WO2013010238A1 - Compositions pharmaceutiques microparticulaires contenant des antiparasitaires pour traitement sous-cutané prolongé, utilisation desdites compositions pharmaceutiques pour la production d'un médicament, et méthode de traitement de parasitoses - Google Patents

Compositions pharmaceutiques microparticulaires contenant des antiparasitaires pour traitement sous-cutané prolongé, utilisation desdites compositions pharmaceutiques pour la production d'un médicament, et méthode de traitement de parasitoses Download PDF

Info

Publication number
WO2013010238A1
WO2013010238A1 PCT/BR2012/000249 BR2012000249W WO2013010238A1 WO 2013010238 A1 WO2013010238 A1 WO 2013010238A1 BR 2012000249 W BR2012000249 W BR 2012000249W WO 2013010238 A1 WO2013010238 A1 WO 2013010238A1
Authority
WO
WIPO (PCT)
Prior art keywords
plga
amphotericin
pharmaceutical compositions
particles
drugs
Prior art date
Application number
PCT/BR2012/000249
Other languages
English (en)
Portuguese (pt)
Inventor
Bartira ROSSI BERGMANN
Wallace PACIENZA LIMA
Camila ALVES BANDEIRA FALCÃO
Original Assignee
Universidade Federal Do Rio De Janeiro - Ufrj
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Universidade Federal Do Rio De Janeiro - Ufrj filed Critical Universidade Federal Do Rio De Janeiro - Ufrj
Publication of WO2013010238A1 publication Critical patent/WO2013010238A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7048Compounds having saccharide radicals and heterocyclic rings having oxygen as a ring hetero atom, e.g. leucoglucosan, hesperidin, erythromycin, nystatin, digitoxin or digoxin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/12Ketones
    • A61K31/121Ketones acyclic
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/28Compounds containing heavy metals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/555Heterocyclic compounds containing heavy metals, e.g. hemin, hematin, melarsoprol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/16Agglomerates; Granulates; Microbeadlets ; Microspheres; Pellets; Solid products obtained by spray drying, spray freeze drying, spray congealing,(multiple) emulsion solvent evaporation or extraction
    • A61K9/1605Excipients; Inactive ingredients
    • A61K9/1629Organic macromolecular compounds
    • A61K9/1641Organic macromolecular compounds obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyethylene glycol, poloxamers
    • A61K9/1647Polyesters, e.g. poly(lactide-co-glycolide)
    • 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

