amphiphilic antimony(v) complexes for oral treatment of ......amphiphilic antimony(v) complexes for...

8
Amphiphilic Antimony(V) Complexes for Oral Treatment of Visceral Leishmaniasis Flaviana R. Fernandes, a Weverson A. Ferreira, b Mariana A. Campos, a Guilherme S. Ramos, a Kelly C. Kato, a Gregório G. Almeida, c José D. Corrêa Junior, d Maria N. Melo, c Cynthia Demicheli, b Frédéric Frézard a Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, a Departamento de Química, Instituto de Ciências Exatas, b Departamento de Parasitologia, Instituto de Ciências Biológicas, c and Departamento de Morfologia, Instituto de Ciências Biológicas, d Universidade Federal de Minas Gerais, Pampulha, Belo Horizonte, Minas Gerais, Brazil The need for daily parenteral administration is an important limitation in the clinical use of pentavalent antimonial drugs against leishmaniasis. In this study, amphiphilic antimony(V) complexes were prepared from alkylmethylglucamides (L8 and L10, with carbon chain lengths of 8 and 10, respectively), and their potential for the oral treatment of visceral leishmaniasis (VL) was evaluated. Complexes of Sb and ligand at 1:3 (SbL8 and SbL10) were obtained from the reaction of antimony(V) with L8 and L10, as evidenced by elemental and electrospray ionization-tandem mass spectrometry (ESI-MS) analyses. Fluorescence probing of hydrophobic environment and negative-staining transmission electron microscopy showed that SbL8 forms kinetically stabi- lized nanoassemblies in water. Pharmacokinetic studies with mice in which the compound was administered by the oral route at 200 mg of Sb/kg of body weight indicated that the SbL8 complex promoted greater and more sustained Sb levels in serum and liver than the levels obtained for the conventional antimonial drug meglumine antimoniate (Glucantime [Glu]). The efficacy of SbL8 and SbL10 administered by the oral route was evaluated in BALB/c mice infected with Leishmania infantum after a daily dose of 200 mg of Sb/kg for 20 days. Both complexes promoted significant reduction in the liver and spleen parasite burdens in relation to those in the saline-treated control group. The extent of parasite suppression (>99.96%) was similar to that achieved after Glu given intraperitoneally at 80 mg of Sb/kg/day. As expected, there was no significant reduction in the parasitic load in the group treated orally with Glu at 200 mg of Sb/(kg day). In conclusion, amphiphilic antimony(V) complexes emerge as an in- novative and promising strategy for the oral treatment of VL. V isceral leishmaniasis (VL) is a systemic parasitic disease which leads to high rates of morbidity and mortality in humans worldwide. Even with scientific advances related to diagnosis, treatment, and prevention over the past 10 years, VL still is a neglected disease leading to 60,000 human deaths/year (1, 2). The clinical manifestations of VL are attributed to obligatory intracel- lular protozoa of the Leishmania donovani complex, and depend- ing on the etiological agent, the disease presents two distinct forms: anthroponotic VL that is endemic in India and Central Africa, caused by L. donovani, and zoonotic VL that occurs in countries of the Mediterranean basin, Central Asia, and the Amer- icas, caused by Leishmania infantum. Even though pentavalent antimonials, including meglumine antimoniate (Glucantime [Glu]) and sodium stibogluconate (Pentostam), are still the first-line drugs in several developing countries for treatment of all forms of leishmaniasis (2), they have several limitations (3, 4, 5). These drugs have to be given paren- terally, daily, for at least 3 weeks (typically, 20 mg of Sb/kg of body weight/day for 20 to 30 days). Antimony therapy is often accom- panied by local pain during intramuscular injections and by sys- temic side effects, requiring very careful medical supervision. Typ- ical side effects include nausea, vomiting, weakness and myalgia, abdominal colic, diarrhea, skin rashes, and hepatotoxicity, to- gether with the most severe pancreatitis and cardiotoxicity. All these factors contribute to compliance difficulties and, eventually, treatment failures. At present, only one oral VL therapy exists. Miltefosine (hexa- decylphosphocholine) was found to exhibit efficacy against VL. In 2002, miltefosine (Impavido; Zentaris GmBH) was registered and licensed in India for oral treatment of VL, but teratogenicity re- stricted its use in women of childbearing age (6, 7, 8). In addition, it has a long half-life (150 h) and requires 28 days of treatment; these factors, when the drug is taken orally on an outpatient basis, make the drug critically prone to low compliance and increase the possibility of the development of drug resistance (9, 10). In this context, new drugs must be developed that are capable of being administered orally with minimal medical supervision, at an affordable price. The oral bioavailability of meglumine antimoniate was found to be enhanced through complexation with cyclodextrin (11), presumably as a result of the formation of a ternary meglumine- Sb-cyclodextrin complex, which maintains the antimonial com- pound depolymerized and sustainedly releases the low-molecu- lar-weight 1:1 Sb-meglumine (12, 13). However, the advance achieved is still limited, because of the insufficient solubility of the ternary complex for application in large animals and the highly hydrophilic character of Sb-meglumine complex (14, 15). In the present study, a novel oral delivery strategy for pentava- lent antimonials was investigated, based on the formation of an amphiphilic antimony(V) complex. Such a complex was obtained Received 30 March 2013 Returned for modification 17 May 2013 Accepted 13 June 2013 Published ahead of print 24 June 2013 Address correspondence to Frédéric Frézard, [email protected]. F.R.F. and W.A.F. contributed equally to this article. Copyright © 2013, American Society for Microbiology. All Rights Reserved. doi:10.1128/AAC.00639-13 September 2013 Volume 57 Number 9 Antimicrobial Agents and Chemotherapy p. 4229 – 4236 aac.asm.org 4229 on August 15, 2017 by guest http://aac.asm.org/ Downloaded from

Upload: others

Post on 26-Jun-2020

6 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Amphiphilic Antimony(V) Complexes for Oral Treatment of ......Amphiphilic Antimony(V) Complexes for Oral Treatment of Visceral Leishmaniasis Flaviana R. Fernandes,a Weverson A. Ferreira,b

Amphiphilic Antimony(V) Complexes for Oral Treatment of VisceralLeishmaniasis

Flaviana R. Fernandes,a Weverson A. Ferreira,b Mariana A. Campos,a Guilherme S. Ramos,a Kelly C. Kato,a Gregório G. Almeida,c

José D. Corrêa Junior,d Maria N. Melo,c Cynthia Demicheli,b Frédéric Frézarda

Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas,a Departamento de Química, Instituto de Ciências Exatas,b Departamento de Parasitologia,Instituto de Ciências Biológicas,c and Departamento de Morfologia, Instituto de Ciências Biológicas,d Universidade Federal de Minas Gerais, Pampulha, Belo Horizonte,Minas Gerais, Brazil

