Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

International Journal of Pharmaceutics 587 (2020) 119692

Contents lists available at ScienceDirect

International Journal of Pharmaceutics


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

Development of a stable oral pediatric solution of hydrochlorothiazide by T


the combined use of cyclodextrins and hydrophilic polymers
Marzia Cirria, Francesca Maestrellia, Natascia Menninia, Lorenzo Di Cesare Mannellib,

Laura Michelib, Carla Ghelardinib, Paola Muraa,
a
Department of Chemistry, University of Florence, Florence, Italy
b
Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Pharmacology and Toxicology Section, University of Florence, Florence, Italy

A R T I C LE I N FO A B S T R A C T

Keywords: Hydrochlorothiazide (HCT) is widely used in pediatrics for hypertension management. Due to the lack of pe-
Hydrochlorothiazide diatric commercial forms, community or hospital pharmacies generally prepare HCT extemporaneous pediatric
Pediatric solution suspensions by dispersing in water a portion of a crushed tablet or the drug powder; however, any dose or
Hydroxypropyl-β-cyclodextrin stability control is usually done on these preparations. Obtaining stable HCT solutions is very challenging, due to
Sulfobutylether-β-cyclodextrin
its low water-solubility and pH-dependent degradation. The aim of this work was to develop a stable 2 mg/mL-
Hydrophilic polymers
HCT oral pediatric solution without using co-solvents. Combined use of cyclodextrins (CD) and hydrophilic
Stability
In vivo diuretic effect polymers was exploited to improve poor HCT solubility and stability. HPβCD and SBEβCD were selected, con-
sidering their safe toxicological profiles, while PVP resulted the best among the tested polymers. Low PVP
concentrations (0.2–1.0%) improved the solubilizing efficiency of both CDs, allowing to reach the prefixed HCT
concentration. Different CD-PVP concentrations were used to prepare several 2 mg/mL-HCT solutions in pH 5.5
buffer. The best stability was shown by solutions containing the highest SBEβCD concentration (25 mM), which
allowed a 3-months stability at 4 °C. In vivo studies on rats showed that such formulation allowed a more
pronounced and more reproducible diuretic effect than the corresponding HCT suspension.

1. Introduction and strategies can be seen and an adequate number of purposely for-
mulated drugs become actually available for children.
Most of the drug products available on the market have no in- The lack of adequate pharmaceutical products authorized for pe-
formation in the labeling for the use in pediatrics and there is a general diatric use led to a wide off-label use of dosage forms intended for
lack of medicines properly assessed and with approved indication for adults, by manipulation of tablets, capsules or concentrated suspensions
the pediatric population, thus making the pharmacological treatment in or solutions for obtaining the required proportion of drug dose to be
children a fundamental public health problem (Shah et al., 2007; Amer. administered. However, these manipulations result in poor or un-
Acad. Pediatr., 2014). In fact, the marked differences in pharmacoki- controlled dosing accuracy, especially harmful when very low dosages
netic/pharmacodynamic of drugs between adult and pediatric patients, are required. Dose calculations errors during manipulations can
would require specific studies for the pediatric population (Ivanovska happen, and they have even been recognized as the most frequent
et al., 2014). Instead, clinical trials in this field are still very limited, medication mistake occurring in pediatric patients (Chua et al., 2010).
mainly due to shortage of available people and high complexity of the Moreover, the stability of extemporaneous formulations is limited and
research, joined to economic and ethical reasons. (Rose, 2008). uncertain, from both a physical, chemical and microbiological point of
The need for increasing the availability of child-specific medicines view, and their bioavailability may be variable and/or unknown (Glass
has been well recognized (Ward et al., 2017) and several efforts have and Haywood, 2006; Skwierczynski and Conroy, 2008). Also, the ex-
been made during the last decades for enhancing pediatric drug de- cipients commonly used in dosage forms for adults, such as pre-
velopment, by introducing suitable guidelines and policies and pro- servatives, sweeteners, fillers, solvents, coating and coloring agents, can
moting incentives to investments in new therapies for pediatric use result harmful for children of various ages (Fabiano et al., 2011) and
(WHO, 2011; EMA, 2013; Bucci-Rechtweg, 2017; Tomasi et al., 2017). the off-label use of drugs in pediatric patients leads to their exposure to
However, it will take some time before the effects of these legislations high risks of unanticipated adverse reactions (Bellis et al., 2013).


