You are on page 1of 9

Journal of Controlled Release 209 (2015) 229–237

Contents lists available at ScienceDirect

Journal of Controlled Release

journal homepage: www.elsevier.com/locate/jconrel

Capreomycin oleate microparticles for intramuscular administration:


Preparation, in vitro release and preliminary in vivo evaluation
Adrián Cambronero-Rojas a,b, Pablo Torres-Vergara a,⁎,1, Ricardo Godoy a, Carlos von Plessing a,
Jacqueline Sepúlveda c, Carolina Gómez-Gaete a,⁎,1
a
Faculty of Pharmacy, University of Concepción, Concepción, Chile
b
Hospital Dr. Fernando Escalante Pradilla, CCSS, San José, Costa Rica
c
Department of Pharmacology, Faculty of Biological Sciences, University of Concepción, Chile

a r t i c l e i n f o a b s t r a c t

Article history: Capreomycin sulfate (CS) is a second-line drug used for the treatment of multidrug-resistant tuberculosis (MDR-
Received 28 July 2014 TB). The adverse effects profile and uncomfortable administration scheme of CS has led to the development of
Received in revised form 24 April 2015 formulations based on liposomes and polymeric microparticles. However, as CS is a water-soluble peptide that
Accepted 3 May 2015
does not encapsulate properly into hydrophobic particulate matrices, it was necessary to reduce its aqueous sol-
Available online 5 May 2015
ubility by forming the pharmacologically active capreomycin oleate (CO) ion pair. The aim of this research was to
Keywords:
develop a new formulation of CO for intramuscular injection, based on biodegradable microparticles that encap-
Capreomycin oleate sulate CO in order to provide a controlled release of the drug with reduced local and systemic adverse effects.
PLGA The CO-loaded microparticles prepared by spray drying or solvent emulsion-evaporation were characterized in
DPPC their morphology, encapsulation efficiency, in vitro/in vivo kinetics and tissue tolerance. Through scanning elec-
Microparticles tron microscopy it was confirmed that the microparticles were monodisperse and spherical, with an optimal size
Spray drying for intramuscular administration. The interaction between CO and the components of the microparticle matrix
Intramuscular was confirmed on both formulations by X-ray powder diffraction and differential scanning calorimetry analyses.
Tuberculosis
The encapsulation efficiencies for the spray-dried and emulsion-evaporation microparticles were 92% and 56%,
respectively.
The in vitro kinetics performed on both formulations demonstrated a controlled and continuous release of CO
from the microparticles, which was successfully reproduced on an in vivo rodent model. The results of the
histological analysis demonstrated that none of the formulations produced significant tissue damage on the
site of injection. Therefore, the results suggest that injectable CO microparticles obtained by spray drying and sol-
vent emulsion-evaporation could represent an interesting therapeutic alternative for the treatment of MDR-TB.
© 2015 Elsevier B.V. All rights reserved.

1. Introduction of some drugs, which can be very uncomfortable for most of patients
[7]. Failure to comply can lead to the apparition of M. tuberculosis strains
Tuberculosis (TB) is an infectious bacterial disease considered as a that are resistant to first-line drugs.
major cause of illness and death in many countries [1,2]. In humans, Multidrug-resistant tuberculosis (MDR-TB) is often the result of a
this disease is mainly caused by contagion with Mycobacterium tubercu- failed treatment with first line drugs due to patient non-compliance,
losis [3] and pulmonary TB is the commonest clinical presentation [4,5]. prescription of a wrong/incomplete drug scheme, or an ineffective
Pharmacological treatment of TB is basically a combination of drugs directly observed treatment short course (DOTS) therapy. Treatment
that must be given (orally or via injection) under a strict scheme, which of MDR-TB involves the use of second-line drugs, which are more
varies according to the degree of resistance held by the bacterial strain expensive and have more adverse effects than first-line drugs [8].
that infects the patient [6]. Total compliance to TB treatments is difficult Capreomycin sulfate (CS), a peptide obtained from strains of Strepto-
to achieve because of their length, the amount of drugs administered, myces capreolum, is a second-line drug used to treat patients infected
the severity of certain adverse effects and the route of administration with isoniazid and rifampicin-resistant M. tuberculosis strains [9,10].
Despite its efficacy, the adverse effects profile of CS and administration
route, based on repeated intramuscular injections for several days,
⁎ Corresponding authors at: Facultad de Farmacia, Universidad de Concepción, Barrio reduce the probabilities of achieving a full recovery from the disease
Universitario s/n, Concepción, Chile.
E-mail addresses: pabltorr@udec.cl (P. Torres-Vergara), cargomez@udec.cl
[11]. As CS is an effective drug when tolerated by the patient, this
(C. Gómez-Gaete). subject has been a matter of interest for many research groups that
1
Carolina Gómez-Gaete and Pablo Torres-Vergara are both senior authors. have addressed the issue with the current advances in microparticle

http://dx.doi.org/10.1016/j.jconrel.2015.05.001
0168-3659/© 2015 Elsevier B.V. All rights reserved.
230 A. Cambronero-Rojas et al. / Journal of Controlled Release 209 (2015) 229–237