The need for daily parenteral administration is an important limitation in the clinical use of pentavalent antimonial drugsagainst leishmaniasis. In this study, amphiphilic antimony(V) complexes were prepared from alkylmethylglucamides (L8 andL10, with carbon chain lengths of 8 and 10, respectively), and their potential for the oral treatment of visceral leishmaniasis (VL)was evaluated. Complexes of Sb and ligand at 1:3 (SbL8 and SbL10) were obtained from the reaction of antimony(V) with L8 andL10, as evidenced by elemental and electrospray ionization-tandem mass spectrometry (ESI-MS) analyses. Fluorescence probingof hydrophobic environment and negative-staining transmission electron microscopy showed that SbL8 forms kinetically stabi-lized nanoassemblies in water. Pharmacokinetic studies with mice in which the compound was administered by the oral route at200 mg of Sb/kg of body weight indicated that the SbL8 complex promoted greater and more sustained Sb levels in serum andliver than the levels obtained for the conventional antimonial drug meglumine antimoniate (Glucantime [Glu]). The efficacy ofSbL8 and SbL10 administered by the oral route was evaluated in BALB/c mice infected with Leishmania infantum after a dailydose of 200 mg of Sb/kg for 20 days. Both complexes promoted significant reduction in the liver and spleen parasite burdens inrelation to those in the saline-treated control group. The extent of parasite suppression (>99.96%) was similar to that achievedafter Glu given intraperitoneally at 80 mg of Sb/kg/day. As expected, there was no significant reduction in the parasitic load inthe group treated orally with Glu at 200 mg of Sb/(kg day). In conclusion, amphiphilic antimony(V) complexes emerge as an in-novative and promising strategy for the oral treatment of VL.

Visceral leishmaniasis (VL) is a systemic parasitic disease whichleads to high rates of morbidity and mortality in humans

worldwide. Even with scientific advances related to diagnosis,treatment, and prevention over the past 10 years, VL still is aneglected disease leading to 60,000 human deaths/year (1, 2). Theclinical manifestations of VL are attributed to obligatory intracel-lular protozoa of the Leishmania donovani complex, and depend-ing on the etiological agent, the disease presents two distinctforms: anthroponotic VL that is endemic in India and CentralAfrica, caused by L. donovani, and zoonotic VL that occurs incountries of the Mediterranean basin, Central Asia, and the Amer-icas, caused by Leishmania infantum.

Even though pentavalent antimonials, including meglumineantimoniate (Glucantime [Glu]) and sodium stibogluconate(Pentostam), are still the first-line drugs in several developingcountries for treatment of all forms of leishmaniasis (2), they haveseveral limitations (3, 4, 5). These drugs have to be given paren-terally, daily, for at least 3 weeks (typically, 20 mg of Sb/kg of bodyweight/day for 20 to 30 days). Antimony therapy is often accom-panied by local pain during intramuscular injections and by sys-temic side effects, requiring very careful medical supervision. Typ-ical side effects include nausea, vomiting, weakness and myalgia,abdominal colic, diarrhea, skin rashes, and hepatotoxicity, to-gether with the most severe pancreatitis and cardiotoxicity. Allthese factors contribute to compliance difficulties and, eventually,treatment failures.

At present, only one oral VL therapy exists. Miltefosine (hexa-decylphosphocholine) was found to exhibit efficacy against VL. In2002, miltefosine (Impavido; Zentaris GmBH) was registered andlicensed in India for oral treatment of VL, but teratogenicity re-

stricted its use in women of childbearing age (6, 7, 8). In addition,it has a long half-life (�150 h) and requires 28 days of treatment;these factors, when the drug is taken orally on an outpatient basis,make the drug critically prone to low compliance and increase thepossibility of the development of drug resistance (9, 10).

In this context, new drugs must be developed that are capableof being administered orally with minimal medical supervision, atan affordable price.

The oral bioavailability of meglumine antimoniate was foundto be enhanced through complexation with cyclodextrin (11),presumably as a result of the formation of a ternary meglumine-Sb-cyclodextrin complex, which maintains the antimonial com-pound depolymerized and sustainedly releases the low-molecu-lar-weight 1:1 Sb-meglumine (12, 13). However, the advanceachieved is still limited, because of the insufficient solubility of theternary complex for application in large animals and the highlyhydrophilic character of Sb-meglumine complex (14, 15).

In the present study, a novel oral delivery strategy for pentava-lent antimonials was investigated, based on the formation of anamphiphilic antimony(V) complex. Such a complex was obtained

Received 30 March 2013 Returned for modification 17 May 2013Accepted 13 June 2013

Published ahead of print 24 June 2013

Address correspondence to Frédéric Frézard, [email protected].

F.R.F. and W.A.F. contributed equally to this article.

Copyright © 2013, American Society for Microbiology. All Rights Reserved.

doi:10.1128/AAC.00639-13

September 2013 Volume 57 Number 9 Antimicrobial Agents and Chemotherapy p. 4229–4236 aac.asm.org 4229

on August 15, 2017 by guest

http://aac.asm.org/

Dow

nloaded from

Page 2: Amphiphilic Antimony(V) Complexes for Oral Treatment of ......Amphiphilic Antimony(V) Complexes for Oral Treatment of Visceral Leishmaniasis Flaviana R. Fernandes,a Weverson A. Ferreira,b

after reaction of antimony(V) with nonionic surfactants from theN-alkyl-N-methylglucamide series (Fig. 1). Improved oral bio-availability and pharmacokinetics of Sb in mice were achievedfrom such a complex, compared to meglumine antimoniate. Theresulting amphiphilic complexes were found to be active by theoral route in a murine model of VL.

MATERIALS AND METHODSMaterials and drugs. N-Decanoyl-N-methylglucamide (MEGA-10 orL10; 100% ultrapure grade) was purchased from Anresco (Solon, OH).N-Octanoyl-N-methylglucamide (MEGA-8 or L8; 98%), potassiumhexahydroxoantimonate [KSb(OH)6], and 1,6-diphenylhexatriene(DPH) were obtained from Sigma-Aldrich Chemical Co. (USA). Glucan-time (Glu) was obtained from Sanofi-Aventis (São Paulo, Brazil). Allother reagents were of at least reagent grade. Double-distilled deionizedwater was used throughout all the experiments.

Animals. Swiss and BALB/c mice (female, 4 to 6 weeks old) wereobtained from CEBIO of the Institute of Biological Sciences, Federal Uni-versity of Minas Gerais (UFMG; Belo Horizonte, Brazil). Free access to astandard diet was allowed, and tap water was supplied ad libitum. Thestudies involving animals were approved by the Ethical Committee forAnimal Experimentation of the UFMG with protocol numbers 215/09and 199/2011.