Corresponding author at: Department of Chemistry, via Schiff 6, 50019 Sesto Fiorentino (FI), Italy.
E-mail address: paola.mura@unifi.it (P. Mura).

https://doi.org/10.1016/j.ijpharm.2020.119692
Received 31 May 2020; Received in revised form 16 July 2020; Accepted 21 July 2020
Available online 24 July 2020
0378-5173/ © 2020 Published by Elsevier B.V.
M. Cirri, et al. International Journal of Pharmaceutics 587 (2020) 119692

Finally, poor palatability and/or taste of these extemporaneous pre- parahydroxybenzoate (0.77 mg/mL) as preservative, and has a 6-
parations can negatively influence the young patient compliance, and months shelf-life (Eur. Paed. Formul., 2019). However, products con-
consequently the relative clinical outcomes. taining high propylene glycol concentrations should not be adminis-
The European Medicinal Agency (EMA) reflection paper on pedia- tered to infants below 4 years, due to possible accumulation problems,
tric formulations indicates oral liquid dosage forms as the form of owing to their limited metabolic pathway, with consequent risks of
choice due to the simplest applicability for administration to infants, toxic effects (EMA, 2006). Moreover, the pediatric use of sucrose as
toddlers and children up to 5 years (EMA, 2006). Liquid formulations sweetener should be generally limited, since it is considered potentially
offer the benefits of great dosing flexibility, swallowing ease, and low cariogenic, and it must be completely avoided in therapy of diabetic
choking risks. On the other hand, they present possible issues con- patients (EMA, 2013).
cerning stability and palatability, acceptable dose volume (preferably Oral liquid HCT pediatric formulations based on solid lipid nano-
≤5 mL for children up to 5 years) and use of child-friendly excipients particles (SLN) and nanostructured lipid carriers (NLC), were pre-
(EMA, 2006). Among oral liquid formulations, solutions are preferred viously successfully developed (Cirri et al., 2017; Cirri et al., 2018a;
to suspensions due to their better acceptance by pediatric patients, less Cirri et al., 2018b). However, in spite of their safety in use, due to the
susceptibility to dosing errors arising from insufficient or non-homo- absence of potentially harmful excipients, higher bioavailability than
geneous re-dispersion of the sediment, and greater ease of administra- simple HCT suspensions, and good stability on storage (up to 3 months
tion, also, if necessary, through enteral feeding tubes. Pediatric oral at 4 °C), such formulations were not suitable for a quick setting-up in
solutions of drugs such as amlodipine (Van der Vossen et al., 2016) or hospital pharmacies, due to the elaborated preparation procedure, re-
lorazepam (Van der Vossen et al., 2017) have been recently successfully quiring a series of steps with the use of specific rather complex appa-
developed. ratus.
Hydrochlorothiazide (HCT) is a thiazide diuretic drug widely used Therefore, based on all these premises, the aim of this work was to
in pediatrics for the hypertension management, and it is part of the List develop a stable oral pediatric aqueous solution of HCT at a target
of Essential Medicines for Children set out by (WHO, 2013)). The re- concentration of 2 mg/mL, easy and fast to prepare also in a pharmacy,
commended pediatric therapeutic HCT dosage is 1 or 2 mg/mL once without using potentially harmful co-solvents, with a well-tolerable pH
daily or two administrations every 12 h, depending on clinical situation value, endowed with an adequate shelf-life, supported by stability
and patient’s response. studies, and thus able to assure right administration dose, efficiency of
However, there are neither age-appropriate, nor liquid HCT for- the therapeutic treatment, and stability during storage.
mulations available on the market. Moreover, the obtainment of HCT Our approach, aimed to overcome the problems of poor solubility
solutions is made very challenging by its low aqueous solubility and its and stability of HCT, was based on the combined use of cyclodextrin
pH-dependent hydrolytic degradation, with the lowest degradation rate (CD) complexation and hydrophilic polymers. CDs are cyclic torus-
around pH 3.0–3.5 (Mollica et al., 1971); Tagliari et al., 2008; Mahajan shaped oligosaccharides consisting of six to eight glucose units, named
et al, 2012). Then, community or hospital pharmacies usually provide α -, β -, and γ-CDs, respectively. CDs, characterized by a hydrophilic
