg / ml, there is no significant effect of fullerol on cell viability. In contrast, from 0.15mg / ml the absorbance value begins to decrease significantly indicating decreased cell viability. This data allows us to infer that the maximum concentration of fullerol to be used without causing cell damage is 0.12 mg / ml.
[059] Para avaliar a atividade antileishmania do fulerol, este foi utilizado em concentração máxima não citotóxica (0,12 mg/ml) no modelo de macrófago infectado por Leishmania infantum (cepa BH46). Como mostrado na Figura 5, o fulerol sozinho reduziu a carga parasitária em cerca de 75% quando comparado ao grupo controle (barra escura). O efeito foi equivalente aquele do Glucantime dado em concentração de Sb 0,75 mg/ml. [059] To evaluate the fullerol antileishmania activity, it was used at a maximum non-cytotoxic concentration (0.12 mg / ml) in the Leishmania infantum-infected macrophage model (strain BH46). As shown in Figure 5, fullerol alone reduced the parasite burden by about 75% compared to the control group (dark bar). The effect was equivalent to that of Glucantime given at a concentration of Sb 0.75 mg / ml.
[060] No estudo de crescimento de células promastigotas de Leishmania infantum na presença de concentrações crescentes de fulerol, na faixa de 0 a 0,2 mg/ml, não foi detectada ação citotóxica do fulerol.  In the study of promastigote cell growth of Leishmania infantum in the presence of increasing concentrations of fullerol in the range of 0 to 0.2 mg / ml, no cytotoxic action of fullerol was detected.
[061 ] Em conclusão, foi demonstrada a ação antileishmania do fulerol no modelo de macrófago infectado por Leishmania infantum. O fato de que o fulerol não exerceu citotoxicidade no modelo de promastigotas sugere uma ação indireta do fulerol no parasita através de ativação do macrófago. Exemplo 5: Influência de diferentes doses de fulerol encapsulado em lipossomas na atividade antileishmania e no perfil de resposta imune, em modelo murino de leishmaniose visceral [061] In conclusion, the antileishmania action of fullerol in the Leishmania infantum-infected macrophage model has been demonstrated. The fact that fullerol did not exert cytotoxicity in the promastigote model suggests an indirect action of fullerol on the parasite through macrophage activation. Example 5: Influence of different doses of liposome-encapsulated fullerol on antileishmania activity and immune response profile in a murine model of visceral leishmaniasis
[062] Este estudo teve como objetivo avaliar se a administração do fulerol encapsulado em lipossomas em doses crescentes resultaria em maior eficácia em modelo murino de leishmaniose visceral, bem como a sua possível ação imunomoduladora in vivo.  [062] This study aimed to evaluate whether administration of liposome-encapsulated fullerol at increasing doses would result in greater efficacy in a murine model of visceral leishmaniasis, as well as its possible immunomodulatory action in vivo.
Avaliação de eficácia in vivo do tratamento com as formulações In vivo efficacy evaluation of formulation treatment
[063] Para os testes de eficácia, foram utilizados camundongos BALB/c fêmeas, com idade de 6-8 semanas e peso de 20-25g provenientes do Centro de Bioterismo do Instituto de Ciências Biológicas da UFMG - CEBIO. Os procedimentos utilizados foram aprovados pelo Comité de Ética em Experimentação Animal da UFMG (protocolo nQ 46/2013). [063] For efficacy testing, female BALB / c mice, aged 6-8 weeks and weighing 20-25g from the Bioterismo Center of the UFMG - CEBIO Institute of Biological Sciences, were used. The procedures used were approved by the UFMG Animal Experimentation Ethics Committee (protocol Q 46/2013).
[064] Os animais foram infectados pela via intraperitoneal com inoculo de Animals were infected intraperitoneally with inoculum of
2χ107/100μΙ promastigotas de L. Infantum (cepa MHOM/BR/1970/BH46). 2χ10 7 / 100μΙ L. Infantum promastigotes (strain MHOM / BR / 1970 / BH46).