Parasites. Leishmania infantum (MHOM/BR/70/BH46) parasiteswere routinely maintained and isolated from golden (Syrian) hamsters(Mesocricetus auratus) and were grown as promastigotes at 26°C in Dul-becco modified Eagle medium (DMEM; Sigma) supplemented with 20%heat-inactivated fetal bovine serum (FBS; Cultilab, Brazil), 2 mM L-glu-tamine, 25 mM HEPES, 50 �M 2-mercaptoethanol, and 20 �g/ml ofgentamicin at pH 7.0.

Preparation and characterization of amphiphilic antimony(V)complexes. The SbL8 and SbL10 compounds were prepared by mixingKSb(OH)6 and L8 or L10 surfactant in water at a 1:3 Sb/surfactant molarratio and a final surfactant concentration of 0.08 M. The mixture washeated at 60°C under agitation until complete solvent evaporation. Theresulting film was redissolved in water at 25°C, and the dispersion wasfinally freeze-dried. The freeze-dried compounds were first characterizedby C, H, and N elemental analysis, inductively coupled plasma opticalemission spectrometry (Sb and K), and thermogravimetry. In the case ofthe SbL8 compound, analysis found the following: C, 47.28; H, 7.85; N,3.75; K, 3.6; Sb, 10.69%. The calculation for C45H87KN3O18Sb (1:3 Sb/L8complex) is as follows: C, 48.3; H, 7.84; N, 3.76; K, 3.5; Sb, 10.88%. In thecase of the SbL10 compound, analysis found the following: C, 49.48; H,8.19; N, 3.62; K, 3.3; Sb, 10.24%. The calculation for C51H99KN3O18Sb(1:3 Sb/L10 complex) is as follows: C, 50.91; H, 8.29; N, 3.49; K, 3.2; Sb,10.12%.

Electrospray ionization-tandem mass spectrometry (ESI-MS) spectrawere collected on a Thermo-Scientific LCQFleet mass spectrometer oper-ating in both the negative and positive modes. Samples were prepared in1:1 acidic water-methanol solutions, and measurements used the follow-ing main instrument parameters: capillary voltage, 3 kV, and cone voltage,30 kV.

The 1H and 13C nuclear magnetic resonance (NMR) spectra were ac-quired at 25°C on a Bruker DRX400-AVANCE spectrometer operating at400.129 MHz under standard conditions using a direct-detection 5-mm1H/13C dual probe with 90° pulse lengths of 7.2 �s for 1H. TMS

[3-(trimethylsilyl)propionic-2,2,3,3-d4 acid, sodium salt] was used as aninternal reference. Samples were prepared in CDCl3 or d6 dimethyl sul-foxide (DMSO) at 25 g/liter of L8. Two-dimensional 1H homonuclearcorrelation spectroscopy (COSY) and heteronuclear multiple quantumcoherence (HMQC) analyses allowed for the assignment of hydrogen andcarbon signals.

The formation of hydrophobic environment in aqueous dispersions ofL8 and SbL8 was investigated using the lipophilic fluorescent probe DPH,as described previously (16). This assay is based on the principle that DPHis not fluorescent in water due to molecular aggregation but fluorescesafter incorporation into a hydrophobic environment, such as those pro-vided by micelles, allowing for determination of the critical micellar con-centration (CMC). Fluorescence measurements were carried out using aCary Eclipse fluorescence spectrometer (Varian Inc.). Temperature wascontrolled at 25°C through a jacketed cuvette holder from a refrigeratedcirculating water bath. To evaluate the dependence of DPH fluorescenceupon L8 concentration, DPH was added from a tetrahydrofuran stocksolution at a final concentration of 5 � 10�7 M to dispersions of L8 orSbL8 in water at various concentrations. After a 24-h incubation at 25°Cunder light protection, the fluorescence intensity was measured at excita-tion and emission wavelengths of 360 and 428 nm, respectively. To eval-uate the kinetic stability of SbL8 and L8 nanoassemblies, dispersions ofSbL8 and L8 were prepared in water at 50 mM and 100 mM, respectively,and were diluted to 7.5 mM. After different times of incubation at 25°C,the interaction of DPH was assessed through measurement of fluores-cence intensity.

Dynamic light scattering (DLS) measurements of L8 and SbL8 disper-sions were performed using a Zetasizer (Nano ZS90; Malvern Instru-ments, United Kingdom). Dispersion Technology Software, version 6.12,was used to collect and analyze the data. The samples were prepared inwater at an L8 concentration of 30 mM, kept at 25°C during the entireexperiment, and analyzed at a fixed angle of 90°. Determined parameterswere the mean hydrodynamic diameter, polydispersity index, and particlecount.

Dispersions of SbL8 and L8 at 30 mM in water were analyzed at 25°Cfor their size distribution and nanoparticle concentration, using a Nano-Sight LM14C (United Kingdom). These parameters were determined ac-cording to the Nanoparticle Tracking Analysis (NTA) technique and us-ing NTA 2.3 software (17). L8 and SbL8 were analyzed using the samecamera gain. This technique is based on the tracking of Brownian motionof individual particles and determination of size according to the Stokes-Einstein equation.

Negative-staining transmission electron microscopy (TEM) analysiswas carried out at the Microscopy Center of UFMG using a TEM operat-ing at an acceleration voltage of 120 KV, line resolution of 0.34 nm, pointresolution of 0.49 nm, and Cs of 6.3 (spherical aberration), implying highcontrast (model Tecnai G2 Spirit, 2006; FEI). For sample preparation, adrop of SbL8 aqueous dispersion (0.001 M Sb) was placed onto a 300-mesh Formvar copper grid (TABB Laboratories Equipment, Berks,United Kingdom) and allowed to adsorb, and the surplus was removed byfilter paper. A drop of 5% (wt/vol) aqueous solution of uranyl acetate wasadded. The surplus water was removed by filter paper, and the sample wasdried at room temperature before the nanostructures were imaged. Rep-resentative electron micrographs were taken at magnifications of �22.000to �30.000.

Pharmacokinetic studies of antimonial compounds in mice. Swissmice received through oral inoculum, with the aid of a needle gauge, 0.2ml of an aqueous solution of either commercial antimonial drug (Glu) orSbL8 complex at 200 mg of Sb/kg of body weight. Mice from each group(n � 5/time) were sacrificed at the following times: 15 and 30 min and 1,2, 3, 6, 12, and 24 h after administration. The pharmacokinetics of Glu wasalso evaluated after administration to mice by the intravenous (i.v.) routeat 300 mg of Sb/kg. Animals (n � 5 to 7/time) received by the tail vein 0.1ml of Glu and were sacrificed at the following time points: 10, 20, 40, 60,and 90 min and 2 and 3 h. Animal sacrifice was done by cervical disloca-

FIG 1 Structure of octanoyl N-methyl-D-glucamide (L8).