technology. Nowadays, pulmonary delivery of inhalable powders solution was dropwise added to a 1 M aqueous solution of CS under
[12–14], liposomes [15,16] and polymeric microparticles [17,18] has magnetic stirring (700 rpm) in order to obtain the CO ion pair. The
been extensively studied as potential approaches because, presumably, obtained suspension was centrifuged at 12,000 rpm for 10 min at
the drug could reach the lungs with a reduced systemic distribution. To room temperature and then the supernatant was discarded, repeating
date, there is a work that attempted to test an inhalable CS formulation the procedure once. The washed product was reconstituted in 40 mL
in healthy volunteers with interesting results [14]. However, in the case of nanopure water and then freeze dried for 15 h. The resulting solid
of liposomes and microparticles the works of Giovagnoli et al. [15,17] (CO) was collected in an amber bottle and stored at 8 °C.
and Ricci et al. [16] showed that the chemical nature of CS limits consid-
erably its encapsulation into hydrophobic matrices because of its high 2.3. Microparticle preparation
aqueous solubility. In order to overcome this limitation, Schoubben
et al. [18] proposed that reducing the aqueous solubility of capreomycin 2.3.1. Spray drying method
through a simple ion pair exchanging reaction with a hydrophobic DPPC and HA microparticles were prepared by the spray drying
counterion could increase the drug loading within the microparticle method, according to the procedure described by Gómez et al., with
and therefore allowing its use as a inhalable dosage form. The results modifications [21]. DPPC and CO were dissolved in ethanol and HA in
showed that the ion pairing of capreomycin with sodium oleate formed ultra-pure water. Then, a 75/25 (v/v) mixture of both ethanolic and
capreomycin oleate, a product with reduced solubility and capable of aqueous solutions was prepared, keeping a solid concentration in the
being loaded at larger amounts into poly(D,L-lactide-co-glycolide) final solution at 2 g/L. The mixture was fed into a B-290 mini spray
(PLGA) microparticles. Furthermore, recently it has been demonstrated dryer (Büchi, Switzerland), which was set up with the following
that CO pharmacological activity is comparable to the showed by CS conditions: inlet temperature: 110 °C, outlet temperature: 60–65 °C,
[19]. air flow rate: 500 L/h, feed-flow rate: 17 mL/min, aspiration: 100%,
Although pulmonary TB constitutes the majority of cases and the ad- nozzle diameter: 0.7 mm.
ministration of inhalable powders could be a significant step forward in Powder samples were stored at room temperature under vacuum in
terms of efficacy, there is still a problem that needs to be solved for pa- a dessicator immediately after their spray-drying to limit the uptake of
tients that suffer extrapulmonary TB. Therefore, a valid alternative of moisture. It has been shown in a previous work that environmental
taking advantage of the known features present in PLGA-based moisture leads to a modification of the supramolecular organization of
microparticulate systems, including controlled release of drug, reduc- chemicals within particles [21]. The yield was calculated using the
tion of adverse effects and increased stability of the active drug among following equation (Eq. (1)):
others, is developing a depot formulation that can be administered
through intramuscular (IM) injection. On recent years, the increasing mass of the powder collected
% yield ¼  100:
number of depot formulations based in microparticles demonstrates initial mass of solids in the solution prior to spray drying
the IM route could be an improvement in terms of compliance [20]. ð1Þ
The preparation of PLGA microparticles through the solvent
emulsion-evaporation method is a well-documented procedure and
easy to implement, but there are other alternatives of excipients and 2.3.2. Solvent emulsion-evaporation method
preparation methods that could be more efficient when considering a PLGA microparticles loaded with CO were prepared through the sol-
large-scale production. The spray drying method is one of the most vent emulsion-evaporation method [23]. Briefly, CO was dissolved in
reliable ways to achieve the latter purpose because the machinery is 0.3 mL of ethanol and 2.2 mL of acetone. The resulting solution was
designed to allow escalation of the process and also, the resulting mixed with 300 mg of PLGA dissolved in 2.5 mL dichloromethane. The
particulate systems can exhibit some useful features [21]. The excipients organic solution was then pre-emulsified with 20 mL of a 0.25% w/v
used for preparation of spray-dried microparticles, including 1,2- PVA aqueous solution by shaking it at 3200 rpm for 1 min with a Vortex
dipalmitoyl-sn-glycero-3-phosphocholine (DPPC) and hyaluronic acid Genie 2 (Scientific Industries Inc. USA). In order to obtain a
(HA) have demonstrated to be highly stable, biocompatible and microemulsion, the pre-emulsion was homogenized for 30 s at
especially, capable to provide a controlled release of drug [21,22]. 8000 rpm with a mixer system (Heidolph, Germany). The organic
In the present work, the preparation of capreomycin oleate phase was evaporated at room temperature under gentle agitation
microparticles for intramuscular administration by spray drying and (700 rpm) and the microparticle suspension was then completed to
solvent emulsion-evaporation is described. Also, this report provides 20 g by weight with nanopure water. Amber vials were used throughout
the first evidence of the in vitro/in vivo performance showed by a partic- the process to provide protection against UV light, which degrades
ulate system that encapsulates CO, aiming for a potential application in capreomycin [24].
patients who suffer from MDR-TB.
2.4. Quantification of capreomycin in samples
2. Materials and methods
The amount of capreomycin within the microparticles and its
2.1. Materials concentration in liquid samples (e.g. medium for in vitro release and
rat serum) was determined by HPLC using a validated method for
Capreomycin sulfate, sodium oleate and hyaluronic acid sodium salt each matrix, based on the conditions described by Rossi et al. [25]
95% (HA) were purchased from Sigma-Aldrich (USA). Poly (lactic-co- with modifications. Analyses were performed in a LaChrom Elite system
glycolic acid) (PLGA 50:50) Resomer® RG502 was provided from (Merck-Hitachi, Japan), using a Kromasil Reverse Phase C-18 column
Boehringer Ingelheim (Germany) and 1,2-dipalmitoyl-sn-glycero-3- (5 μm, 250 mm), a mobile phase composed of acetonitrile-KH2PO4 buff-
phosphocholine (DPPC) by Genzyme Pharmaceuticals (Switzerland). er solution (pH 2.2; 0.2 M) with 0.2% of heptafluorobutyric acid
All other chemicals and reagents were of the highest purity grade (7.5:92.5 v/v) at isocratic flow rate of 1 ml min−1. The UV detection of
commercially available. capreomycin was set up at 268 nm.