[065] O tratamento iniciou-se 7 dias após inoculação da cepa BH46 e teve duração de 20 dias. Os animais foram alocados em 6 grupos experimentais (n = 6 por grupo) que receberam os seguintes tratamentos por via intraperitoneal:  [065] Treatment began 7 days after BH46 strain inoculation and lasted 20 days. The animals were allocated to 6 experimental groups (n = 6 per group) receiving the following intraperitoneal treatments:
[066] Grupo 1 : tratado com o glucantime (120mg Sb/kg/dia - 20 doses);  Group 1: treated with glucantime (120mg Sb / kg / day - 20 doses);
[067] Grupo 2: tratado com o fulerol livre em solução NaCI 0,15 M (0,05mg/kg/4dias Group 2: treated with free fullerol in 0.15 M NaCl solution (0.05mg / kg / 4 days
- 5 doses); - 5 doses);
[068] Grupo 3: tratado com solução NaCI 0,15 M contendo fulerol encapsulado em lipossomas (0,05mg/kg/4dias - 5 doses);  Group 3: treated with 0.15 M NaCl solution containing liposome-encapsulated fullerol (0.05mg / kg / 4 days - 5 doses);
[069] Grupo 4: tratado com solução NaCI 0,15 M contendo fulerol encapsulado em lipossomas (0,2mg/kg/4dias - 5 doses);  Group 4: treated with 0.15 M NaCI solution containing liposome encapsulated fullerol (0.2mg / kg / 4 days - 5 doses);
[070] Grupo 5: tratado com solução de lipossomas vazios (mesma dose de lipídeos de que nos grupos 4 e 5);  Group 5: treated with empty liposome solution (same lipid dose as in groups 4 and 5);
[071 ] Grupo 6: tratado com salina (controle).  [071] Group 6: saline treated (control).
[072] Ao final do tratamento, os animais foram submetidos à eutanásia por deslocamento cervical, após sedação com 8mg/kg cetamina e 5mg/kg de xilazina via intraperitoneal. Os órgãos fígado e baço foram coletados para determinação de carga parasitária por PCR quantitativa (qPCR). [073] Para determinar o perfil de citocinas no fígado, os órgãos foram pesados individualmente e homogeneizados em PBS contendo Tween 20 a 0,05% e inibidores de protease (phenylmethilsulfonyl fluoride 0,1 m M; benzethonium chloride 0,1 mM; EDTA 10 mM e aprotinin A 20 Kl). Após centrifugação a 8,000g por 10 min a 4 QC, os sobrenadantes foram utilizados para dosagem das citocinas (IL-4, IL-10, TNF-alfa e IFN-gamma) por ELISA usando kit comercial e seguindo as instruções do fabricante. [072] At the end of treatment, the animals were euthanized by cervical dislocation after sedation with 8mg / kg ketamine and 5mg / kg xylazine intraperitoneally. Liver and spleen organs were collected for determination of parasite load by quantitative PCR (qPCR). [073] To determine the cytokine profile in the liver, the organs were individually weighed and homogenized in PBS containing 0.05% Tween 20 and protease inhibitors (0.1 mM phenylmethilsulfonyl fluoride; 0.1 mM benzethonium chloride; EDTA 10 mM and aprotinin A 20 Kl). After centrifugation at 8,000g for 10 min at 4 Q C, the supernatants were used for determination of cytokines (IL-4, IL-10, TNF-alpha and IFN-gamma) by ELISA using a commercial kit and following the manufacturer's instructions.
[074] A Figura 6 (A e B) e a Figura 7 mostram os resultados da carga parasitária no fígado e baço, respectivamente. Podemos observar uma redução significativa da carga parasitária tanto no fígado quanto no baço, nos grupos tratados com fulerol encapsulado em lipossomas (0,05 e 0,2 mg/kg) e com Glucantime, quando comparados ao grupo salina. É também interessante notar que o fulerol livre administrado a cada 4 dias não promoveu redução significativa da carga parasitária e que a forma encapsulada foi significativamente mais efetiva que a forma livre. O aumento da dose de fulerol lipossomal de 0,05 para 0,2 mg/kg resultou em redução mais pronunciada da carga parasitária (Figura 6A e Figura 7), sendo que não foi possível detectar o parasito no fígado dos animais dentro o limite de detecção do método de PCR (Figura 6B).  [074] Figure 6 (A and B) and Figure 7 show the results of parasitic burden on the liver and spleen, respectively. Significant reduction in parasite load in both liver and spleen was observed in the liposome-encapsulated fullerol (0.05 and 0.2 mg / kg) and Glucantime treated groups compared to the saline group. It is also interesting to note that free fullerol administered every 4 days did not significantly reduce parasite load and that the encapsulated form was significantly more effective than free form. Increasing the liposomal fullerol dose from 0.05 to 0.2 mg / kg resulted in a more pronounced reduction in parasitic load (Figure 6A and Figure 7), and it was not possible to detect the parasite in the liver of animals within detection of the PCR method (Figure 6B).