Fernandes et al.

4230 aac.asm.org Antimicrobial Agents and Chemotherapy

on August 15, 2017 by guest

http://aac.asm.org/

Dow

nloaded from

Page 3: Amphiphilic Antimony(V) Complexes for Oral Treatment of ......Amphiphilic Antimony(V) Complexes for Oral Treatment of Visceral Leishmaniasis Flaviana R. Fernandes,a Weverson A. Ferreira,b

tion after ketamine-xylazine anesthesia. Blood samples were then col-lected from the brachial plexus, and the serum was recovered and frozen at�20°C. The livers were also recovered, homogenized, and subjected todigestion with nitric acid in a dry block (MA 4004; Marconi, São Paulo,Brazil), as described previously (18). Antimony was determined in dilutedserum and digested liver by electrothermal atomic absorption spectrom-etry (ETAAS) using a Perkin-Elmer AA600 graphite furnace atomic ab-sorption spectrometer, as described previously (15, 18). The analyticalmethods for determination of Sb in the serum and liver were validated andshowed suitable levels of precision (coefficient of variation [CV] � 5%),accuracy (80 to 120% analyte recovery), and linearity (range of 10 to 180�g of Sb/liter). The quantification limits of the analytical methods were240 �g of Sb/liter and 0.93 �g of Sb/g for the serum and liver, respectively.Pharmacokinetic parameters were determined using the R-STRIP 4.03computer program. Experimental Sb concentration-time data were sub-jected to iterative weighted nonlinear least-squares regression (19). A bi-exponential model with oral bolus input was chosen. Fitted parametersincluded maximum concentration of Sb (Cmax), the time for reaching thepeak Sb concentration (Tmax), the mean residence time of Sb in the 0- to24-h interval (MRT), the area under the concentration-time curve in the0- to 24-h interval (AUC), and the coefficient of partition of Sb to the liver(Kp � AUC in liver/AUC in serum).

Antileishmanial activity in murine model of visceral leishmaniasis.Groups of BALB/c mice (n � 7 to 10) were infected in the tail vein with1 � 106 late-log-phase L. infantum promastigotes. The treatment wasstarted 8 days after infection, with daily doses for 20 days. The first exper-iment included 5 groups that received the following formulations: SbL8by the oral route (46 mg in 200 �l of water, equivalent to 200 mg of Sb/kg,for each daily dose), L8 alone by the oral route (same dose as in the grouptreated with SbL8), Glu by the oral route (58 �l of Glu plus 142 �l ofsaline, equivalent to 200 mg of Sb/kg, for each daily dose), Glu by theintraperitoneal (i.p.) route (24 �l of Glu plus 77 �l of saline, equivalent to80 mg of Sb/kg, for each daily dose), and saline by the oral route (200 �l ofNaCl [0.15 M]). The second experiment included 3 groups that receivedthe following formulations: SbL10 by the oral route (48 mg in 200 �l ofwater, equivalent to 200 mg of Sb/kg, for each daily dose), Glu by the

intraperitoneal route (24 �l of Glu plus 77 �l of saline, equivalent to 80 mgof Sb/kg, for each daily dose), and saline by the oral route.

Twenty-one days after the beginning of treatment, the animals weresacrificed and the spleen and liver were harvested for determination ofparasite burdens by limiting dilution, as previously described (20). Briefly,organs were weighed and fragmented, and a tissue homogenate was ob-tained in 1 ml of DMEM plus 20% FBS at pH 7.0. Each tissue homogenatewas serially diluted (10-fold) in a 96-well flat-bottom microtiter plates(Nunc; Nunclon). Samples, in duplicate, were incubated at 23°C. Thewells containing motile promastigotes were identified with a microscope(Axiovert 25; Zeiss), and the parasite burden was determined from thehighest dilution at which promastigotes had grown after 12 days of incu-bation, as follows: parasite burden � 10highest dilution. Data are presented aslog parasite burden/mg of organ. The resulting transformed data (nor-mally distributed) were statistically analyzed using one-way analysis ofvariance (ANOVA) followed by Bonferroni posttest.

A parasite suppression index (SI) was also calculated by the followingformula: SI � 100 � [100 � (median of parasite burden/mg of organ oftreated mice)/(median of parasite burden/mg of organ of control mice)].In the former experiment, a fragment of the liver was also used for deter-mination of Sb, as described above.

RESULTSPhysicochemical characterization of amphiphilic antimonycomplexes. Complexation of Sb(V) with the surfactants L8 (Fig.1) and L10 was achieved through reaction of KSb(OH)6 with thesurfactant in water at a 1:3 molar ratio. According to elemental,thermogravimetry, and conductimetry analyses as well as K andSb determinations, the resulting product was characterized as apotassium salt containing Sb and the surfactant at a 1:3 molarratio. Interestingly, the resulting products dissolved at high con-centration in nonpolar solvents, such as chloroform and DMSO,and also in water.

Figure 2 presents the ESI-MS spectrum of SbL8 in the negativemode, and Table 1 displays the main species identified in SbL8 and

FIG 2 ESI-MS spectrum of SbL8 in the negative mode.

Amphiphilic Antimony(V) Complexes

September 2013 Volume 57 Number 9 aac.asm.org 4231

on August 15, 2017 by guest

http://aac.asm.org/

Dow

nloaded from

Page 4: Amphiphilic Antimony(V) Complexes for Oral Treatment of ......Amphiphilic Antimony(V) Complexes for Oral Treatment of Visceral Leishmaniasis Flaviana R. Fernandes,a Weverson A. Ferreira,b

SbL10 and their proposed structures. Accordingly, the main peakwas attributed to a 1:3 ratio of Sb to ligand complex. ESI-MSanalysis of the 1,078.5 species identified in SbL8 led to the appear-ance of m/z 757.5 ions corresponding to 1:2 complexes of Sb to L8,indicating the release of one L8 molecule.

To get insight into the site of Sb(V) binding to the surfactantmolecule, SbL8 was further characterized using 1H and 13C NMRin CDCl3. No significant change of the chemical shift of L8 hydro-gens was observed upon complexation (data not shown). On theother hand, as illustrated in Table 2, comparative 13C NMR anal-yses of L8 and its complex showed changes of the resonance of C1,C1�, C4, and C5, supporting the complexation of Sb(V) with thepolar head of the surfactant. According to 1H NMR spectra ob-tained in d6 DMSO, integration of the proton signals related tohydroxyls (5- to 4-ppm range) gave values of 4.9 and 2.5 for L8 andits complex, respectively. These data strongly indicate that Sb(V)coordinates between 2 and 3 hydroxyls in L8.