HCT extemporaneous pediatric suspensions produced by dispersion of outer surface and a hydrophobic central cavity, are known for their
the drug powder in sweetened solutions, or suspension of a portion of a ability to form inclusion complexes with a variety of lipophilic drugs,
crushed tablet in suitable commercially available suspension bases, thus thus improving their solubility and/or chemical or physical stability
running into all the disadvantages above illustrated (Glass and (Kurkov and Loftsson, 2011; Jansook et al., 2018). Several studies re-
Haywood, 2006; Chua et al., 2010; Skwierczynski and Conroy, 2008; ported the positive effect of water-soluble polymers in improving CD
Fabiano et al., 2011). complexing, solubilizing and stabilizing efficacy, (Loftsson and
Numerous efforts have been made to prepare stable HCT suspen- Fridriksdottir, 1998; Mura et al., 2001; Ammar et al., 2006; Cirri et al.,
sions. Tagliari et al. (2009) developed a pH 3.3 flocculated HCT sus- 2006; Taupitz et al., 2013; Wang et al., 2013), due to the synergistic
pension containing 0.6% carboxymethylcellulose sodium, well re-dis- effect of polymer and CD on the formation of drug:CD:water-soluble
persible after soft agitation; however, data about its stability on storage polymer ternary systems (Loftsson et al., 1994; Ribeiro et al., 2003;
are lacking. Santoveña et al. (2012) prepared four different oral pe- Loftsson and Másson, 2004).
diatric HCT suspensions, using glycerol as wetting agent, hydro- Some studies concerning the characterization of HCT interactions in
xypropyl-methylcellulose or methylcellulose as stabilizer, and adjusting solution with βCD are reported in literature (Denadai et al., 2006;
pH at 3.0–3.5 with citric acid, to improve drug stability (Mollica et al., Onnainty et al., 2013). The HCT-βCD complex prepared by fluid bed
1971); the best result was obtained with the formulation containing showed an increase in HCT intrinsic dissolution rate and diuretic effect
20% glycerol and 1% methylcellulose, which allowed a 3-weeks stabi- (Pires et al., 2011). Complexation with βCD increased HCT solubility,
lity when stored light-protected at 5 °C. Other authors proved that HCT- protected it from hydrolysis up to 30 days and enhanced its in vivo
spironolactone suspensions (5/5 mg/mL) obtained by commercial ta- bioavailability (Mendes et al., 2016). However, these authors reported a
blets pulverization and then powder dispersion in an aqueous vehicle water solubility value for HCT of 0.037 mg/mL, and obtained a max-
containing an Ora-plus®/Ora-sweet® 1:1 mixture as suspending agent imum solubility increase by βCD complexation at pH 3.3 up to
(pH 4.2–4.4) were stable up to 60 days storage at 5 or 25 °C (Allen and 0.080 mg/mL. Altamimi et al. (2018) showed that βCD complexation
Erickson, 2011). Extemporaneous HCT aqueous suspensions prepared was more effective than the use of surfactants such as sodium lauryl
using drug raw material showed a maximum 7-days shelf life when sulfate or Tween 20 in improving HCT solubility (reaching a maximum
stabilized by adjusting pH to 3.3 (Mendes et al., 2013). A 60 days- solubility value of about 1.4 mg/mL) and in preventing its degradation
stability was found for HCT suspensions (2 mg/mL) obtained by dis- up to 60 days at room temperature.
persing the drug in a 6.5% Syrspend®SF-PH4 aqueous vehicle (pH 4.0) However, it is well known that safety in use and complexing/solu-
(Binson et al., 2019). bilizing power of βCD are limited by its rather low water solubility,
The only study in literature on HCT solutions development regarded which led to the search for more soluble βCD-derivatives (Brewster and
a 0.5 mg/mL solution (Li, 2013), that was the maximum drug con- Loftsson, 2007). Moreover, some of these highly-soluble derivatives,
centration giving a clear solution, according to its saturation solubility such as in particular hydroxypropyl-β-cyclodextrin (HPβCD) and sul-
(≈0.57 mg/mL, Baka et al., 2008; Altamimi et al., 2018). The mono- fobutylether-β-cyclodextrin (SBEβCD), in addition to the higher com-
graphy of this HCT 0.5 mg/mL oral solution is reported on the new on- plexing and solubilizing power, further possess improved toxicological
line European Paediatric Formulary (2019), and consists in a sucrose profiles compared to native βCD (Brewster and Loftsson, 2007), thus
solution (200 mg/mL) buffered at pH 3.2, to reduce drug hydrolysis, making them more suitable for pediatric use.
added with propylene glycol (2.75 mg/mL) as co-solvent and methyl- Therefore, in the present study HPβCD and SBEβCD were selected as