  • the invention relates to a pharmaceutical composition containing amphotericin B or antimonial compounds such as meglumine antimoniate, sodium stibogluconate and hydrated antimony (III) potassium tartrate (Sb 111 ), or a nitrated chalcone, in which case it would be 3-nitro-2-hydroxy-4,6-dimethoxycyclones which we have named it CH8.
  • amphotericin B or antimonial compounds such as meglumine antimoniate, sodium stibogluconate and hydrated antimony (III) potassium tartrate (Sb 111 ), or a nitrated chalcone, in which case it would be 3-nitro-2-hydroxy-4,6-dimethoxycyclones which we have named it CH8.
  • CH8 nitrate chalcones
  • This invention also relates to the use of these pharmaceutical compositions, a medicament and the treatment of parasitic diseases.
  • Parasitic diseases including malaria, leishmaniasis, trypanosomes, among others, are considered one of the major public health problems in the world. These diseases, however, are concentrated in countries with low-income populations, such as countries in Africa, Asia, and South America. This is largely due to the lack of public policies on health and sanitation, these countries have the highest number of cases and deaths from these parasites.
  • Leishmaniasis is a complex of diseases caused by various protozoan species of the genus Leishmania, which can cause a spectrum of clinical forms in humans and other vertebrates. These can be classified into Cutaneous form, which includes the simple, diffuse, mucocutaneous, often disfiguring, and often lethal Visceral forms.
  • the parasite exists as a flagellated promastigote in the phlebotomine vector, and as a flush amastigote within mononuclear phagocytes of the vertebrate host.
  • the privileged intracellular location makes it difficult to access active drugs, which makes this disease difficult to treat.
  • Antimonial salts are synthetically obtained from antimonic acid and N-methyl glucamine.
  • the administration Intramuscular or intravenous antimoniate is rapidly absorbed, so that after 48 hours 90% of the initial dose has already been excreted by the kidneys. Repeated high doses are required to maintain the therapeutic level of antimony in the tissues. This results in a variety of side effects such as myalgia, pancreatitis, pancytopenia, hepato and cardiotoxicity. Pancreatitis seems to be the frequent cause of nausea and abdominal pain (GASSER et al, 1994).
  • Pentamidine has similar efficacy to antimonials (TUON et al 2008), but produces other side effects such as hypoglycemia, diabetes, tachycardia, hypotension, nephrotoxicity and local pain on alternate 4mg / kg.day administration for 30 days intramuscularly .
  • Amphotericin B is a polyenic antibiotic isolated from the bacteria Streptomyces sp, initially used as an antifungal drug, especially in systemic infections.
  • a systemic drug should remain in circulation for as long as necessary for its therapeutic effect, within a safe range of high efficacy combined with low toxicity, and a minimum of repetitive doses.
  • encapsulating a drug in appropriate micro and nanoparticles can help direct it to your target tissue or cell.
  • Controlled drug release systems have several advantages over conventional systems, such as: 1) Greater control of the release of the active ingredient, reducing the appearance of toxic and subtherapeutic doses; 2) Use of less quantity of active principle; 3) Resulting in lower cost; 4) Longer administration interval; better treatment acceptance by the patient and the possibility of directing the active ingredient to its specific target.
  • liposomes The first to be developed, and the only ones already approved for intravenous systemic use, are liposomes. From the 1980s onwards various biodegradable polymers were and are being used in the preparation of micro and nanoparticles, including synthetic poly (lactic co-glycolic) polymers (PLGA), in addition to chitosan and polyhydroxyalkanoate (PHA) biopolymers. Of smaller size, in the order of 1 -10 nm, we have the cyclodextrin; and more recently spherical and branched polymers, dendrimers, which are being studied for their pharmaceutical viability.
  • synthetic poly (lactic co-glycolic) polymers PLGA
  • PHA polyhydroxyalkanoate
  • Liposomes were the first particles delineated for antileishmanial drug vectorization.
  • Amphotericin B encapsulated in negatively charged unilamellar liposomes (SUNDAR & RAI, 2002).
  • Recent studies on the toxicity and effectiveness of meglumine antimoniate liposomal formulations in dogs with visceral leishmaniasis have shown that the formulation significantly reduced the parasitic burden of bone marrow in dogs and did not cause significant changes in liver and renal function in the dose (6.5 mg Sb / kg / dose) with four doses (4 days apart) (FRÉZARD, F. & MICHALICK, MS M, 2008).
  • lipid-associated amphotericin B formulations have led to a remarkable improvement in leishmaniasis chemotherapy.