To address the changes in the physicochemical behavior of L8upon complexation with Sb(V), the nanoassemblies formed inaqueous solution were characterized through DPH fluorescenceprobing of hydrophobic environment, as well as size and concen-tration analyses of nanoparticles by DLS, TEM, and NTA.

As shown in Fig. 3A, the lipophilic fluorescent probe DPHexhibited an increase of the fluorescence intensity when incubatedwith L8 dispersion in water, from a concentration of about 50mM. This property was expected from the reported value of crit-ical micellar concentration (CMC) of L8 (21). Interestingly, theSbL8 complex formed a hydrophobic environment from a lowerL8 concentration of about 10 mM, indicating an enhanced ther-modynamic stability of the nanoassemblies.

The dispersions of SbL8 in water were found to be thermody-namically stable for at least 1 month upon storage at room tem-

perature. This long-term stability was evidenced through macro-scopic evaluation and DPH incorporation (data not shown). Onthe other hand, upon dilution of a concentrated SbL8 solution(from 50 mM to 7.5 mM in water), a slow decrease of DPH incor-poration was observed (Fig. 3B), indicating that SbL8 nanoassem-blies dissociate slowly after dilution. Indeed, micelle nanoassem-blies are expected to dissociate upon dilution below the CMC;however, surfactant micelles usually show immediate dissocia-tion, as observed in this study in the case of noncomplexed L8micelles (Fig. 3B). Thus, SbL8 nanoassemblies exhibited unusualkinetic stabilization upon dilution, supporting the model thatthese nanoparticles do not readily dissociate following adminis-tration and may act as a sustained release system of Sb.

The size distribution of nanoparticles in L8 and SbL8 aqueousdispersions could not be fully assessed by DLS because of highpolydispersity (L8 and SbL8) and low count (L8). DLS analysis ofSbL8 suggests the coexistence of micelle-type particles with adiameter of a few nanometers, together with much larger nano-assemblies with a mean diameter in the 100-nm range (data notshown). This interpretation was supported by transmissionelectron microscopy analysis of SbL8 dispersion in water afternegative staining. As illustrated in Fig. 4, two main populationsof nanoparticles were identified: one population with meandiameter in the 20- to 30-nm range and the other one showinga mean diameter of 80 nm. NTA, with expected sensitivity fornanoparticles with diameter higher than 30 nm (17), revealedthe presence of nanoparticles with mean hydrodynamic diameter inthe range of 115 to 140 nm in both L8 and SbL8 (Table 3). Inter-

TABLE 1 ESI-MS characterization of SbL8 and SbL10 complexes

Anionic species

m/z

SbL8 SbL10

[(2L-6H)SbV]� 757.3 813.8[(2L-4H)SbV(O)]� 775.3 831.9[(2L-4H)SbV(OH)2]� 793.3 845.8[(2L-6H)SbV(O)2]� 877.3[(5L-12H)(SbV)2]2� 917.9 988.0[(3L-6H)SbV]� 1,078.5a 1,162.6a

[(5L-12H�K)(SbV)2]� 1,874.8a Peak of higher intensity.

TABLE 2 13C NMR data for L8 and SbL8 in CDCl3

Carbon(s)

Chemical shift (, ppm)

L8 SbL8

2= 33.08/33.66 33.14/33.69 �0.06/�0.033= 25.05/25.66 25.08/25.61 �0.03/0.054=-7= 22.65/29.12/29.44/

31.7922.66/29.14/29.49/

31.82�0.01/�0.02/�0.05/

�0.038= 14.09 14.09 0.011� 34.57/37.55 34.38/37.28 0.19/0.272 72.05 72.05 01 51.74/52.82 51.48/52.65 0.26/0.173 70.21 70.29 0.084 and 5 71.85 or 73.13 71.67 or 73.23 0.18 or �0.16 63.89 63.84 0.05

FIG 3 Interaction of DPH fluorescent probe with L8 and SbL8 aqueous dis-persions at 25°C. (A) Fluorescence intensity of 0.5 �M DPH after 24 h ofincubation with L8 or SbL8 dispersions, as a function of L8 concentration. (B)Fluorescence intensity of 0.5 �M DPH in SbL8 or L8 dispersion at differenttimes after dilution of concentrated aqueous solution (50 mM for SbL8 and100 mM for L8) to 7.5 mM L8. a.u., arbitrary units.

Fernandes et al.

4232 aac.asm.org Antimicrobial Agents and Chemotherapy

on August 15, 2017 by guest

http://aac.asm.org/

Dow

nloaded from

Page 5: Amphiphilic Antimony(V) Complexes for Oral Treatment of ......Amphiphilic Antimony(V) Complexes for Oral Treatment of Visceral Leishmaniasis Flaviana R. Fernandes,a Weverson A. Ferreira,b

estingly, the concentration of nanoparticles was much higherin SbL8 than L8 dispersion, in accordance with the higher CMCof L8.

Pharmacokinetic studies of SbL8 in mice by the oral route.SbL8 given by the oral route was evaluated in mice and comparedto the hydrophilic commercial antimonial drug Glu, regardingpharmacokinetic profiles of Sb in the serum and liver. Figure 5shows the level of Sb in the serum and liver in Swiss mice afteradministration of SbL8 and Glu at the same dose of Sb (200 mg ofSb/kg). Table 4 displays the serum pharmacokinetic parameters.Both compounds were found to be rapidly absorbed; however,SbL8 promoted more sustained serum Sb levels compared to se-rum Glu levels, with significantly greater serum Sb concentrationsafter 6 and 24 h (Fig. 5A). Accordingly, the area under the curve(AUC) and mean residence time (MRT) were about 4-fold greaterfor SbL8 than for Glu (Table 4). It noteworthy that Sb reached theliver more rapidly from SbL8 than Glu and that SbL8 promoted

significantly higher Sb levels in the 0.5- to 3-h period (Fig. 5B),resulting in a 2.4-fold-greater AUC in this tissue (Table 4). Thesedata suggest that part of SbL8 is retained in the liver after absorp-tion into the portal system before being released into the blood-stream. This is in contrast with Glu, whose pharmacokinetic datasuggest an accumulation of Sb in the liver after entrance in theblood circulation. The lower MRT of Glu is also consistent withthe fast renal excretion of this water-soluble drug (4).