2
M. Cirri, et al. International Journal of Pharmaceutics 587 (2020) 119692

complexing partners for HCT, while hydrophilic polymers poly- The series of HCT simple solutions were prepared by adding the
vinylpyrrolidone, hydroxypropylmethylcellulose and sodium carbox- exactly weighed drug amount (200 mg) in 100 mL of pH 5.5 phosphate
ymethylcellulose were tested for their effectiveness as ternary compo- buffer, and keeping under magnetic stirring at ambient temperature, in
nents. In addition, a maltitol-syrup (Lycasin®80/55) was added to the the presence of different suitable concentrations of SBEβCD or HPβCD
developed formulations, in order to enhance their taste, making them and hydrophilic polymer (as indicated by phase-solubility studies), al-
more palatable and also suitable for children with diabetic diseases. The lowing to obtain complete drug solubilization and achieve the target
developed formulations were tested for physical, chemical and micro- drug solubility (2 mg/mL).
bial stability on storage and for in vivo diuretic activity in rats. The same procedure was followed for preparation of the series of
HCT syrups, where a 20% of the total volume of phosphate buffer was
2. Materials and methods replaced by Lycasin®80/55.

2.1. Materials 2.5. Stability studies under storage

Hydrochlorothiazide (HCT) was a gift from Menarini (Italy). Chemical, physical and microbiological stability studies of the dif-
Hydroxypropyl-β-Cyclodextrin (Kleptose®HP, HPβCD) with an average ferent formulations were performed during 3 months on samples stored
molar substitution (MS) of 0.85 (average molecular weight 1443 g/mol) at room temperatures and at 4 °C, in light-protected glass vials.
and Lycasin®80/55, a maltitol syrup (55% maltitol on dry basis) with Chemical stability studies were performed by biweekly monitoring
an average viscosity of 3000 mPa*s at 20 °C, were kindly donated from the residual drug concentration by spectrophotometric assay (UV–Vis
Roquette (Lestrem, France), while sulfobutylether-β-Cyclodextrin 1601 Shimadzu spectrometer, Kyoto, Japan) at λ = 272.2 nm (see
(Captisol®, SBEβCD) from Ligand Pharmaceuticals, Inc. (San Diego, Section 2.3). In the light of the high accuracy and precision demon-
CA). Polyvinylpyrrolidone K30 (average molecular weight 40000, strated by the spectrophotometric technique, and the absence of in-
PVP), hydroxypropyl-methylcellulose (HPMC), Sodium carbox- terferences in the drug assay, it was considered suitable also for stabi-
ymethylcellulose (NaCMC) and sodium benzoate were supplied by lity studies. In fact, these were merely aimed at determining the
Sigma Aldrich Co. (St. Louis, USA). Purified water was obtained by residual drug concentration present in solution, and not at separating
reverse osmosis (Elix® Millipore, MD, USA). All other chemicals were of and quantifying its eventual degradation products, already known and
analytical grade. characterized (Gumieniczek et al., 2018). HCT formulations were con-
sidered stable when drug concentration remained above 90% of the
2.2. Phase-solubility studies initial concentration, without showing variations in physical properties.
Physical stability was checked weekly monitoring, by visual in-
An excess amount of HCT was added to pH 5.5 phosphate buffer spection, any eventual modification in organoleptic properties such as
aqueous solutions containing increasing concentrations of HPβCD or odour, clarity and colour, appearance of precipitation. Eventual pH
SBEβCD (0–25 mM) in the presence or the absence of a fixed amount of changes of the solutions were controlled by pH measurements, per-
hydrophilic polymer (PVP, HPMC or CMC). The suspensions were kept formed in triplicate using a Basic 20 pH-meter (Crison Instruments,