2.2. Capreomycin oleate preparation 2.5. Capreomycin oleate loading within microparticles

Capreomycin oleate was prepared by the method described by The extraction of CO from the microparticle matrix was performed
Schoubben et al. [18] with modifications. Briefly, a 3.5 M sodium oleate by adding 1 mL of methanol or acetonitrile on a weighted amount of
A. Cambronero-Rojas et al. / Journal of Controlled Release 209 (2015) 229–237 231

spray-dried and emulsion-evaporation microparticles, respectively. For Medcenter Einrichtungen, Germany) set up at 37 °C under magnetic
the latter, a previous extra step had to be included, as the particles were stirring at 100 rpm. At predetermined time intervals; 1 mL of medium
obtained by this last method. Briefly, a weighted amount of suspension was withdrawn, being replaced with an equal volume of fresh and
was centrifuged at 14,000 rpm for 15 min at 25 °C in a Z320 centrifuge warm medium. After filtration (0.22 μm PVDF filter), samples were
(Hermle Scientific, Germany) and the supernatant was discarded, stored at 4 °C until their analysis by HPLC as described above. The sam-
leaving the particles ready for the extraction procedure. ples proved to be stable in medium for at least 30 days at 4 °C (data not
The encapsulation efficiency was assessed as the ratio between the shown) and all the experiments were performed at least in triplicate.
determined and theoretical amounts of CO loaded (Eq. (2)) and all the
measurements were carried out in triplicate, expressing the variation 2.10. In vivo studies
as S.D.
2.10.1. Pharmacokinetic study
actual amount of capreomycin The in vivo experiments were conducted according to the University
Encapsulation efficiency ¼  100: ð2Þ
theoretical amount of Concepcion Ethical Committee Regulations for Animal Handling.
Male and female Sprague Dawley rats (250–300 g) obtained from the
University animal unit were used and kept in groups of 5 per cage, at
2.6. Morphological analysis and particle size
room temperature with free access to food and water. CS, CO and CO-
loaded microparticles were administered intramuscularly (n = 3 for
Samples were analyzed in a JMS-6380 LV scanning electronic micro-
each treatment) into semi-membranosus or semi-tendinosus muscles,
scope (JEOL, Japan). The microparticles obtained by solvent emulsion-
at a dose equivalent to 20 mg/kg of capreomycin [26]. The specimens
evaporation were isolated by centrifugation to remove any excess of
were anesthetized with an intraperitoneal solution of ketamine in a sin-
PVA and then were reconstituted in ultrapure water. Samples were
gle 60 mg/kg dose. Samples of blood were withdrawn at predetermined
coated with a gold layer of 150 Å of thickness, using a S 150 metallizer
time intervals by cardiac puncture and after obtaining the required vol-
(Edwards, England). The particle size of the microparticles was
ume of blood the specimen was sacrificed by cervical dislocation. Serum
described by the volume-number mean diameter, calculated from the
samples were obtained by centrifugation of the collected blood at
number distribution estimated (assuming spherical shape) by measur-
5000 rpm for 5 min and were immediately stored at −20 °C until analy-
ing approximately 200 particles located in an arbitrarily chosen area,
sis. The concentration of capreomycin in serum was determined with
with the TRO111-020 software (JEOL, Japan). Span, a mathematical
the HPLC method described above. From the serum concentrations ob-
value related to the granulometric dispersion, was calculated according
tained after each treatment, a time-concentration curve was construct-
to Eq. (3).
ed and certain pharmacokinetic parameters were determined including
D90‐D10 area under the curve (AUC) calculated by the trapezoidal rule method,
Span ¼ ð3Þ maximum concentration reached (Cmax) and time necessary to reach
D50
the maximum serum concentration (tmax).
where D10, D50 and D50 are the particles diameters at 10, 50 and 90%,
respectively, of the accumulated number-distribution calculated from 2.10.2. Histological analysis
the sample. The effect of CS, CO, blank and CO-loaded microparticles on tissue in-
tegrity at the site of injection was assessed by histological analysis of
2.7. Differential scanning calorimetry muscle samples exposed to the compounds and formulations. Rats
were sacrificed by cervical dislocation at predetermined time intervals
DSC analyses were carried out in a DSC-822e system (Mettler post-injection in order to obtain samples from semi-membranous and
Toledo, Germany) coupled to a Haake EK90/MT Intra-Cooler (Haake, semi-tendinous muscles. Samples were fixed in 4% (w/v) paraformalde-
Germany). Approximately 1 mg of sample was weighted into an alumi- hyde dissolved in phosphate buffer saline (pH 7.4), then impregnated
num pan and the analysis parameters were set up in the range of and embedded in paraffin. Each sample was cut with a RM 2145 micro-
20–300 °C, with a scanning rate 10 °C/min under a dynamic nitrogen tome (Leica Microsystems, Switzerland) in order to obtain a 10 μm in
atmosphere (N2 flow rate: 20 mL/min). thickness slice that was treated in xylol to eliminate the paraffin and
finally stained with Mayer's hematoxylin and eosin. After staining,
2.8. Powder X-ray diffraction (XRD) samples were mounted in a glass slide prior to optical microscope
observation with 10× and 40× zoom objectives. The analysis of samples
X-ray diffraction patterns of drugs, excipients, blank an CO-loaded included the study of the following markers of tissue injury: presence of
microparticles were obtained from an EndeavorD4 X-ray diffractometer inflammatory cells, damage of muscle fibers, hyperemia in epimysium,
(Brüker, Germany), built with a fine-focus, Ni filtered Cu Kα1 (λ = 1.54 hyperemia in perimysium, hyperemia in endomysium, hemorrhage in
A) beam, operated at 40 kV and 20 mA. Samples were mounted on a perimysium, hemorrhage in epimysium and presence of fibroblasts.
plate volume of about 1 cm3 and the analyses were performed at
room temperature in the 2θ range between 5° and 45°. 3. Results and discussion

2.9. In vitro release kinetics of capreomycin oleate from microparticles 3.1. Optimization of microparticle formulation and morphological analysis

The in vitro release analysis of CO from the microparticles was For preparation of microparticles by the spray drying method, DPPC
performed under sink conditions (CO aqueous solubility 40 μg/mL), and HA were used as matrix components. DPPC has a high entrapment
using 10 mM HEPES buffer saline (150 mM NaCl, pH 7.4) as medium. capacity of lipophilic compounds [22,27,28], while HA is a polysaccha-
An accurately weighted amount of microparticles, equivalent to 2 mg ride that has been used to improve the physical properties of the
of CO, was resuspended in 10 mL of medium and added into a pre- powders obtained by spray drying and also to modulate the release of
hydrated dialysis membrane pore size of 12,000 kDa. In the case of active compounds [22,29]. In order to optimize the physical and
microparticles obtained by solvent emulsion-evaporation, they were morphological properties of the final formulation, the excipient ratio
centrifuged at 6000 rpm for 10 min before being resuspended. The filled was modified but maintaining the amount of CO (5% w/w) constant in
membranes were placed into beakers containing 240 mL of medium. the mixture. Microparticles with increasing concentrations of HA
Experiments were carried out within a Venticell oven (MMM (5, 15 and 25% w/w) were elaborated and the amount of DPPC was
232 A. Cambronero-Rojas et al. / Journal of Controlled Release 209 (2015) 229–237