  • These lipid-associated formulations such as AmBisome®, encapsulated in unilamellar liposomes, Abelcet®, incorporated into lipid complex; and Amphocil® as a colloidal suspension in cholesterol were evaluated in clinical tests for visceral and mucocutaneous leishmaniasis.
  • AmBisome® has shown greater efficiency by favoring its approval by the Food and Drug Administration (FDA).
  • Side effects mainly reduced nephrotoxicity by decreasing renal elimination and increasing plasma half-life.
  • Another advantage is the possibility of administering larger doses of the drug in a shorter period of time.
  • the limiting factor of the use of these liposomal formulations is the high phosphatidylcholine, which makes the access to treatment impossible for most of the affected population (GOLENSER et al. 1999).
  • the polymeric micro and nanoparticle polymer systems are drug carrier systems that include capsules and spheres. Its major advantage over liposomes is its greater stability and can be lyophilized for storage. Factors such as type and proportion of polymers in the formulation determine important characteristics such as stability and crystalline structure and biodegradability, which influences the release rate of the drug (SCHAFFAZICK & GUTERRES, 2003).
  • Poly (lactide) polymer, or PLA is one of the most studied for biological application, due to its high biodegradability and biocompatibility, having already approved clinical use in the constitution of sutures and biodegradable orthopedic prostheses.
  • PLA nanospheres have also been successfully tested as carriers of various antileishmaniasis, such as primaquine, pentamidine, bacopasaponin and atovaquone, for the experimental treatment of intravenous visceral leishmaniasis.
  • various antileishmaniasis such as primaquine, pentamidine, bacopasaponin and atovaquone
  • 200 nm PLA nanoparticles were successfully tested by our group on the delivery of nitrated chalcone (CH8) DMC for subcutaneous treatment of L. amazonensis infected mice (TORRES-SANTOS er a /., 1999). .
  • CH8 DMC nitrated chalcone
  • PLGA is also interesting as a controlled drug delivery system. This polymer has also been approved by the FDA as biodegradable structures used in orthopedic screws and studs, sutures, and meniscus and cartilage repair systems (ELIAZ and KOST, 2000). In addition, PLGA is widely used in cosmetic medicine for subcutaneous wrinkle filling, with a high degree of acceptance by patients. Polymer formulations approved for human use are all directed to the treatment of cancer in polymeric microcapsules (> 1000 nm) and have potential application in the case of deposit formation for slow and continuous local release such as in localized vaccines or chronic treatments.
  • the first FDA approved PLGA product was the
  • LupronDepot in 1996 (TAP Pharmaceutical Products Inc), 75:25 lactide / glycolide microparticles containing Leuprolide for the treatment of advanced prostate cancer. Administered locally, LupronDepot forms a depot and releases the drug gradually and can be administered at intervals of up to 4 months, replacing daily injections of free Leuprolide (KADA et al., 1994). PLGA microparticles are already being tested in humans as vaccine delivery systems. This clinical trial is underway in Brazil in patients with advanced neck and neck cancer, developed by SILVA, C. L and collaborators.
  • Poly (lactide) polymer or PLA
  • PLA is one of the most studied for the preparation of microparticles for biological application, due to its high bidegradability and biocompatibility, having already approved clinical use in the constitution of biodegradable suture threads, as well as PLGA
  • PLA nanospheres have also been successfully tested as carriers of various antileishmaniasis, such as primaquine, pentamidine, bacopasaponin and atovaquone, for the experimental treatment of intravenous visceral leishmaniasis.
  • the nitrogenous chaffs referred to herein are 3-nitro-2-hydroxy-4,6-dimethoxychalcona, which we will refer to as CH8 for ease.
  • the first object of this invention relates to pharmaceutical compositions containing a pharmaceutically effective amount of PLGA particle encapsulated antileishmanial drugs or drugs.
  • the second object of this invention relates to the use of pharmaceutical compositions containing a pharmaceutically effective amount of amphotericin B, chalcone nitrated (CH8) or antimony compounds, encapsulated in PLGA particles directed to the manufacture of a medicament for the subcutaneous treatment of inflammatory skin diseases.
  • infectious skin or skin regions such as leishmaniasis, mycoses, leprosy, psoriasis, tropical ulcer, warts, cavernous hemangioma in mammalian animals.