at constant temperature (25 °C) under magnetic stirring at 500 rpm up Spain).
to equilibrium (48 h), and then an aliquot was filtered (0.45 µm pore Microbiological stability was checked by biweekly monitoring, by
size) and UV assayed according to the procedure described below visual inspection, of possible cloudiness phenomena or possible mold
(section 2.3). No interferences on the spectrophotometric assay of HCT formation.
due to the presence of CD and/or polymer were found. Each test was
performed in triplicate. 2.6. In vivo studies
The apparent stability constant (K1:1) of the HCT-CD complexes
were calculated from the slope of the phase-solubility diagrams and the In vivo studies were carried out using male Sprague-Dawley rats
drug solubility in the medium (Higuchi and Connors, 1965). (Envigo, Varese, Italy), weighing around 200–250 g at the beginning of
The solubilizing efficiency (SE) was obtained by the ratio of drug the experimental procedure. Rats, housed in CeSAL (Centro
solubility in the presence of the highest CD concentration used and that Stabulazione Animali da Laboratorio, University of Florence, Italy)
of the plain drug. were used not earlier than seven days after they arrived. Rats were
housed in 26x41 cm cages (4 in each one), kept at room temperature
2.3. HCT spectrophotometric assay (23 ± 1 °C) with a 12 h light/dark cycle (light at 7 a.m), and nourished
according to a standard laboratory diet, providing water ad libitum. All
Drug concentration in phase-solubility and storage stability studies animal treatments were performed in agreement with the Directive
was spectrometrically determined at 272.2 nm (Shimadzu UV/vis 1601, 2010/63/EU of the European Parliament and of the European Union
Tokyo, Japan). The analytical method was validated according to the Council (22 September 2010) on protection of animals used for scien-
ICH Q2(R1) guidelines (2005). Five standard solutions of the drug were tific purposes. The Florence University ethical policy is in compliance
prepared in the concentration range from 3.0 to 20 mg/L. The equation with the Guide for Care and Use of Laboratory Animals of US National
of the regression line was y = 0.0623x + 0.003, r2 = 0.9991. The LOQ Institutes of Health (NIH Publication No. 85–23, revised 1996;
(Limit of Quantitation) and LOD (Limit of Detection) values were University of Florence assurance number: A5278-01). Formal approval
0.5 mg/L and 0.15 mg/L, respectively. The precision of the method (as to perform the experiments was obtained from the Italian Ministry of
repeatability), determined by a six times analysis of a same drug Health (No. 54/2014-B) and from the Animal Subjects Review Board of
sample, was 0.55%. The accuracy of the method, expressed as % re- Florence University. Experiments involving animals have been reported
covery of a known added drug amount, was 100.5 ± 1.5%. in agreement with ARRIVE guidelines (McGrath and Lilley, 2015).
Every effort was undertaken to lower the number of animals utilised
2.4. Preparation of HCT liquid formulations and minimize their suffering.
The HCT diuretic effect was evaluated following the same procedure
Two different series of liquid HCT (2 mg/mL) formulations were of a previous study of the Authors (Cirri et al. (2017), in accordance
prepared, using, respectively, a simple pH 5.5 phosphate buffer or a 80/ with the method reported by Compaore et al. (2011). Rats were sub-
20 v/v phosphate buffer (pH 5.5):Lycasin®80/55 mixture, (with or divided into four groups (n = 5) for the acute (single oral dose) study,
without 0.05% w/v sodium benzoate as preservative) as solvent. and they had free access to both water and food. Prior to treatment, an