adjusted to keep the total solids concentration at 2 g/L. Independent of 3.2. Drug loading
the ratio of the excipients the yield of the process remained at 45 ±
6%. The micrographs of the prepared formulations are shown in Fig. 1. The encapsulation efficiency value obtained for the formulation pre-
At the lowest HA amount used (Fig. 1A), the particles formed aggregates pared by spray drying was 92 ± 8% (n = 5). This result is consistent
and this behavior can be attributed to the tendency of DPPC to form with those described by several reports that regarded this method as
highly cohesive powders. Increasing the HA concentration to 15% w/w being capable of providing high encapsulation values, because the drop-
did not revert the aggregation (Fig. 1B) but at 25% w/w, the resulting lets formed by atomizing the solution contain exactly equal amounts of
particles showed a spheric shape with a smooth surface and less aggre- the components present in the mixture, so each formed droplet
gation (Fig. 1 C). The formulation composed of DPPC 70%, HA 25% and becomes a microparticle when dried [33]. On the other hand, the
5% CO gave microparticles with a volume-number mean diameter of emulsion-evaporation method did not provide the same outcome,
8.86 μm and was selected for further studies. despite the change in lipophilicity done to capreomycin in order to
In the case of microparticles prepared by solvent emulsion- increase its encapsulation into hydrophobic matrices. The amount of
evaporation method (Fig. 2), the formulation parameters were adjusted CO incorporated into the particles was 56 ± 7% (n = 5), value that
in function of the amount of CO used, as the amount of PLGA 50:50 were could be explained by the diffusion of drug from the microdroplets
kept constant. The microparticles prepared with 1 and 5 mg of CO during the evaporation phase [34], or a reduced interaction between
(Fig. 2A and B, respectively) were spherical, monodisperse and without CO and the polymeric matrix.
presence of drug crystals in their surface. When the amount of CO was
increased to 10 mg (Fig. 2C) the particles had crystals adsorbed in its 3.3. Structural analysis
surface, whereas particles containing 15 mg (Fig. 2D) resulted to be
extremely agglomerated and with a great number of pores, probably be- In order to evaluate any possible interactions between CO and the
cause PLGA was unable to encapsulate such amount of drug. As the formulation components, DSC and XRD analyses were carried out on
presence of crystals in the surface is not desired when it comes to both microparticulate formulations. The DSC curves obtained from the
achieving a controlled release of drug from the polymeric matrix, the formulation components used in the method of spray drying (Fig. 3,
formulation loaded with 5 mg provided the best results in this regard, top) show for HA a wide endothermic band in the temperature range
with a volume-number mean diameter of 3.60 μm. The particle size of 30 to 150 °C, which may reflect loss of the volatile components,
distribution and Span value of the selected formulations prepared by while at 230 °C there is an exothermic peak attributed to the degrada-
both methods are summarized in Table 1. tion of the polysaccharide (Fig. 3 top, curve A) [35]. In the case of
Regarding the size of the chosen microparticulate formulations, the DPPC, two characteristic endothermic peaks were observed including
selection was done to overcome certain shortcomings related to the a pretransition peak corresponding to a gel-to-ripple (Tp) phase at
spray-drying technique, including condensation in the drying chamber, ~67 °C and the bilayer main phase transition (Tm) to the liquid crystal-
increase of the output temperature and clogging of the spraying nozzle line phase at ~72 °C (Fig. 3 top, curve B) [36,37]. The CO curve (Fig. 3 top,
due to the increased viscosity of solutions with a higher concentration of curve C) shows a typical endothermic band corresponding to degrada-
solids (larger particle size), among others. Therefore, the emulsion/ tion of the peptidic fraction that conforms the hydrophobic ion pair
evaporation microparticles were prepared with a particle size as similar [18]. The DSC curve of the physical mixture shows again the previously
as possible in order to make a better comparison of the results obtained mentioned peaks of DPPC and HA but not the peak corresponding to CO,
in further studies. probably due to the low amount of drug present in the mixture (Fig. 3.
Although it has been clearly demonstrated that formulations with a top, curve D). The thermograms of both unloaded and CO-loaded
particle size b 10 μm can suffer phagocytosis from macrophages microparticle formulations are similar as they only show the main
surrounding the site of injection, this issue was assumed to not be a phase transition temperature of DPPC (Fig. 3 top, curves E and F,
major inconvenience at the moment of outlining the in vivo experi- respectively).
ments. The evidence from reports that tested microparticle systems de- In the case of microparticles prepared by solvent emulsion-
signed for pulmonary [30,31] and intramuscular [32] delivery of drugs evaporation, DSC curves (Fig. 3, bottom) for CO, PLGA, physical mixture
has proved that despite the smaller particle size and subsequent phago- and unloaded or CO-loaded microparticles were generated as well.
cytosis, it is possible to reach therapeutic plasma concentrations or at Thermograms for PLGA and unloaded microparticles exhibited a glass
least modify the pharmacokinetics to the point of achieving measurable temperature transition around 40 °C (Fig. 3 bottom, curves B and D).
levels in the target tissue when compared to the non-encapsulated The endothermic band corresponding to denaturation of CO was not ob-
drug. Furthermore, the concerns about the safety of polymer-based served for both the physical mixture and CO-loaded microparticles
pulmonary delivery systems in chronic treatments helped to make the (Fig. 3 bottom, curves C and E), which only have the endothermic
decision of using the intramuscular route in this study. peak associated with the glass transition temperature of the polymer.

Fig. 1. SEM micrographs of microparticles prepared by the spray-drying method. Percentage of HA employed in the formulation (% w/w): 5 (A), 15 (B), 25 (C).
A. Cambronero-Rojas et al. / Journal of Controlled Release 209 (2015) 229–237 233

Fig. 2. SEM micrographs of microparticles prepared by the solvent emulsion-evaporation method. Amount of CO employed in the formulation (mg): 1 (A), 5 (B), 10 (C), 15 (D).