The pharmacokinetic parameters of SbL8 and Glu by the oralroute were also compared to those of Glu given intravenously at300 mg of Sb/kg (Table 4). Even though oral antimonial com-pounds exhibited much lower values for serum Cmax and AUCthan does intravenous Glu, these oral drugs showed much longerMRTs. Interestingly, the AUC determined in the liver for oralSbL8 was close to that for intravenous Glu. Furthermore, the co-efficient of partition of the drug to the liver (Kp � AUC liver/AUCserum) was more favorable for the oral drugs than for intravenousGlu (Table 4). These results suggest that a higher targeting of Sb tothe liver was achieved when the drugs were given by the oral route.

Efficacies of SbL8 and SbL10 in a murine model of visceralleishmaniasis. The observation that the amphiphilic antimonialdrug promoted greater concentrations of Sb in the liver comparedto Glu led us to further evaluate the antileishmanial efficacies ofSbL8 and SbL10 in BALB/c mice infected with Leishmania infan-tum, as an experimental model of VL. In the first experiment, theefficacy of oral SbL8 at 200 mg of Sb/kg/day was compared to thatof Glu given by the oral and i.p. routes at 200 and 80 mg of Sb/kg/day, respectively. Control groups received saline and L8 orally.However, treatment of mice with L8, given at the same dose as inSbL8 group, had to be interrupted because of acute toxicity, with50% animal loss after 10 days of treatment. The fact that no animaldied during the treatment with SbL8 strongly suggests that com-plexation of L8 with Sb(V) reduced its acute toxicity. Figure 6shows the parasite burdens in the livers and spleens of mice after20 days of treatment. Oral SbL8 promoted a significant reductionin the liver and spleen parasite burdens compared to those in thecontrol group (saline). These data represent 99.999% and99.987% parasite suppression in the liver and spleen, respectively.In contrast, Glu given orally did not significantly reduce the liver

FIG 4 Negative-staining transmission electron microscopy analysis of SbL8dispersion in water. (Bottom) Representative TEM image of nanoparticles inSbL8 dispersion. (Top) Histogram showing the particle size distribution witha 10-nm range.

TABLE 3 NTA analyses of L8 and SbL8 at 30 mM in water

CompoundDiam, mean diam �SDa (nm)

Particle concn(108/ml)

L8 138 � 61 2.9SbL8 114 � 47 11.7a Data are the means and standard deviations of 2,297 and 455 tracks for SbL8 and L8,respectively.

FIG 5 Pharmacokinetics of Sb in the serum (A) and liver (B) in Swiss miceafter administration of SbL8 and Glu at 200 mg of Sb/kg by the oral route. Dataare shown as means � SEMs (n � 5 to 7). *, P � 0.05 (Mann-Whitney test).

Amphiphilic Antimony(V) Complexes

September 2013 Volume 57 Number 9 aac.asm.org 4233

on August 15, 2017 by guest

http://aac.asm.org/

Dow

nloaded from

Page 6: Amphiphilic Antimony(V) Complexes for Oral Treatment of ......Amphiphilic Antimony(V) Complexes for Oral Treatment of Visceral Leishmaniasis Flaviana R. Fernandes,a Weverson A. Ferreira,b

or spleen parasite load. Furthermore, oral Glu was significantlyless effective than oral SbL8 in reducing the liver parasite load. Thegroup treated intraperitoneally with Glu, which served as a posi-tive control, showed a significant parasite suppression in both theliver and spleen. The concentration of Sb in the liver was alsodetermined just after treatment, as an attempt to correlate theantileishmanial activity to the liver Sb concentration. As shown inFig. 7, the Sb concentration in the liver was significantly greaterafter oral SbL8 and intraperitoneal Glu than after oral Glu. Inter-estingly, the concentration of Sb did not differ significantly be-tween the groups that received oral SbL8 and intraperitoneal Glu.

In a second experiment, the efficacy of SbL10 given orally at200 mg of Sb/kg/day was evaluated in the same experimentalmodel and was compared to that of Glu given intraperitoneally at80 mg of Sb/kg/day. Figure 8 shows the parasite burdens in thelivers and spleens of mice after 20 days of treatment. Importantly,oral SbL10 promoted a significant reduction in both the liver andspleen parasite burdens, compared to the values for the saline

group, showing suppression levels (99.984% in liver and 99.969%in spleen) close to those achieved after intraperitoneal Glu(99.963% in liver and 99.995% in spleen).

These data, taken together, established for the first time the oralefficacy of amphiphilic antimony(V) complexes in a murinemodel of VL.

DISCUSSION

The present work reports for the first time the preparation ofamphiphilic Sb(V) compounds from two nonionic surfactants ofthe N-alkyl-N-methylglucamide series (L8 and L10). These com-pounds consist essentially of 1:3 complexes of Sb to ligand. Theamphiphilic character of the resulting complexes is evidenced bytheir abilities to dissolve in both organic solvent and water and toself-associate in aqueous solution, forming nanoassemblies. Asmajor results of pharmacological evaluations, SbL8 exhibited animproved oral bioavailability of Sb and more favorable pharma-cokinetic parameters in the serum and liver of mice than obtainedwith Glu. Importantly, the resulting amphiphilic complexesshowed efficacy by the oral route in a murine model of VL.

Conventional pentavalent antimonials are considered inactivewhen given by the oral route. A previous pharmacokinetic study ofmeglumine antimoniate with dogs indicated an oral bioavailabil-ity of about 10% (15). Such a low bioavailability can be attributedto the highly hydrophilic character of meglumine antimoniate andalso to its tendency to polymerize in concentrated aqueous solu-tions (22). As an attempt to improve the oral bioavailability ofpentavalent antimony, a strategy based on the formation of am-phiphilic antimony(V) complex was investigated. This strategy issupported by previous works showing improved delivery of metalacross biological membranes through their complexation with li-

TABLE 4 PK parameters of SbL8 and Glu in Swiss mice

Drug (route)Dose(mg/kg)

Serum PK parameters Liver PK parameters

Cmax

(mg/liter) Tmax (h)AUC(mg · h/liter) MRT (h)

AUC(�g · h/g) MRT (h)

Kp (AUC liver/AUC serum)

SbL8, oral 200 2.2 0.8 39.6 10.8 131.8 9.3 3.3Glu, oral 200 2.9 1.3 10.3 2.6 55.0 11.1 5.3Glu, i.v. 300 1,652 0 474 0.29 189.9 0.74 0.4

FIG 6 Parasite load in the liver (A) and spleen (B) in BALB/c mice infectedwith Leishmania infantum, after 20 days of treatment with oral SbL8 (200 mg ofSb/kg/day), oral Glu (200 mg of Sb/kg/day), intraperitoneal Glu (80 mg ofSb/kg/day), or saline. Data are shown as means � SEMs (n � 7). **, P � 0.01;***, P � 0.001 (one-way ANOVA followed by Bonferroni posttest).