  • the third object of this invention is a pharmaceutical composition containing a therapeutically effective amount of amphotericin B, chalcone nitrite (CH8) or PLGA particle encapsulated antimonial compounds, and pharmaceutically acceptable excipients.
  • the last object of this invention is a method of treating parasitic diseases comprising subcutaneously applying a therapeutically effective amount of amphotericin B, chalcone nitrite (CH8) or antimonial compounds encapsulated in PLGA microparticles to a mammalian animal carrying leishmaniasis.
  • compositions comprising a pharmaceutically effective amount of PLGA particle encapsulated antileishmanial drugs or drugs, having as examples of drugs such as Amphotericin B, chalcone nitrate (CH8) and antimonial compounds.
  • the active agent may constitute about 5 to 85% of the weight of the formulation. Varied concentration of active compounds may enable faster initial release into tissue after local administration. In an exemplary variation as shown below, we use 10% Amphotericin B relative to the polymer (w / w).
  • medicaments or drugs used are amphotericin B, chalcone nitrate (CH8) and antimonial compounds such as meglumine antimoniate, sodium stibogluconate and potassium hydrate (IIIb) tartrate, combined or not.
  • glycolic and lactic acid copolymers are used and the percentage of each poly (lactic-co-glycolic acid) monomer may be about 15-85%, about 25-75%, about 35-65%. %, or the proportion of 50-50% may be employed.
  • the second object of this invention relates to the use of pharmaceutical compositions containing a pharmaceutically effective amount of amphotericin B or antimonial compounds, preferably Glucantime and PLGA particle-encapsulated nitrated chalcone (CH8) for the manufacture of a medicament for the subcutaneous treatment of inflammatory and infectious skin diseases or in skin regions (dermis, epidermis and appendages) such as leishmaniasis, mycosis, leprosy, psoriasis, tropical ulcer, warts, cavernous hemangioma in mammalian animals.
  • a pharmaceutically effective amount of amphotericin B or antimonial compounds preferably Glucantime and PLGA particle-encapsulated nitrated chalcone (CH8) for the manufacture of a medicament for the subcutaneous treatment of inflammatory and infectious skin diseases or in skin regions (dermis, epidermis and appendages) such as leishmaniasis, mycosis, leprosy, psoria
  • the third object of this invention is a pharmaceutical composition containing a therapeutically effective amount of amphotericin B or antimonial compounds such as PLGA particle encapsulated nitrated glucosin and chalcone (CH8) in addition to pharmaceutically acceptable excipients.
  • amphotericin B or antimonial compounds such as PLGA particle encapsulated nitrated glucosin and chalcone (CH8) in addition to pharmaceutically acceptable excipients.
  • the last object of this invention is a method of treating parasitic diseases comprising subcutaneously applying a therapeutically effective amount of Amphotericin B or antimonial compounds such as PLGA microparticle-encapsulated nitrided Glucantime and chalcone (CH8) to a mammalian animal carrying leishmaniasis.
  • Amphotericin B or antimonial compounds such as PLGA microparticle-encapsulated nitrided Glucantime and chalcone (CH8)
  • PLGA particles containing Amphotericin B and antimonial compounds will be produced by the multiple emulsification method (Ai / 0 / A 2 ), followed by the solvent evaporation method.
  • the polymer (PLGA) will be dissolved in dichloromethane (organic phase), to this organic phase will be added 1 ml pH 7.4 buffer containing the drugs (10% relative to the polymer (w / w)), and The mixture of the phases will be promoted under ultra-stirring at 7000rpm for 30 seconds to form a single emulsion (Al 10).
  • Such a single emulsion will be dispersed in the continuous phase (A 2 ) under 7000rpm ultrahomogenization for 60 seconds, a 3% aqueous polyvinyl acid (PVA) solution will be added to the system and solvent evaporation will be under stirring at 400 rpm for 4 hours.
  • PVA polyvinyl acid
  • the microparticle suspension will be centrifuged at 4000 rpm for 5 minutes, washed with distilled water 3 times and frozen in -20 ° C freezer for subsequent freeze drying for 16 hours.
  • the active agent may constitute about 5 to 85% of the weight of the formulation.
  • Determination of drug content in the particles is important to ensure the correct dose to be used in the trials.
  • Amphotericin B contents in PLGA particles will be determined by HPLC.
  • Antimony (Sb) levels will be determined by atomic absorption.
  • PLGA particles will have the drug release rate determined in physiological buffer pH 7.2 at 37 ° C. For this, 10 mg of particle will be incubated in 1 ml of PBS, plus 0.1% azide, under constant agitation at 80rpm and 37 ° C. Samples will be collected by centrifugation, taken from 500 ⁇ at times ranging from 1 hour to 270 days. Samples will be analyzed by UV-HPLC to determine the content of the released amphotericin B, the antimony content will be analyzed by atomic absorption. The curve will be constructed:% release x time.