The absence of a CO-related peak can be attributed again to the low 3.4. In vitro release studies
amount present in the formulation, since the physical mixture did not
show the band associated to denaturation of the compound. The results of release kinetics performed on free CS and CO, CO-
Confirmation of the physical state of CO within the microparticles loaded spray-dried and solvent emulsion-evaporation microparticles
was done by powder X-ray diffraction. The diffractogram of pure DPPC release profiles are displayed in Fig. 5.
showed peaks in the range of 21–23° 2θ indicating the presence of a As it was expected, the dissolution and diffusion of free CS from the
multi-lamellar bilayer structure (Fig. 4 top, curve A) [36], while HA dialysis bag was fast and complete after 8 h (Fig. 5, top). For CO the dis-
shows the expected lack of peaks characteristic of amorphous structure solution and diffusion rate was slower, demonstrating that the chemical
(Fig. 4 top, curve B). The diffraction pattern of CO confirms its crystalline modification can change the rate of dissolution of capreomycin under
nature (Fig. 4 top, curve C) and the physical mixture of the components the described experimental conditions.
used for preparation of the microparticles revealed the presence of The release profiles of CO from microparticles show that both formu-
peaks that belong CO (Fig. 4 top, curve D), but those were not observed lations exhibit a similar behavior (Fig. 5, bottom). In the first 8 h there is
in the drug-loaded microparticle diffractogram (Fig. 4 top, curve F). The a burst release that can be attributed to the adsorbed drug on the surface
X-ray pattern of the DPPC/HA microparticles greatly differs from the or encapsulated in the outermost layers of the particles. From then, the
patterns obtained from the raw materials, suggesting a different organi- release of CO became continuous, reaching approximately a 50% of re-
zation of both components within the microparticles and an interaction leased drug after 16 days. This behavior remained fairly constant but
between them. These interactions together with the dispersion of CO at the spray-dried particles released the totality of the drug after 26 days,
the molecular level within the particulate matrix might impact the while solvent emulsion-evaporation particles continued releasing drug
release of drug from the formulation. The results obtained for the micro- until day 28.
particles prepared by solvent emulsion-evaporation (Fig. 4, bottom) XRD analyses from previous reports have demonstrated that the mi-
showed a similar outcome, suggesting that CO is dispersed at a molecu- croparticles prepared with DPPC and HA have a particular structure that
lar level in the microparticles obtained by both methods. allows a controlled release of drug as HA is “sandwiched” between DPPC
polar head groups [21]. The interaction between CO and the matrix may
be determined by the oleate group, which could be embedded within
the hydrophobic fractions delaying the release of drug until HA is hy-
Table 1 drated. Moreover, the mechanisms involved on the release of drug
Microparticle size of the selected formulations shown as number distribution. from PLGA microparticles are well known and the profiles obtained in
Method Diameters (μm) ± DS Span this study suggest that the release of CO follows a triphasic pattern.
The first phase corresponds to a fast release of CO that is adsorbed on
D10 D50 D90
the surface of the particle, followed by a second phase in which the
Spray drying 2.06 ± 0.71 7.85 ± 4,27 9.14 ± 2.03 0.87 drug is slowly released from the polymeric matrix that undergoes
Solvent emulsion-evaporation 1.01 ± 0.08 2.94 ± 0,25 3.12 ± 0.96 0.72
through a hydration and erosion process. Finally, the third phase
234 A. Cambronero-Rojas et al. / Journal of Controlled Release 209 (2015) 229–237

Fig 3. DSC curves. Top: HA (A), DPPC (B), CO (C), physical mixture (D), unloaded micro-
particles (E), CO-loaded microparticles (F). Bottom: CO (A), PLGA (B), physical mixture
(C), unloaded microparticles (D), CO-loaded microparticles (E).
Fig 4. XRD diffraction patterns. Top: DPPC (A), HA (B), CO (C), physical mixture (D),
unloaded microparticles (E), CO-loaded microparticles (F). Bottom: CO (A), PLGA (B),
physical mixture (C), unloaded microparticles (D), CO-loaded microparticles (E).
of faster release is often attributed to the onset of the erosion process
[38].

The encapsulation of CO into lipophilic microparticules had an even


3.5. In vivo pharmacokinetic study more marked effect on the disposition of the drug. From the time con-
centration profiles (Fig. 6, bottom), it can be said that both spray-dried
The time-concentration profiles of the studied formulations and and solvent emulsion-evaporation microparticles are capable to keep
their pharmacokinetic parameters are presented in Fig. 6 and Table 2, sustained serum concentrations of capreomycin for at least 6 days,
respectively. with a tmax of 8 h and an observed Cmax of 13.75 and 15.29 μg/mL, re-
The profile obtained from an intramuscular administration of CS spectively. The fast rise in serum concentrations of capreomycin can
(Fig. 6, top) is consistent with the available data, which describes be explained by the transfer of drug that is bound or trapped in the sur-
capreomycin as a drug that is quickly excreted after the intramuscular face of the microparticles [22,38], which may be favored by the abun-
administration of an aqueous solution [11]. The influence of the reduc- dant and stable blood muscular irrigation. The oscillant pattern after
tion in CS aqueous solubility is showed in the CO time-concentration reaching Cmax is probably a product of the controlled release of CO
profile, as there is a significant modification in the pharmacokinetic from the microparticle, whose structure suffers a gradual erosion that
behavior of capreomycin, expressed through an increase in tmax and a provides free drug and leads to a complete disintegration of the matrix
reduction of Cmax. In terms of pharmacokinetic parameters, CO reaches [41]. The obtained AUC values for the microparticulate formulations
its peak concentration at 24 h post-application (20.52 μg/mL), whereas could be justified by employing the same approach utilized to describe
for CS, Cmax (29.32 μg/mL) is reached 1 h after its administration. the differences between the pharmacokinetics of CS and CO. In this
Despite the observed changes, the outcome is not so surprising because case, the controlled and sustained release of drug provided by both for-
as capreomycin is a drug whose elimination goes under first-order mulations becomes another rate-limiting factor that will modify the in-
kinetics [39,40], its rate is a function of the concentration present in tramuscular absorption of capreomycin from the site of injection, thus
serum, so the administration of a salt with reduced solubility will reducing the peak concentrations in serum and consequently the elim-
influence the absorption from the site of injection, becoming the rate- ination rate, resulting finally in an increased residence time. Due to the
limiting step that alters the elimination phase of the drug. The use of exploratory nature of this study it was not possible to characterize in de-
insoluble salts is a well-documented method employed to increase the tail the kinetics involved on each phase, but from what is observed in
residence time of drugs that are quickly excreted when administered the calculated pharmacokinetic parameters and time-concentration
intramuscularly [9,10]. profiles the outcome supports the proposed hypothesis.
A. Cambronero-Rojas et al. / Journal of Controlled Release 209 (2015) 229–237 235