FIG 7 Sb concentration in the liver in BALB/c mice infected with Leishmaniainfantum, after 20 days of treatment with oral SbL8 (200 mg of Sb/kg/day), oralGlu (200 mg of Sb/kg/day), or intraperitoneal Glu (80 mg of Sb/kg/day). Dataare shown as means � SEMs (n � 7). *, P � 0.05; **, P � 0.01 (Kruskal-Wallisfollowed by Dunn’s multiple-comparison posttest).

Fernandes et al.

4234 aac.asm.org Antimicrobial Agents and Chemotherapy

on August 15, 2017 by guest

http://aac.asm.org/

Dow

nloaded from

Page 7: Amphiphilic Antimony(V) Complexes for Oral Treatment of ......Amphiphilic Antimony(V) Complexes for Oral Treatment of Visceral Leishmaniasis Flaviana R. Fernandes,a Weverson A. Ferreira,b

pophilic ligand, as, for example, in Tc-99m HMPAO (99mTc-labeled hexamethylpropyleneamine oxime) (23) and satraplatin(24). N-Alkyl-N-methylglucamides were chosen as ligands for thispurpose, because their polar head moiety is N-methylglucamine,which is the ligand of Sb(V) in meglumine antimoniate. Thus,these new complexes may be seen as amphiphilic meglumine an-timoniate derivatives.

Regarding the pharmacokinetic data (Table 4), SbL8 showed ahigher AUC than oral Glu, which may be explained by its amphi-philic character and its more effective permeation across the gas-trointestinal tract. The aggregated state of SbL8 and the low rate ofdissociation of these nanoassemblies probably contribute to themore sustained serum Sb levels, also evidenced by the greaterMRT. Another contributing factor may be the different mecha-nisms and rates of Sb elimination after absorption of SbL8 andGlu. It is also noteworthy that SbL8 promoted higher levels of Sbin the liver in mice, presumably because of its improved retentionin the liver during the first-pass removal and the longer half-life ofthe amphiphilic drug.

To get insight into the mode of action of SbL8, in vitro studieswere also carried out (data not shown). Cytotoxicity evaluation ofSbL8 against peritoneal macrophages indicated that it is about6-fold more cytotoxic than L8 (concentration of L8 that decreasedcell viability by 50% [CC50], 0.78 versus 4.9 mM). This is in con-trast with the in vivo acute toxicity that was observed for L8, butnot for SbL8. When evaluated for antileishmanial activity in theLeishmania amazonensis-infected macrophage model, neither L8

nor SbL8 showed significant activity up to 0.3 mM for SbL8 and to1 mM for L8 (data not shown). In the same model, Glu showedantileishmanial activity, but at a rather high concentration of Sb(50% inhibitory concentration [IC50] � 4.6 mM). Thus, the pres-ent data suggest that SbL8 suffers a transformation in vivo thatreduces its cytotoxicity. Furthermore, SbL8 probably exerts itsantileishmanial action after activation, through a process that maybe more effective in vivo than in vitro. This activation step mayconsist in the reduction of Sb(V) into Sb(III), in accordance withthe generally accepted model for the mode of action of pentavalentantimonials (4). The fact that treatment with oral SbL8 and thatwith i.p. Glu promoted about the same hepatic levels of Sb andalso reduced the liver parasite burden to similar extents furthersupports the model that SbL8 action is mediated by the metal,similarly to Glu. Nevertheless, oral SbL8 and i.p. Glu cannot beconsidered equally effective, since SbL8 was given at a dose thatyielded 2.5-fold more Sb than Glu. Furthermore, when SbL8 wasgiven orally at 80 mg of Sb/kg/day for 20 days, no significant re-duction of parasite load could be detected in the same murinemodel of VL (data not shown).

Compared to our previous cyclodextrin-based oral formula-tion of meglumine antimoniate (11, 15), the most significant ad-vantages of the present amphiphilic formulation are its muchgreater solubility in water (SbL8 aqueous dispersions were pre-pared at concentrations as high as 0.7 M Sb) and its superiorbioavailability and sustained drug release property. Indeed, whenevaluated in dogs, cyclodextrin-based oral formulation showedlimited increases of bioavailability (15 versus 10%) and MRT (6.8versus 4.1 h) compared to those of noncomplexed meglumineantimoniate (15).

The present work indicates that the formation of nanoassem-blies not only allows for the application of very high doses ofamphiphilic Sb with reduced acute toxicity but also contributes tothe sustained action of the metal. Regarding the structure of thesenanoassemblies, TEM and DLS analyses support the formation ofmicelles in both L8 and SbL8; however, NTA evidenced the for-mation of large nanostructures to a greater extent in SbL8. Nev-ertheless, the exact structure of SbL8 nanoaggregates requires fur-ther investigation. The kinetic stability of SbL8 nanoassembliesindicates that they do not readily dissociate following dilution inbiological fluids. This property may be attributed to the low rate ofdissociation of the Sb(V)OO bond (22, 25). This property may beexplored in the future through the use of these nanostructures ascarrier systems of other lipophilic drugs. Indeed, polymeric mi-celles or micelles formed from polymeric surfactants are beingintensively studied as drug carrier systems, taking advantage oftheir kinetic stability (26, 27).

From the point of view of drug development, the present workrepresents promising preliminary evidence that the amphiphilicSb(V) complex is a feasible strategy to achieve oral efficacy ofpentavalent antimonials against VL. Further steps include selec-tion of the most effective and least toxic surfactant, as well asdetailed toxicological evaluations.

ACKNOWLEDGMENTS

We acknowledge the Brazilian agencies CNPq, FAPEMIG, and CAPES forfinancial support. F.F., C.D., and M.N.M. are recipients of a researchfellowship from CNPq.

FIG 8 Parasite load in the liver (A) and spleen (B) in BALB/c mice infectedwith Leishmania infantum after 20 days of treatment with oral SbL10 (200 mgof Sb/kg/day), intraperitoneal Glu (80 mg of Sb/kg/day), or saline. Data areshown as means � SEMs (n � 7). ***, P � 0.001 (one-way ANOVA followedby Bonferroni posttest).

Amphiphilic Antimony(V) Complexes

September 2013 Volume 57 Number 9 aac.asm.org 4235

on August 15, 2017 by guest

http://aac.asm.org/

Dow

nloaded from

Page 8: Amphiphilic Antimony(V) Complexes for Oral Treatment of ......Amphiphilic Antimony(V) Complexes for Oral Treatment of Visceral Leishmaniasis Flaviana R. Fernandes,a Weverson A. Ferreira,b

REFERENCES1. Guerin PJ, Olliaro P, Sundar S, Boelaert M, Croft SL, Desjeux P,

Wasunna MK, Bryceson AD. 2002. Visceral leishmaniasis: current statusof control, diagnosis, and treatment, and a proposed research and devel-opment agenda. Lancet Infect. Dis. 2:494 –501.