[075] É importante ressaltar que o tratamento com fulerol lipossomal mostrou-se significativamente mais efetivo que o tratamento convencional com Glucantime.  [075] Importantly, liposomal fullerol treatment was significantly more effective than conventional Glucantime treatment.
[076] Visando esclarecer o mecanismo de ação do fulerol na eliminação do parasito no fígado e evidenciar uma possível ação imunomoduladora, foi avaliado o perfil da resposta imunológica ao final do tratamento, através da dosagem das citocinas IL-4, IL-10, TNF-alfa e IFN-gamma no tecido. Como mostrado na Figura 8, na análise comparativa entre os grupos tratados e o controle (salina), o Fulerol lipossomal na dose de 0,2 mg/kg induziu uma perfil imunológico proinflamatório, com aumento significativo de TNF-alfa e IFN-gamma, sem variação significativa de IL-10 e IL-4. Por outro lado, o grupo Glucantime mostrou indução de apenas IL-4 e IL-10. [076] In order to clarify the mechanism of action of fullerol in the elimination of the parasite in the liver and to show a possible immunomodulatory action, the profile of the immune response at the end of treatment was evaluated by measuring the cytokines IL-4, IL-10, TNF. alpha and IFN-gamma in the tissue. As shown in Figure 8, in the comparative analysis between treated and control (saline) groups, liposomal fullerol at a dose of 0.2 mg / kg induced a proinflammatory immune profile, with a significant increase in TNF-alpha and IFN-gamma, no significant variation of IL-10 and IL-4. On the other hand, the Glucantime group showed induction of only IL-4 and IL-10.
[077] Essa dado evidencia uma forte ação imunomoduladora do fulerol lipossomal no fígado que contribuiu muito provavelmente para a eliminação do parasito neste tecido. TNF-alfa é uma citocina conhecida por estimular hepatócitos a sintetizar e secretar proteínas de fase inflamatória aguda (proteína ligada a manose, proteína C reativa) que servem para ativar sistema complemento e quimiotaxia de células fagocíticas (células dendríticas, macrófagos e neutrófilos). [078] Em conclusão, demonstramos que: i) o fulerol administrado na forma lipossomal em dose de 0,2 mg/kg/4dias promove uma forte redução da carga parasitária no fígado e no baço em modelo murino de leishmaniose visceral, sendo que houve eliminação completa do parasito no fígado; ii) a atividade antileishmania da formulação lipossomal é muito maior que aquela do medicamento padrão (Glucantime); iii) a eficácia da formulação lipossomal no fígado parece resultar da indução de perfil imunológico proinflamatório; iv) a formulação lipossomal do fulerol pode encontrar aplicações no tratamento de outras patologias acometendo o fígado e para as quais um perfil imunológico proinflamatório seria beneficiai. [077] This data shows a strong immunomodulatory action of liposomal fullerol in the liver that most likely contributed to the elimination of the parasite in this tissue. TNF-alpha is a cytokine known to stimulate hepatocytes to synthesize and secrete acute inflammatory phase proteins (mannose-linked protein, C-reactive protein) that serve to activate phagocytic complement and chemotaxis system (dendritic cells, macrophages and neutrophils). In conclusion, we have shown that: i) Liposomal fullerol administered at a dose of 0.2 mg / kg / 4 days promotes a marked reduction in parasite burden in the liver and spleen in a murine model of visceral leishmaniasis, and complete elimination of the parasite in the liver; ii) the antileishmania activity of the liposomal formulation is much higher than that of the standard drug (Glucantime); iii) the efficacy of the liposomal formulation in the liver seems to result from the induction of proinflammatory immune profile; iv) the liposomal formulation of fullerol may find applications in the treatment of other conditions affecting the liver and for which a proinflammatory immune profile would be beneficial.