  • Particle size and distribution analysis of b obtained by centrifugation and lyophilization will be performed using the distribution analyzer.
  • particle size Coulter LS230 Small amounts of particles will be dispersed in PA absolute ethanol with the aid of ultrasound until they reach the obscuration index required by the device.
  • the average particle size will be expressed as the average volume diameter (D4,3).
  • the particle diameter will be calculated at D 0 , 9, D 0 , ie D 0 , 5, which means the particle diameter corresponding to 90, 50 and 10% of the cumulative distribution and the polydispersion will be given by the span index, which will be calculated by (OD, 9 - OD, 1 / OD, 5).
  • the particles analyzed in C by SEM will also have their average size evaluated by the Coulter LS230 analyzer.
  • PLGA particles will be dispersed in water with pH corrected to 7.0 and zeta potential measurements will be determined on a Zetamaster apparatus (Malvern Instruments). Microparticle dispersion for potential zeta measurements will be prepared in duplicate and each sample will be measured 0 times.
  • Macrophages will be isolated from the peritoneal of BALB / c mice by adherence to 24-well culture plates infected with L amazonensis GFP promastigotes and treated with the formulations for 72h. After the incubation time, the cells will be scraped off and the parasitic load will be assessed by fluorimeter on a plate fluorimeter (485 nm excitation, 528 nm emission) (ROSSI-BERGMANN, et al., 1999) (FLx800, Bio-Tek Instruments, Inc) for L amazonensis or microscopic counting.
  • Macrophages will be isolated from the BALB / c mouse peritoneum by adherence to 24-well culture plates, empty or drug-containing PLGA particles will be added and after 72 hours the Crop supernatant will be evaluated for NO (Nitric Oxide) production by Griss methodology and for microbicidal activity evaluation and the presence of lactate dehydrogenase (LDH) enzyme for evaluation of necrosis toxicity by lactate dosage (Kit Dolles) .
  • NO Nitric Oxide
  • LDH lactate dehydrogenase
  • the ear swelling test (MEST) test It is recommended by the OECD and ANVISA and will be performed for skin immunotoxicity assessment (allergenicity),
  • MEST evaluates the increase in ear thickness as a sign of skin sensitization.
  • Animals will receive 3 topical applications of 100 ⁇ l of the formulations every 2 days (days 0, 2 and 4) in the dorsal region (induction phase). After 5 days, 10ul of the substances will be topically applied to one ear and to the other 10ul of the formulation will be applied intradermally (challenge phase). This is the time required for the body to produce the immune response. Ear swelling measurements will be evaluated at 6, 18, 36, 48, and 72 hours after challenge. Skin sensitization is positive when there is an increase in ear thickness above 10% compared to the control group.
  • Oxazolone (0.3%) is the most used substance as a positive control in these skin sensitization tests and, according to the literature, promotes a 30% increase in ear thickness in relation to the measurement before the challenge.
  • the animals will be treated intralesionally, blood will be collected and plasma obtained by centrifugation will be pretreated following the methodology proposed by (WANG & MORRIS, 2005; PAGANGA & RICE-EVANS, 1997 and-MANACH et al., 1996). lowering the pH to 4 using H 3 PO 4 ; acidifying with acetonitrile and centrifuging. Acidification of samples is important to break possible binding to plasma proteins. After this procedure plasma samples will be analyzed using UV-HPLC for amphotericin B and atomic absorption for antimonials.
  • Pharmacokinetic parameters (Cmax; ASC 0- -; t1 ⁇ 2; Vd; and Cl) will be obtained using software (ADAPT: Pharmacokinetic / p armacodynamic systems analysis) produced by the University of Southern California, available for free download.
  • ADAPT Pharmacokinetic / p armacodynamic systems analysis
  • the treated ears will be removed after different times and macerated in acetonitrile.
  • the suspensions will be clarified by centrifugation and processed as described for plasma.
  • Figure 1 shows the result of particle size analysis of PLGA particles containing Glucantime (meglumine antimoniate) or Amphotericin B, where Figure 1A represents the empty microparticles, 1 B the microparticles containing Glucantime and 1 C the microparticles containing Amphotericin B.
  • Figure 2 shows the analysis of PLGA particle morphology assessed by SEM and the images obtained by scanning electron microscopy (SEM) of empty PLGA particles (A), Glucantime (B), with Amphotericin B (C). Increases by 1500X, 3500X and 5000X, respectively.