2. World Health Organization. 2010. Control of the leishmaniases. Reportof a meeting of the WHO Expert Committee on the Control of Leishman-iases, Geneva, 22 to 26 March 2010. WHO Technical report series 949.World Health Organization, Geneva, Switzerland.

3. Berman JD. 1997. Human leishmaniasis: clinical, diagnostic, and chemo-therapeutic developments in the last 10 years. Clin. Infect. Dis. 24:684 –703.

4. Frézard F, Demicheli C, Ribeiro RR. 2009. Pentavalent antimonials: newperspectives for old drugs. Molecules 14:2317–2336.

5. Marsden PD. 1985. Pentavalent antimonials: old drugs for new diseases.Rev. Soc. Bras. Med. Trop. 18:187–198.

6. Berman JJ. 2008. Treatment of leishmaniasis with miltefosine: 2008 sta-tus. Expert Opin. Drug Metab. Toxicol. 4:1209 –1216.

7. Croft SL, Engel J. 2006. Miltefosine— discovery of the antileishmanialactivity of phospholipid derivatives. Trans. R. Soc. Trop. Med. Hyg.100(Suppl 1):S4 –S8.

8. Sundar S, Jha TK, Thakur CP, Engel J, Sindermann H, Fischer C, JungeK, Bryceson A, Berman J. 2002. Oral miltefosine for Indian visceralleishmaniasis. N. Engl. J. Med. 347:1739 –1746.

9. Chappuis F, Sundar S, Hailu A, Ghalib H, Rijal S, Peeling RW, Alvar J,Boaelaert M. 2007. Visceral leishmaniasis: what are the needs for diagno-sis, treatment and control? Nat. Rev. 5:873– 882.

10. Sundar S, Singh A, Rai M, Prajapati VK, Singh AK, Ostyn B, BoelaertM, Dujardin JC, Chakravarty J. 2012. Efficacy of miltefosine in thetreatment of visceral leishmaniasis in India after a decade of use. Clin.Infect. Dis. 55:543–550.

11. Demicheli C, Ochoa R, Silva JBB, de Melo AL, Falcão CAM, Rossi-Bergmann B, Sinisterra RD, Frézard F. 2004. Oral delivery of meglumineantimoniate-beta-cyclodextrin complex for treatment of leishmaniasis.Antimicrob. Agents Chemother. 48:100 –103.

12. Frézard F, Martins PS, Bahia APCO, le Moyec L, de Melo AL, PimentaAMC, Salerno M, da Silva JBB, Demicheli C. 2008a. Enhanced oraldelivery of antimony from meglumine antimoniate/�-cyclodextrin nano-assemblies. Int. J. Pharm. 347:102–108.

13. Martins PS, Ochoa R, Pimenta AMC, Ferreira LAM, de Melo AL, daSilva JBB, Sinisterra RD, Demicheli C, Frézard F. 2006. Mode of actionof �-cyclodextrin as an absorption enhancer of the water-soluble drugmeglumine antimoniate. Int. J. Pharm. 325:39 – 47.

14. Frézard F, Demicheli C. 2010. New delivery strategies for the old penta-valent antimonial drugs. Expert Opin. Drug Deliv. 7:1343–1358.

15. Ribeiro RR, Ferreira WA, Martins PS, Neto RL, Rocha OG, Le MoyecL, Demicheli C, Frézard F. 2010. Prolonged absorption of antimony(V)by the oral route from non-inclusion meglumine antimoniate-beta-cyclodextrin conjugates. Biopharm. Drug Dispos. 31:109 –119.

16. Subuddhi U, Mishra AK. 2007. Micellization of bile salts in aqueousmedium: a fluorescence study. Colloids Surf. 57:102–107.

17. Filipe V, Hawe A, Jiskoot W. 2010. Critical evaluation of nanoparticletracking analysis (NTA) by NanoSight for the measurement of nanopar-ticles and protein aggregates. Pharm. Res. 27:796 – 810.

18. Schettini DA, Ribeiro RR, Demicheli C, Rocha OGF, Melo MN, Mi-chalick MSM, Frézard F. 2006. Improved targeting of antimony to thebone marrow of dogs using liposomes of reduced size. Int. J. Pharm. 315:140 –147.

19. Yamaoka K, Nakagawa T, Uno T. 1978. Application of Akaike’s infor-mation criterion (AIC) in the evaluation of linear pharmacokinetic equa-tions. J. Pharmacokinet. Biopharm. 6:165–175.

20. Titus RG, Marchand M, Boon T, Louis JA. 1985. A limiting dilutionassay for quantifying Leishmania major in tissues of infected mice. Para-site Immunol. 7:545–555.

21. Walter A, Suchy SE, Vinson PK. 1990. Solubility properties of the alkyl-methylglucamide surfactants. Biochim. Biophys. Acta 1029:67–74.

22. Frézard F, Martins PS, Barbosa MC, Pimenta AM, Ferreira WA, deMelo JE, Mangrum JB, Demicheli C. 2008b. New insights into thechemical structure and composition of the pentavalent antimonial drugs,meglumine antimonate and sodium stibogluconate. J. Inorg. Biochem.102:656 – 665.

23. Ell PJ, Hocknell JML, Jarritt PH, Cullum ID, Lui D, Campos-Costa D,Nowotnik DP, Pickett RD, Canning LR, Neirinckx RDA. 1985. A99TCm labelled radiotracer for the investigation of cerebral vascular dis-ease. Nucl. Med. Commun. 6:437– 441.

24. Neidle S, Snook CF, Murrer BA, Barnard CFJ. 1995. Bis (acetato)amminedichloro (cyclohexylamine) platinum(IV), an orally active anti-cancer drug. Acta Crystallogr. C 51:822– 824.

25. dos Santos Ferreira C, Pimenta AMC, Demicheli C, Frezard F. 2006.Characterization of reactions of antimoniate and meglumine antimoniatewith a guanine ribonucleoside at different pH. Biometals 19:573–581.

26. Gong J, Chen M, Zheng Y, Wang S, Wang Y. 2012. Polymeric micellesdrug delivery system in oncology. J. Control. Release 159:312–323.

27. Torchilin VP. 2007. Micellar nanocarriers: pharmaceutical perspectives.Pharm. Res. 24:1–16.

Fernandes et al.

4236 aac.asm.org Antimicrobial Agents and Chemotherapy

on August 15, 2017 by guest

http://aac.asm.org/

Dow

nloaded from