3
M. Cirri, et al. International Journal of Pharmaceutics 587 (2020) 119692

oral dose of 2.5 mL/100 g body weight of physiological saline (0.9% Table 1
NaCl) was administered to all rats (Wiebelhaus et al., 1965), so that to Stability constant (K1:1) and solubilizing efficiency (SE) of HCT complexes with
impose a uniform water and salt load. An hour later, rats received the HPβCD or SBEβCD in the presence of 0.2% w/v of hydrophilic polymer at 25 °C
following oral treatments: group I (control), 0.9% saline; group II, HCT in pH 5.5 phosphate buffer.
suspension 10 mg kg−1; group III, HCT- SBEβCD solution, 10 mg kg−1; CD K 1:1 (M−1) SEa
group IV, HCT- SBEβCD syrup, 10 mg kg−1. Immediately after ad-
HPβCD 106 ± 3 3.1
ministration, the rats were put in metabolic cages (one per cage),
HPβCd + PVP 101 ± 5 3.5
purposely designed to separate fecal material from urine and the cu- HPβCd + HPMC 107 ± 4 3.2
mulative urine and feces output at various times (1, 2, 4, 6 and 24 h) HPβCd + NaCMC 128 ± 5 3.2
was measured (Asif et al., 2014). The diuretic activity was expressed in SBEβCD 213 ± 4 4.7
mL/100 g of body weight (Asif et al., 2014), while the diuretic index SBEβCd + PVP 173 ± 4 4.9
SBEβCd + HPMC 214 ± 4 4.7
was calculated by the ratio between the diuresis of rats treated with the
SBEβCd + NaCMC 253 ± 3 4.7
test formulation and that of the control group (Danamma et al., 2011).
The gastrointestinal motility was evaluated measuring the number of a
ratio between drug water solubility in presence of 25 mM CD used and
faecal pellets produced by the animals during the entire experiment, at alone.
the same times considered for the diuretic activity.
Each value represents the mean ± SEM of 5 rats per group, per- both the selected CDs in the presence of a fixed amount (0.2% w/v) of
formed in two different experimental sets. The analysis of variance was three different hydrophilic polymers, i.e. PVP, NaCMC and HPMC, in
performed by ANOVA. A Bonferroni’s significant difference procedure order to select the most effective ternary component. Phase-solubility
was used as post-hoc comparison. P values of < 0.05 were considered studies were performed in pH 5.5 buffer solution, selected, as explained
significant. Data were analysed using the ‘Origin 9′ software above, as the medium for the preparation of the final oral HCT for-
(OriginLab, Northampton, MA, USA). mulations. AL-type phase-solubility diagrams were obtained for both
CDs in the presence of all the tested polymers, indicating the formation
3. Results and discussion of soluble inclusion complexes (Higuchi and Connors, 1965). The ob-
tained values of apparent stability constants (K1:1) and Solubilizing
The objective of this study was the development of a stable oral Efficiency (SE) are collected in Table 1, together with the corresponding
liquid HCT pediatric formulations with a target concentration of 2 mg/ data previously obtained for HCT:HPβCD and HCT:SBEβCD inclusion
mL, without the use of potentially harmful co-solvents, and with a well- complexes in the absence of polymers (Maestrelli et al., 2020), for
tolerable pH value, higher than the pH 3–4 values commonly used to comparison purposes.
improve its stability against hydrolysis (Mollica et al., 1971; Tagliari As known, the affinity of ionizable drugs, such as HCT, for the in-
et al., 2008; Mahajan et al, 2012) in the until now developed aqueous clusion into the hydrophobic cavity of CDs is inversely related to the
drug suspensions (Mendes et al., 2013, Binson et al., 2019), as well in ionization degree of the host molecule. In particular, in the case of HCT,
the existing 0.5 mg/mL HCT solution (Eur. Paed. Formul., 2019), considering its pKa value (7.9, according to O'Neil, 2013), a decrease of
CD complexation was exploited to reach the prefixed goal, in the the complex stability constant is expected with increasing the pH value,