Claims

REIVINDICAÇÕES
1. COMPOSIÇÕES FARMACÊUTICAS ANTILEISHMANIA caracterizadas por compreender fulerenos solúveis em água. 1. ANTILEISHMANY PHARMACEUTICAL COMPOSITIONS characterized by comprising water soluble fullerenes.
2. COMPOSIÇÕES FARMACÊUTICAS ANTILEISHMANIA de acordo com a reivindicação 1 , caracterizadas pelos fulerenos solúveis em água possuírem de 60 a 1 20 carbonos com adição de grupamentos hidroxila.  ANTILEISHMANY PHARMACEUTICAL COMPOSITIONS according to Claim 1, characterized in that the water-soluble fullerenes have from 60 to 120 carbons with the addition of hydroxyl groups.
3. COMPOSIÇÕES FARMACÊUTICAS ANTILEISHMANIA de acordo com as reivindicações 1 e 2, caracterizadas pelo fulereno ser preferencialmente o fulerol ANTILEISHMANE PHARMACEUTICAL COMPOSITIONS according to Claims 1 and 2, characterized in that fullerene is preferably fullerol.
(C6oOH22-24)-(C60 OH22-24) -
4. COMPOSIÇÕES FARMACÊUTICAS ANTILEISHMANIA de acordo com as reivindicações de 1 a 3, caracterizadas pelos fulerenos solúveis em água estarem associados ou não a um ou mais fármacos leishmanicidas ou imunomoduladores. ANTILEISHMANY PHARMACEUTICAL COMPOSITIONS according to Claims 1 to 3, characterized in that the water-soluble fullerenes are associated or not with one or more leishmanicidal or immunomodulatory drugs.
5. COMPOSIÇÕES FARMACÊUTICAS ANTILEISHMANIA de acordo com a reivindicação 4, caracterizadas pelos fármacos leishmanicidas serem selecionados do grupo compreendendo antimoniais pentavalentes, preferencialmente o antimoniato de meglumina; anfotericina B; alopurinol; cetoconazol; itraconazol; fluconazol; posaconazol; tucaresol; paramomicina; pentamidina; miltefosina, sitamaquina, iminoquimod; azitromicina; buparvaquona; tamoxifeno; terbinafina; furazolidona; fluoroquinolona; domperidona; derivado de alquil-lisofosfolipídeo ou de alquil-fosfolipídeo; aza-esterol; licocalcona A; maesabalid III; tricotecenos; n-acetil-cisteina; quinolina 3-substituída e os imunomoduladores serem selecionados do grupo compreendedo peptídeos ou proteínas imunogênicas derivadas de Leishmania, interferon gama, citocinas, lipídeo A, oligonucleotídeos, imunoprotetores e antiinflamatórios esteroidais.  ANTILEISHMANIA PHARMACEUTICAL COMPOSITIONS according to claim 4, characterized in that the leishmanicidal drugs are selected from the group comprising pentavalent antimonials, preferably meglumine antimoniate; amphotericin B; allopurinol; ketoconazole; itraconazole; fluconazole; posaconazole; tucaresol; paramomycin; pentamidine; miltefosine, sitamaquine, iminoquimod; azithromycin; buparvaquone; tamoxifen; terbinafine; furazolidone; fluoroquinolone; domperidone; alkyl lysophospholipid or alkyl phospholipid derivative; aza-sterol; licocalcona A; maesabalid III; trichothecenes; n-acetyl cysteine; 3-substituted quinoline and immunomodulators are selected from the group comprising Leishmania-derived immunogenic peptides or proteins, interferon gamma, cytokines, lipid A, oligonucleotides, immunoprotectants, and steroidal anti-inflammatories.
6. COMPOSIÇÕES FARMACÊUTICAS ANTILEISHMANIA de acordo com a reivindicação de 1 a 5, caracterizadas pelos fulerenos solúveis em água estarem associados ou não a nanossistemas carreadores e/ou sistemas de liberação controlada de fármacos selecionados do grupo compreendendo ciclodextrinas, polímeros, polímeros mucoadesivos, vesículas lipídicas, lipossomas, polimersomas, nanopartículas lipídicas sólidas, micro- e nanopartículas poliméricas, micro- e nanocapsulas, micro- e nanoemulsões, dendrímeros, micelas, micelas poliméricas, nanopartículas inorgânicas, adesivos transdérmicos, matrices poliméricas implantáveis e dispositivos que possam ser