(2 μg/mL) reported for capreomycin sulfate on virulent strains of


M. tuberculosis [11] and a significantly larger exposure to CO, expressed
as AUC. Capreomycin oleate has demonstrated to show activity over
susceptible M. tuberculosis strains that is comparable to the obtained
with CS [19], so in this regard it can be assumed that a CO formulation
should be capable to perform well in a infection in vivo challenge study.
If the results of this study are compared to previous publications that
tested their formulations on animal and human models, it must be
noted that the inhalable powder developed by Fiegel et al. and García-
Contreras et al. [12,39] have the advantage of a reduced systemic expo-
sure to capreomycin, but still require the administration of multiple
doses in order to maintain concentrations within the therapeutic
range. The phase I trial of an inhalable capreomycin formulation in
healthy volunteers performed by Dharmadhikari et al. [14] confirms
this statement. From a practical perspective, depot IM formulations
are generally well tolerated and can solve many issues related to com-
pliance, as patients do not need to be reminded about taking their
drugs and is possible to sort out cases in which the patient is not
collaborative.

3.6. Histological analysis

The results from the histological analysis performed on biopsies of


semi-membranous and semi-tendinous muscle exposed to a single
dose of CO-loaded microparticles prepared by both methods are
shown on Fig. 7.
From the micrographs, it can be observed that after 8 h of injection
Fig 5. Top: dissolution kinetics of CS and CO as free drug in HEPES buffer under sink con- there was presence of inflammatory cells with hyperemia in
ditions. Bottom: Release kinetics of CO from microparticles in HEPES buffer under sink
endomysium and perimysium (Fig. 7A and D). Hemorrhage in the stud-
conditions. Results are expressed as mean ± SD (n = 3).
ied areas was mild to moderate without presence of eosinophils and
scarce fibroblast growth. In biopsies obtained at 48 h (Fig. 7B and
Summarizing the obtained results, there are some points to high- E) there is a clear decrease of inflammatory cells and an improvement
light. A single 20 mg/kg dose of CO-loaded microparticles provides in the lesions. At 192 h (Fig. 7C and F), the tissue showed no signs of in-
sustained concentrations of the drug for at least 8 days, which are con- flammation or hyperemia. The population of fibroblasts and satellite
siderably over the minimum inhibitory concentration (MIC) value cells was low at 48 and 192 h of sampling, without signs of necrosis.
As can be seen, the findings are consistent with the events that arise
after an intramuscular injection [42,43].
It must be noted that the obtained results must be considered as a
preliminary evaluation and should be reproduced with multiple doses
of the selected formulations in order to conclude if the encapsulation
of capreomycin into microparticles has a beneficial effect in terms of tol-
erability, as a single dose not necessarily reflects the changes in tissue
integrity that would appear under a chronic treatment. Nevertheless,
as the injection of CO-loaded microparticles did not show significant dif-
ferences regarding tissue damage and wound recovery when compared
to the exposure to free CS or CO and blank microparticles (data not
shown), the outcome could set a trend for further studies. Furthermore,
the amount of evidence that supports the biocompatibility and
biodegradability of PLGA and DPPC would lead to think that a chronic
intramuscular treatment with microparticulate formulations is feasible
[44].

4. Conclusion

This work has successfully attempted to develop two


microparticulate formulations that provide a controlled release of
capreomycin after their intramuscular administration. The pharmacoki-
netic analysis of the in vivo study performed in rats showed that both
formulations kept sustained concentrations of the drug for several
days without significant tissue damage on the site of injection. Howev-
er, the main difference between the studied formulations is that the
spray-dried microparticles can be prepared at a larger scale and encap-
sulate more efficiently CO in this particular case, so these advantages
Fig 6. Time–concentration profiles obtained from intramuscular administration of the
constitute a selling point for pharmaceutical companies and health
studied formulations to Sprague-Dawley rats. Top: CS and CO as free drug. Bottom: CO- organizations that are interested in counting with a new formulation
loaded microparticles. Results are expressed as mean ± SD (n = 3). capable to give an effective treatment of MDR-TB. Further work will
236 A. Cambronero-Rojas et al. / Journal of Controlled Release 209 (2015) 229–237

Table 2
Pharmacokinetic parameters of the studied formulations. Results are expressed as mean ± SD (n = 3).

Formulation Time AUC 0–t Observed Observed


(h) (μg × h × mL−1) tmax (h) Cmax (μg/mL)

CS 0–8 60.13 ± 1.13 1 29.32 ± 1.27


CO 0–96 747.61 ± 62.07 24 20.52 ± 0.74
Spray-dried microparticles 0–240 2210.01 ± 73.11 8 13.75 ± 2.15
Solvent emulsion-evaporation microparticles 2127.49 ± 202.64 8 15.29 ± 1.84

Fig. 7. Micrographies (10×) of rat muscle biopsies inoculated with CO-loaded microparticles prepared by the spray drying method (a–c), group inoculated with CO-loaded microparticles
prepared by solvent emulsion-evaporation method (d–f), at different time points: 8 h (left column), 48 h (central column) and 192 h (right column).