  • the theoretical incorporation of amphotericin B into microparticles is 10% (w / w). Samples were made in triplicate.
  • Figure 4 shows the erosion analysis of amphotericin B-containing PLGA microparticles by scanning electron microscopy (SEM) images. Images A and B at time zero, images C and D with time equal to 30 days in PBS buffer and images E and F with time equal to 120 days in PBS buffer.
  • SEM scanning electron microscopy
  • Figure 5 shows the efficacy of amphotericin B encapsulation in single dose PLGA microparticles in the treatment of intralesional murine cutaneous leishmaniasis, where BALB / c mice were infected with 2x10 6 L.amazonensis GFP promastigotes in the ear. After 10 days of infection, the animals were randomly separated into groups of 4. The free amphotericin B treated group received intralesional injections twice a week with 50 ⁇ g doses of the drug for 4 weeks (total of 8 doses). The PLGA / Amphotericin B treated group received a single dose of the formulation intralesionally with 50 ⁇ g Amphotericin B dose (1 dose).
  • A The lesion growth (infected ear thickness minus thickness before infection) was accompanied during the experiment with the aid of caliper.
  • Figure 6 shows intralesional murine cutaneous leishmaniasis treatment, where BALB / c mice were infected with 2x0 6 L.amazonensis GFP promastigotes in the ear. After 7 days of infection, the animals were randomly separated into groups of 4.
  • treatment 1 the PLGA / amphotericin B treated group received a dose of the formulation intralesionally with a dose of 25 ⁇ g amphotericin B.
  • the free amphotericin B treated group and Liposomal amphotericin B received a dose of 1.25 ⁇ g of the drug.
  • Controls received a single administration of PLGA of 12.5 ⁇ g or 10ul of PBS.
  • the PLGA / amphotericin B treated group received a dose of the formulation intralesionally with 50 ⁇ g amphotericin B dose.
  • the free amphotericin B and liposomal amphotericin B treated group received a 50 ⁇ g dose of the drug.
  • Controls received a single administration of 500ug or 10ul of PBS PLGA.
  • the lesion growth was followed during the experiment with the aid of caliper.
  • Figure 08 shows the assessment of cardiac, hepatic and renal toxicity.
  • Animal blood was drawn 67 days after infection for dosages of: (a) serum TGO levels; (b) serum TGP levels; (c) serum creatinine serum levels.
  • Negative Control PBS.
  • Positive control ICC 4 .
  • Figure 9 shows the determination of the best drug ratio in the formulation.
  • PLGA microparticles containing Amphotericin B added in different proportions to the polymer (5%, 10% and 20% as indicated) were prepared. The particles were then washed and dried. To determine the best process yield, the dried particles were fully dissolved in Acetonitrile after stirring for 24 hours at 100 rpm. The samples were then centrifuged at 10,000 rpm for 5 minutes and the supernatant was removed for quantification of the HPLC incorporated drug content.
  • Table 1 The average particle size expressed as volume average diameter (D 4.3).
  • Table 2 The surface charge of the particles was evaluated by the Zeta Potential. Zeta SD Potential Specification
  • the particle morphology was also evaluated by SEM and the images are represented in Figure 2.
  • SEM morphological analysis of the In particles, we can observe the formation of clusters that may explain the observed heterogeneity in population size distribution when Glucantime or Amphotericin B are encapsulated.
  • EXAMPLE 2 EVALUATION OF EFFECTIVENESS OF IN VITRO PARTICULATION, RELEASE AND DEGRADATION
  • Amphotericin B uptake rate in PLGA microparticles can be seen in Figure 3A.
  • the Average incorporation rate was 89.2%, with this yield the particle amphotericin B content is 8.92% and not the 10% (w / w) Amphotericin B in theoretical PLGA microparticles .
  • EXAMPLE 3 EVALUATION OF THE EFFICACY OF COMPOSITIONS IN THE TREATMENT OF SKIN LEISHMANIASIS
  • amphotericin B-containing PLGA microparticles reduced the lesion growth at a single dose of 50 pg / kg as shown in Figure 5 (A), the Amphotericin-treated group. B free, received intralesional injections twice a week with doses of 50 ⁇ g of the drug for 4 weeks (total of 8 doses). With the evaluation of the parasitic load (B) we can conclude that amphotericin B / PLGA promoted a significant decrease in fluorescence compared to the placebo group, although the size of the lesion decreased there was no statistical difference between the group treated with empty microparticles. and the placebo group when we compared the parasitic load.
  • the group treated with the conventional liposome CH8 formulation was more effective than the group treated with a much higher dose of 200 ⁇ g of free cream lanete CH8.
  • the other groups that were treated with the empty vehicles presented similar profile to the untreated group, indicating specific activity of chalrada nitrada (CH8).