attempt to improve at the same time drug solubility and stability in due to a progressive decrease in drug hydrophobicity with increasing its
aqueous solution. Our previous studies already proved the solubilizing ionized fraction in solution. However, at pH 5.5 the drug is still largely
ability of different βCD-derivatives towards HCT (Maestrelli et al., present in the unionized form, and previous studies performed on the
2020). Among the examined derivatives, SBEβCD and HPβCD were complex of HCT with native βCD showed that there was not significant
selected for the present study, due to their safer toxicological profiles difference between the K1:1 values determined at pH 3.0 and pH 5.5
than both native βCD or methylated-βCDs (Brewster and Loftsson, (Onnainty et al., 2013).
2007), which make them suitable for parenteral and pediatric use. The obtained K1:1 values indicated that only the addition of NaCMC
With a view to further improve the drug solubility, and reach the led to an increase of the complex stability with both CDs, but it did not
desired target value of 2/mg/mL, the possible favorable effect of the enhance the solubilizing power of HPβCD and SBEβCD toward the
presence of hydrophilic polymers, such as NaCMC, PVP, or HPMC, in drug, while HPMC affected neither complexing nor solubilizing prop-
enhancing the CD solubilizing and stabilizing ability towards HCT has erties of both CDs.
been evaluated, based on the positive results reported in literature On the contrary, PVP was the only hydrophilic polymer able to in-
about the effectiveness of this combined approach (Loftsson and crease both the intrinsic HCT solubility (S0) (0.77 mg/mL vs 0.62 mg/
Fridriksdottir, 1998; Mura et al., 2001; Ammar et al., 2006; Cirri et al., mL), and the CD solubilizing efficiency, even though a slight decrease of
2006; Taupitz et al., 2013; Wang et al., 2013). These polymers were the complex stability constant was observed, due to the increased value
selected also considering their wide and consolidated use as safe of S0.
pharmaceutical excipients (Schwarz, 1990: Rowe et al., 2009; Franco Therefore, based on these findings, PVP was selected for further
and De Marco, 2020). phase-solubility studies, to evaluate the effect of the increase of its
A pH 5.5 phosphate buffer was selected as aqueous medium to concentration (up to 1% w/v) on the CD solubilizing power, and in-
prepare the final HCT solutions, as a good compromise between drug vestigate a possible synergistic effect between polymer and CD in en-
permeability, stability, and tolerability. In fact, based on its pKa value hancing HCT solubility. The results of these studies are shown in Fig. 1,
(7.9, according to O'Neil, 2013), HCT at pH 5.5 was still predominantly and summarized in Table 2 in terms of K1:1, SE and highest HCT con-
present in the unionized form, which is more permeable than the io- centration achieved in solution (Csmax). The reduction of the stability
nized one through the biological membranes. Moreover, even though constant values of HCT complexes with both HPβCD and SBEβCD be-
lower pH values, around 3.0–3.5, should provide a higher drug stability came more evident with increasing the PVP concentration in the
(Mollica et al., 1971; Tagliari et al., 2008; Mahajan et al, 2012), pH 5.5 medium, due to the almost constant values of the slopes of the related
should assure a better tolerability for pediatric patients, considering phase-solubility diagrams (see Fig. 1) joined to the concomitant in-
they have a gastric pH around 4.5 (Nguyen et al., 2015). crease in drug intrinsic solubility S0. Interestingly, a progressive in-
crease in CD solubilizing efficiency was observed, even though there
3.1. Phase-solubility studies was not a true synergistic effect between polymer and CD, since the
total increase in drug solubility was almost equal to the sum of the
An initial screening was conducted by performing experiments with