be focused on testing the formulation in higher mammalian models in [6] J.A. Caminero, G. Sotgiu, A. Zumla, G.B. Migliori, Best drug treatment for multidrug-
resistant and extensively drug-resistant tuberculosis, Lancet Infect. Dis. 10 (2010)
order to assess its safety and efficacy compared to the current treatments. 621–629.
[7] E.L. Corbett, C.J. Watt, N. Walker, D. Maher, B.G. Williams, M.C. Raviglione, C. Dye,
The growing burden of tuberculosis: global trends and interactions with the HIV
Disclosure epidemic, Arch. Intern. Med. 163 (2003) 1009–1021.
[8] R. Pandey, Z. Ahmad, Nanomedicine and experimental tuberculosis: facts, flaws, and
The authors report no conflicts of interest in this work. future, Nanomedicine 7 (2011) 259–272.
[9] S.C. Sweetman, Martindale: the complete drug reference, 37th ed. Pharmaceutical
Press, London, 2011.
[10] P. American Society of Health-System, AHFS drug information 2013, American Soci-
Acknowledgments
ety of Health-System Pharmacists, Bethesda, Md., 2013
[11] Capreomycin, Tuberculosis (Edinb.) 88 (2008) 89–91.
This work was funded by The University of Concepción, through the [12] L. Garcia-Contreras, J. Fiegel, M.J. Telko, K. Elbert, A. Hawi, M. Thomas, J.
DIUC 212.074.048-1.0 grant. The authors would like to acknowledge the VerBerkmoes, W.A. Germishuizen, P.B. Fourie, A.J. Hickey, D. Edwards, Inhaled
large porous particles of capreomycin for treatment of tuberculosis in a guinea pig
help of Dr. Francisco Mucientes and Dr. Miguel Zárraga for their advice model, Antimicrob. Agents Chemother. 51 (2007) 2830–2836.
with histological analysis and preparation of capreomycin oleate, [13] J. Fiegel, L. Garcia-Contreras, M. Thomas, J. VerBerkmoes, K. Elbert, A. Hickey, D.
respectively. Mr. Cambronero-Rojas is supported by the International Edwards, Preparation and in vivo evaluation of a dry powder for inhalation of
capreomycin, Pharm. Res. 25 (2008) 805–811.
Cooperation Agency of Chile (AGCI) and INNOVA BIOBIO 12.211- [14] A.S. Dharmadhikari, M. Kabadi, B. Gerety, A.J. Hickey, P.B. Fourie, E. Nardell, Phase I,
EM.TES and he would like to thank Laboratory Pasteur SA. single-dose, dose-escalating study of inhaled dry powder capreomycin: a new ap-
proach to therapy of drug-resistant tuberculosis, Antimicrob. Agents Chemother.
57 (2013) 2613–2619.
References [15] S. Giovagnoli, P. Blasi, C. Vescovi, G. Fardella, I. Chiappini, L. Perioli, M. Ricci, C. Rossi,
Unilamellar vesicles as potential capreomycin sulfate carriers: preparation and
[1] D.L. Longo, D.L. Kasper, J.L. Jameson, A.S. Fauci, S.L. Hauser, J. Loscalzo, Harrison's physicochemical characterization, AAPS PharmSciTech 4 (2003) E69.
Principles of Internal Medicine, Eighteenth edition McGraw-Hill Medical, [16] M. Ricci, S. Giovagnoli, P. Blasi, A. Schoubben, L. Perioli, C. Rossi, Development of li-
McGraw-Hill, London, New York; London, 2012. posomal capreomycin sulfate formulations: effects of formulation variables on pep-
[2] World Health Organization, Global tuberculosis control: surveillance, planning, fi- tide encapsulation, Int. J. Pharm. 311 (2006) 172–181.
nancing, WHO report 2011, WHO Press, Switzerland, 2011. [17] S. Giovagnoli, P. Blasi, A. Schoubben, C. Rossi, M. Ricci, Preparation of large porous
[3] P. Muttil, C. Wang, A.J. Hickey, Inhaled drug delivery for tuberculosis therapy, biodegradable microspheres by using a simple double-emulsion method for
Pharm. Res. 26 (2009) 2401–2416. capreomycin sulfate pulmonary delivery, Int. J. Pharm. 333 (2007) 103–111.
[4] L. Ziganshina, S. Squire, Fluoroquinolones for treating tuberculosis, Cochrane Data- [18] A. Schoubben, P. Blasi, S. Giovagnoli, M. Ricci, C. Rossi, Simple and scalable method
base of Systematic Reviews 2013, Issue 6, John Wiley & Sons, Ltd., Chichester, UK, for peptide inhalable powder production, Eur. J. Pharm. Sci. 39 (2010) 53–58.
2008. [19] A. Schoubben, P. Blasi, M.L. Marenzoni, L. Barberini, S. Giovagnoli, C. Cirotto, M.
[5] A. Sosnik, A.M. Carcaboso, R.J. Glisoni, M.A. Moretton, D.A. Chiappetta, New old chal- Ricci, Capreomycin supergenerics for pulmonary tuberculosis treatment: prep-
lenges in tuberculosis: potentially effective nanotechnologies in drug delivery, Adv. aration, in vitro, and in vivo characterization, Eur. J. Pharm. Biopharm. 83
Drug Deliv. Rev. 62 (2010) 547–559. (2013) 388–395.
A. Cambronero-Rojas et al. / Journal of Controlled Release 209 (2015) 229–237 237