Landscapes

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

Abstract

L'invention concerne une composition pharmaceutique contenant de l'amphotéricine B, de la nitrochalcone (CH8) ou du glucantime, encapsulés dans des microparticules de polymères biodégradables à libération lente, un procédé d'encapsulation de l'agent pharmaceutique dans les particules, l'utilisation de ces compositions pharmaceutiques, un médicament et le traitement de parasitoses.
PCT/BR2012/000249 2011-07-18 2012-07-17 Compositions pharmaceutiques microparticulaires contenant des antiparasitaires pour traitement sous-cutané prolongé, utilisation desdites compositions pharmaceutiques pour la production d'un médicament, et méthode de traitement de parasitoses WO2013010238A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
BRPI1103229-4 2011-07-18
BRPI1103229-4A BRPI1103229B1 (pt) 2011-07-18 2011-07-18 Composições farmacêuticas microparticuladas contendo antiparasitários para terapia subcutânea prolongada, uso das ditas composições farmacêuticas para a produção de um medicamento e método de tratamento de parasitoses

Publications (1)

Publication Number Publication Date
WO2013010238A1 true WO2013010238A1 (fr) 2013-01-24

Family

ID=47557597

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/BR2012/000249 WO2013010238A1 (fr) 2011-07-18 2012-07-17 Compositions pharmaceutiques microparticulaires contenant des antiparasitaires pour traitement sous-cutané prolongé, utilisation desdites compositions pharmaceutiques pour la production d'un médicament, et méthode de traitement de parasitoses

Country Status (3)

Country Link
BR (1) BRPI1103229B1 (fr)
CO (1) CO6950469A2 (fr)
WO (1) WO2013010238A1 (fr)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015048371A1 (fr) * 2013-09-30 2015-04-02 Zoetis Llc Formulations de spiro-isoxazoline à action prolongée
WO2015177820A1 (fr) * 2014-05-19 2015-11-26 Università Degli Studi "Magna Graecia" Di Catanzaro Systèmes nanoparticulaires visant à véhiculer des médicaments pour le traitement de pathologies liées à une infection par leishmania

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008157614A2 (fr) * 2007-06-21 2008-12-24 Yale University Administration soutenue de médicament à partir de microparticules polymères biodégradables

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008157614A2 (fr) * 2007-06-21 2008-12-24 Yale University Administration soutenue de médicament à partir de microparticules polymères biodégradables

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
KAYSER, O. ET AL.: "Formulation and biopharmaceutical issues in the development of drug delivery systems for antiparasitic drugs", PARASITOL RESEARCH, vol. 90, 2003, pages S63 - S70 *
MEDDA, S ET AL.: "Phospholipid Microspheres: A novel delivery mode for targeting antileishmanial agent in experimental leishmaniasis", JOURNAL OF DRUG TARGETING, vol. 11, no. 2, 2003, pages 123 - 128 *
ORDÓNEZ-GUTIÉRRREZ, L. ET AL.: "In vitro effect of new formulations of amphoterecin B on amastigote and promastigote form of Leishmania infantum.", INTERNATIONAL JOURNAL OF ANTIMICROBIAL AGENTS, vol. 30, 2007, pages 325 - 261 *
ROY, P. ET AL.: "Andrographolide nanoparticles in leishmaniasis: Characterization and in vitro evaluations.", INTERNATIONAL JOURNAL OF NANOMEDICINE, vol. 5, 2010, pages 1113 - 1121 *