implantados. ANTILEISHMANY PHARMACEUTICAL COMPOSITIONS according to Claim 1 to 5, characterized in that the water-soluble fullerenes are associated or not with carrier-based nanosystems and / or controlled release systems selected from the group comprising cyclodextrins, polymers, mucoadhesive polymers, lipid vesicles. , liposomes, polymersomes, solid lipid nanoparticles, polymeric micro- and nanoparticles, micro- and nanocapsules, micro- and nanoemulsions, dendrimers, micelles, polymeric micelles, inorganic nanoparticles, transdermal patches, implantable polymer matrices and implantable devices.
7. COMPOSIÇÕES FARMACÊUTICAS ANTILEISHMANIA de acordo com as reivindicações de 1 a 6, caracterizadas pelos fulerenos solúveis em água estarem ou não encapsulados em lipossomas, podendo os lipossomas ser compostos por fosfolipídeos, colesterol e um lipídeo com carga negativa ou positiva, na presença ou ausência e de polímeros e/ou ligantes acoplados em suas superfícies. ANTILEISHMANY PHARMACEUTICAL COMPOSITIONS according to Claims 1 to 6, characterized in that the water-soluble fullerenes are or are not encapsulated in liposomes, and the liposomes may be composed of phospholipids, cholesterol and a negatively or positively charged lipid in the presence or absence. and of polymers and / or binders coupled to their surfaces.
8. COMPOSIÇÕES FARMACÊUTICAS ANTILEISHMANIA de acordo com a reivindicação de 7, caracterizadas pelos lipossomas serem constituídos preferencialmente de fosfatidilcolina, colesterol e fosfatidilglicerol. ANTILEISHMANIA PHARMACEUTICAL COMPOSITIONS according to claim 7, characterized in that the liposomes are preferably composed of phosphatidylcholine, cholesterol and phosphatidylglycerol.
9. COMPOSIÇÕES FARMACÊUTICAS ANTILEISHMANIA de acordo com as reivindicações de 1 a 6, caracterizadas por serem administradas por via parenteral, preferencialmente endovenosa, intramuscular, subcutânea, intralesional e intraperitoneal; via oral, preferencialmente na forma de cápsulas, drágeas, comprimidos, xaropes ou elixir; tópica, preferencialmente na forma de pomada, loção, pasta ou gel. ANTILEISHMANIA PHARMACEUTICAL COMPOSITIONS according to claims 1 to 6, characterized in that they are administered parenterally, preferably intravenously, intramuscularly, subcutaneously, intralesionally and intraperitoneally; orally, preferably in the form of capsules, dragees, tablets, syrups or elixir; topical, preferably in the form of ointment, lotion, paste or gel.
10. USO DAS COMPOSIÇÕES FARMACÊUTICAS ANTILEISHMANIA descritas nas reivindicações de 1 a 9, caracterizado por ser na preparação de medicamentos para o tratamento das leishmanioses visceral, mucocutânea e cutânea, preferencialmente a leishmaniose visceral e as composições compreenderem preferencialmente o fulerol associado com o antimoniato de meglumina encapsulados em lipossomas. USE OF THE ANTILEISHMANIA PHARMACEUTICAL COMPOSITIONS described in claims 1 to 9, characterized in that they are in the preparation of medicaments for the treatment of visceral, mucocutaneous and cutaneous leishmaniasis, preferably visceral leishmaniasis and the compositions preferably comprise the fullerol associated with meglumine antimoniate. encapsulated in liposomes.
PCT/IB2014/067384 2013-12-30 2014-12-29 Antileishmanial compositions containing fullerol and use thereof WO2015101918A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
BRBR1020130338664 2013-12-30
BR102013033866A BR102013033866A2 (en) 2013-12-30 2013-12-30 antileishmania compositions containing fullerol and use
BRBR1020140324470 2014-12-23
BR102014032447-0A BR102014032447B1 (en) 2014-12-23 2014-12-23 ANTILEISHMANIA COMPOSITIONS CONTAINING FULEROL AND USE