[20] R.C. Mundargi, V.R. Babu, V. Rangaswamy, P. Patel, T.M. Aminabhavi, Nano/micro oxidative stress, VEGF expression, and vascular leakage in a benzo(a)pyrene-fed
technologies for delivering macromolecular therapeutics using poly(D, L-lactide- mouse model, J. Pharm. Pharmacol. 57 (2005) 851–860.
co-glycolide) and its derivatives, J. Control. Release 125 (2008) 193–209. [33] A.A. da Silva Jr., J.R. de Matos, T.P. Formariz, G. Rossanezi, M.V. Scarpa, E.S. do Egito,
[21] C. Gomez Gaete, N. Tsapis, L. Silva, C. Bourgaux, E. Fattal, Morphology, structure and A.G. de Oliveira, Thermal behavior and stability of biodegradable spray-dried micro-
supramolecular organization of hybrid 1,2-dipalmitoyl-sn-glycero-3-phosphatidyl- particles containing triamcinolone, Int. J. Pharm. 368 (2009) 45–55.
choline-hyaluronic acid microparticles prepared by spray drying, Eur. J. Pharm. Sci. [34] X. Song, Y. Zhao, S. Hou, F. Xu, R. Zhao, J. He, Z. Cai, Y. Li, Q. Chen, Dual agents loaded
34 (2008) 12–21. PLGA nanoparticles: systematic study of particle size and drug entrapment efficien-
[22] C. Gomez-Gaete, N. Tsapis, L. Silva, C. Bourgaux, M. Besnard, A. Bochot, E. Fattal, Su- cy, Eur. J. Pharm. Biopharm. 69 (2008) 445–453.
pramolecular organization and release properties of phospholipid-hyaluronan mi- [35] K. Kafedjiiski, R.K. Jetti, F. Foger, H. Hoyer, M. Werle, M. Hoffer, A. Bernkop-Schnurch,
croparticles encapsulating dexamethasone, Eur. J. Pharm. Biopharm. 70 (2008) Synthesis and in vitro evaluation of thiolated hyaluronic acid for mucoadhesive drug
116–126. delivery, Int. J. Pharm. 343 (2007) 48–58.
[23] C. Gomez-Gaete, N. Tsapis, M. Besnard, A. Bochot, E. Fattal, Encapsulation of dexa- [36] C.W. Park, X. Li, F.G. Vogt, D. Hayes Jr., J.B. Zwischenberger, E.S. Park, H.M. Mansour,
methasone into biodegradable polymeric nanoparticles, Int. J. Pharm. 331 (2007) Advanced spray-dried design, physicochemical characterization, and aerosol disper-
153–159. sion performance of vancomycin and clarithromycin multifunctional controlled re-
[24] Council of the Royal Pharmaceutical Society of Great Britain, The Pharmaceutical lease particles for targeted respiratory delivery as dry powder inhalation aerosols,
Codex: Principles and practice of Pharmaceutics, 12 ed. Pharmaceutical Press, UK, Int. J. Pharm. 455 (2013) 374–392.
London, 1994. [37] S.A. Meenach, F.G. Vogt, K.W. Anderson, J.Z. Hilt, R.C. McGarry, H.M. Mansour, De-
[25] C. Rossi, G. Fardella, I. Chiappini, L. Perioli, C. Vescovi, M. Ricci, S. Giovagnoli, S. sign, physicochemical characterization, and optimization of organic solution ad-
Scuota, UV spectroscopy and reverse-phase HPLC as novel methods to determine vanced spray-dried inhalable dipalmitoylphosphatidylcholine (DPPC) and
Capreomycin of liposomal fomulations, J. Pharm. Biomed. Anal. 36 (2004) 249–255. dipalmitoylphosphatidylethanolamine poly(ethylene glycol) (DPPE-PEG) micropar-
[26] P. Le Conte, F. Le Gallou, G. Potel, L. Struillou, D. Baron, H.B. Drugeon, Pharmacokinet- ticles and nanoparticles for targeted respiratory nanomedicine delivery as dry pow-
ics, toxicity, and efficacy of liposomal capreomycin in disseminated Mycobacterium der inhalation aerosols, Int. J. Nanomedicine 8 (2013) 275–293.
avium beige mouse model, Antimicrob. Agents Chemother. 38 (1994) 2695–2701. [38] S. Fredenberg, M. Wahlgren, M. Reslow, A. Axelsson, The mechanisms of drug re-
[27] V. Codrons, F. Vanderbist, B. Ucakar, V. Preat, R. Vanbever, Impact of formulation and lease in poly(lactic-co-glycolic acid)-based drug delivery systems — a review, Int.
methods of pulmonary delivery on absorption of parathyroid hormone (1–34) from J. Pharm. 415 (2011) 34–52.
rat lungs, J. Pharm. Sci. 93 (2004) 1241–1252. [39] L. Garcia-Contreras, P. Muttil, J.K. Fallon, M. Kabadi, R. Gerety, A.J. Hickey, Pharmaco-
[28] C. Gervelas, A.L. Serandour, S. Geiger, G. Grillon, P. Fritsch, C. Taulelle, B. Le Gall, H. kinetics of sequential doses of capreomycin powder for inhalation in guinea pigs,
Benech, J.R. Deverre, E. Fattal, N. Tsapis, Direct lung delivery of a dry powder formu- Antimicrob. Agents Chemother. 56 (2012) 2612–2618.
lation of DTPA with improved aerosolization properties: effect on lung and systemic [40] B. Reisfeld, C.P. Metzler, M.A. Lyons, A.N. Mayeno, E.J. Brooks, M.A. Degroote, A phys-
decorporation of plutonium, J. Control. Release 118 (2007) 78–86. iologically based pharmacokinetic model for capreomycin, Antimicrob. Agents
[29] S.J. Kim, S.K. Hahn, M.J. Kim, D.H. Kim, Y.P. Lee, Development of a novel sustained re- Chemother. 56 (2012) 926–934.
lease formulation of recombinant human growth hormone using sodium [41] V. Sáenz, J. Hernández, J. Peniche, Las microesferas como sistemas de liberación
hyaluronate microparticles, J. Control. Release 104 (2005) 323–335. controlada de péptidos y proteínas, Biotecnol. Apl. 24 (2007) 98–108.
[30] S. Tiwari, A.P. Chaturvedi, Y.B. Tripathi, B. Mishra, Macrophage-specific targeting of [42] C. Thuilliez, L. Dorso, P. Howroyd, S. Gould, F. Chanut, R. Burnett, Histopathological
isoniazid through mannosylated gelatin microspheres, AAPS PharmSciTech 12 lesions following intramuscular administration of saline in laboratory rodents and
(2011) 900–908. rabbits, Exp. Toxicol. Pathol. 61 (2009) 13–21.
[31] R.K. Verma, J. Kaur, K. Kumar, A.B. Yadav, A. Misra, Intracellular time course, phar- [43] O. Low, H. Lemke, G. Reich, G. Machnik, Studies on the quantification of local tissue
macokinetics, and biodistribution of isoniazid and rifabutin following pulmonary injury following intramuscular injection of aqueous solutions. VIII. The local lesion
delivery of inhalable microparticles to mice, Antimicrob. Agents Chemother. 52 after application of turimycin in different preparations, Exp. Pathol. 17 (1979)
(2008) 3195–3201. 513–515.
[32] N. Bandi, S.P. Ayalasomayajula, D.S. Dhanda, J. Iwakawa, P.W. Cheng, U.B. Kompella, [44] A. Bandhyopadhya, S. Bose, Characterization of Biomaterials, 1 ed. Elsevier, Oxford,
Intratracheal budesonide-poly(lactide-co-glycolide) microparticles reduce UK, 2013..

You might also like