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015048371A1 (fr) * 2013-09-30 2015-04-02 Zoetis Llc Formulations de spiro-isoxazoline à action prolongée
CN105579029A (zh) * 2013-09-30 2016-05-11 硕腾服务有限责任公司 长效螺异恶唑啉制剂
AU2014324837B2 (en) * 2013-09-30 2019-01-17 Zoetis Services Llc Long-acting spiro-isoxazoline formulations
CN105579029B (zh) * 2013-09-30 2019-03-05 硕腾服务有限责任公司 长效螺异恶唑啉制剂
US10350196B2 (en) 2013-09-30 2019-07-16 Zoetis Services Llc Long-acting spiro-isoxazoline formulations
WO2015177820A1 (fr) * 2014-05-19 2015-11-26 Università Degli Studi "Magna Graecia" Di Catanzaro Systèmes nanoparticulaires visant à véhiculer des médicaments pour le traitement de pathologies liées à une infection par leishmania

Also Published As

Publication number Publication date
CO6950469A2 (es) 2014-05-20
BRPI1103229B1 (pt) 2020-10-06
BRPI1103229A2 (pt) 2013-12-24

Similar Documents

Publication Publication Date Title
JP6574228B2 (ja) 局所麻酔薬のデポー製剤及びその調製方法
ES2699692T3 (es) Sistema de depósito que comprende acetato de glatiramer
EP2741775B1 (fr) Nanoparticules polymériques pour des photosensibilisateurs
Matos et al. A review of current treatments strategies based on paromomycin for leishmaniasis
EP1313453B1 (fr) Vecteurs particulaires destines a ameliorer l'absorption orale de principes actifs
US20030118539A1 (en) Polyamines and analogs for protecting cells during cancer chemotherapy and radiotherapy
PT1143933E (pt) Novas formulações de cocleatos isolados em hidrogel, processo de preparação e sua utilização para a cedência de moléculas biologicamente relevantes.
BR112015012162B1 (pt) formulação de suspensão tópica e formulação de suspensão de minociclina tópica para usar no tratamento de acne vulgaris
EP1827401A2 (fr) Compositions et methodes destinees a traiter des affections de l'ongle
US10792244B2 (en) Parenteral sustained-release delivery of carvedilol disperse systems
US20220354801A1 (en) Multifunctional Nanoparticles For Prevention And Treatment Of Atherosclerosis
JP2009507006A (ja) 鉄キレート剤を含む医薬組成物
CN114949220B (zh) 一种靶向损伤节段背根神经节的pH响应镇痛药及其应用
de Santana et al. Nanoparticles for the treatment of visceral leishmaniasis
WO2013146386A1 (fr) Préparation d'anesthésique local à libération prolongée
Peng et al. Gout therapeutics and drug delivery
WO2013010238A1 (fr) Compositions pharmaceutiques microparticulaires contenant des antiparasitaires pour traitement sous-cutané prolongé, utilisation desdites compositions pharmaceutiques pour la production d'un médicament, et méthode de traitement de parasitoses
WO2022271951A1 (fr) Formulations à libération prolongée comprenant un modulateur sélectif du récepteur des androgènes
Jiang et al. Binary ethosomes-based transdermal patches assisted by metal microneedles significantly improve the bioavailability of carvedilol
BR112019017724A2 (pt) Sistemas depot de glatiramer para tratar formas progressivas de esclerose múltipla
Karimunnisa et al. Development and evaluation of a novel drug delivery system for albendazole
De Santana et al. Nanotechnology as an alternative to improve the treatment of cutaneous leishmaniasis: A systematic review of the literature
RU2548722C1 (ru) Противоопухолевое лекарственное средство пролонгированного действия на основе противоопухолевого препарата, ингибитора синтеза эстрогенов - анастрозола
Harwansh et al. Recent Updates on Transdermal Drug Delivery Approaches for the Management of Gout and its Clinical Perspective
KR20220125027A (ko) 부피바카인 리포좀 기반의 리포좀 약물전달체 제조방법 및 이에 의해 제조된 서방형 제제

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: 12814712

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 14032937

Country of ref document: CO

122 Ep: pct application non-entry in european phase

Ref document number: 12814712

Country of ref document: EP

Kind code of ref document: A1