Publications (1)

Publication Number Publication Date
WO2015101918A1 true WO2015101918A1 (en) 2015-07-09

Family

ID=53493341

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2014/067384 WO2015101918A1 (en) 2013-12-30 2014-12-29 Antileishmanial compositions containing fullerol and use thereof

Country Status (1)

Country Link
WO (1) WO2015101918A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TW516958B (en) * 1997-10-29 2003-01-11 Nat Science Council Water-soluble fullerenol pharmaceutical composition
BRPI1107183A2 (en) * 2011-12-29 2013-11-19 Univ Minas Gerais PHARMACEUTICAL COMPOSITIONS CONTAINING POLYHYDROXY-FULLERENE [C60 (OH) N] AND USE

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TW516958B (en) * 1997-10-29 2003-01-11 Nat Science Council Water-soluble fullerenol pharmaceutical composition
BRPI1107183A2 (en) * 2011-12-29 2013-11-19 Univ Minas Gerais PHARMACEUTICAL COMPOSITIONS CONTAINING POLYHYDROXY-FULLERENE [C60 (OH) N] AND USE

Similar Documents

Publication Publication Date Title
Gutiérrez et al. New approaches from nanomedicine for treating leishmaniasis
Zarif et al. Cochleates: new lipid-based drug delivery system
Pham et al. Strategies for the design of orally bioavailable antileishmanial treatments
US20140037715A1 (en) Disulfiram formulation and uses thereof
WO2015023858A2 (en) Uses of phospholipid conjugates of synthetic tlr7 agonists
TW200936182A (en) Agent for enhancing anti-tumor effect comprising oxaliplatin liposome preparation, and anti-tumor agent comprising the liposome preparation
CN115484984A (en) Method for improving stability of pharmaceutical composition comprising high penetration drug and pharmaceutical composition obtained thereby
Duran et al. State of the art of nanobiotechnology applications in neglected diseases
ES2333889T3 (en) PHARMACEUTICAL COMPOSITIONS FOR THE TREATMENT OF LEISHMANIASIS.
Ramos et al. Antileishmanial activity of fullerol and its liposomal formulation in experimental models of visceral leishmaniasis
WO2015101918A1 (en) Antileishmanial compositions containing fullerol and use thereof
De Santana et al. Nanotechnology as an alternative to improve the treatment of cutaneous leishmaniasis: A systematic review of the literature
BR102014032447B1 (en) ANTILEISHMANIA COMPOSITIONS CONTAINING FULEROL AND USE
BR102013033866A2 (en) antileishmania compositions containing fullerol and use
JP4966855B2 (en) Sterol-enriched mixed layered amphotericin-inserted liposome in saline and method for preparing the same
Frézard et al. Nanostructures for improved antimonial therapy of leishmaniasis
Soni et al. Novel Therapeutic Approaches for the Treatment of Leishmaniasis
de Melo Barbosa et al. Lipid nanoparticles for the treatment of neglected tropical diseases
US9700540B2 (en) Dronedarone for use in leishmaniasis, formulations and associations for use in leishmaniasis
US20040265366A1 (en) Metallothioneine-containing liposomes
US20220370412A1 (en) Inhibition of neurological injuries due to infections via administration of butanetap and analogs thereof
Gupta et al. Leishmaniasis-drugs, nanotechnology based delivery systems and recent patents survey
JP2019520407A (en) Cochleated antifungal compounds for central nervous system delivery and treatment of cryptococcal infections
US20200069585A1 (en) Compositions for upregulating defensin expression
BR102020022824A2 (en) Composition, pharmaceutical composition, use of a stable topical composition comprising a nanoemulsion, and of at least one antileishmanial compound, and, method for treating cutaneous leishmaniasis

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: 14876606

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: 14876606

Country of ref document: EP

Kind code of ref document: A1