4
M. Cirri, et al. International Journal of Pharmaceutics 587 (2020) 119692

Fig. 1. Phase-solubility diagrams in pH 5.5 buffered water of hydrochlorothiazide (HCT) with HPβCD (A) or SBEβCD (B), alone (●), or in the presence of PVP at 0.2%
(□), 0.5% (▴) or 1% w/v (○).

Table 2 recommended only in parenteral dosage forms in neonates (EMA,


Stability constant (K1:1) and Solubilizing Efficiency (SE) of complexes of hy- 2006).
drochlorothiazide (HCT) with the tested CDs in the presence of different PVP
concentrations (0.2, 0.5 and 1% w/v) at 25 °C in pH 5.5 phosphate buffer, and
HCT concentration achieved (Cs max) in the presence of 25 mM CD. 3.3. Stability studies of HCT liquid formulations
−1 a
CD % PVP K1:1 (M ) SE Cs max (mg/mL)
All the developed HCT solutions were stored both at room tem-
HPβCD — 106 ± 3 3.1 1.9 perature and at 4 °C, protected from light, and drug concentration,
HPβCD 0.2 101 ± 5 3.5 2.1 possible pH changes, or organoleptic properties modifications or mi-
HPβCD 0.5 84 ± 3 3.7 2.2 crobial contaminations (in terms of mold formation) were biweekly
HPβCD 1.0 78 ± 2 3.8 2.3 monitored. The stability was considered acceptable as long as the
SBEβCD — 213 ± 4 4.7 2.9
SBEβCD 0.2 173 ± 4 4.9 3.0
percentage of HCT remained above 90% of the initial value, and no
SBEβCD 0.5 164 ± 5 5.0 3.1 organoleptic modifications or microbiological contamination occurred.
SBEβCD 1.0 155 ± 4 5.1 3.2 The results of chemical stability studies, in terms of residual HCT
concentration, of the series of pH 5.5 aqueous solutions are depicted in
a
ratio between drug water solubility in presence of 25 mM CD concentration Fig. 2 (A, B, C). Samples stored at room temperature in the absence of
and alone.
0.05 %w/v Na Benzoate showed a faster drug degradation than the
corresponding refrigerated samples, reaching the minimum acceptable
solubility increases given by each carrier separately. value of residual concentration after only 4 weeks (Fig. 2A). Moreover,
However, our first goal was achieved: in fact the joined presence of also mold formation was observed at this time.
PVP and HPβCD allowed to achieve the desired HCT concentration Storage at 4 °C enabled a slowing down of HCT degradation rate: all
(2 mg/mL), not attainable with HPβCD alone, even at the highest the samples showed higher values of HCT residual concentration during
concentration used (25 mM); on the other hand, addition of PVP al- 4 weeks compared to those stored at room temperature (Fig. 2B).
lowed to reduce the amount of SBEβCD to reach the target HCT solu- However, also in these storage conditions, mold formation was detected
bility (2 mg/mL) from 25 mM up to 13 mM. in all the samples after 4 weeks. The only exception was the solution
containing 25 mM SBEβCD, that showed the highest stability, main-
3.2. Preparations of HCT liquid formulations taining a suitable residual concentration up to 8 weeks, without dis-
playing microbiological contamination. Solutions 2 containing 0.05%
Based on the data obtained by phase-solubility studies, four dif- w/v Na Benzoate, stored at 4 °C, showed a longer stability period for all
ferent 2 mg/mL HCT solutions in pH 5.5 phosphate buffer were then set the examined formulations, maintaining an acceptable residual HCT
up, containing, respectively, 25 mM SBEβCD alone, 15 mM SBEβCD concentration during 8 weeks, without showing any mold formation.
with 0.2% PVP, 25 mM HPβCD with 0.2% PVP and 20 mM HPβCD with Among them, the solutions containing the highest CD concentrations
1% PVP were the best, and, in particular, the solution containing 25 mM
A second series of analogous formulations was also prepared where SBEβCD was the only one stable up to three months (Fig. 2C). The
a 20% of the pH 5.5 phosphate buffer solution was replaced by a higher improvement in drug chemical stability observed in the presence
maltitol syrup (Lycasin®80/55), with the aim to exploit its flavouring of SBEβCD than HPβCD can be attributed to the higher stability of its
and sweetening properties for improving taste and palatability of the complex with the drug (see Table 2), considering that the stronger the
formulations. Moreover, the low-glycemic and insulinemic indexes of complex, the lesser the amount of free drug subjected to degradation.
Lycasin®80/55 make it ideal for the preparation of sugar-free syrups. In Complexation with native βCD provided a protective effect up to
addition, the composition of this maltitol syrup has been specifically 30 days against HCT hydrolytic degradation in unbuffered aqueous
developed to generate very little acid production when in contact with solution (Mendes et al., 2016).
the plaque bacteria, so that it is considered tooth-friendly. Moreover, 25 mM SBEβCD also exhibited a preservative effect
The two series of formulations, as solutions or syrups, were pre- against mold formation, previously observed by other Authors and at-
pared both in absence and in presence of Na Benzoate (0.05% w/v) as tributed to its high ability to reduce the water activity of the formula-
preservative, since it is considered suitable for daily use in pediatric tion, which is considered one of the most important factors affecting the
formulations (Embrechts et al., 2010). In fact, its use is not growth of microorganisms (Mosher et al., 2005).

5
M. Cirri, et al. International Journal of Pharmaceutics 587 (2020) 119692

Fig. 3. Stability of the different hydrochlorothiazide (HCT) pH 5.5 buffered


syrups stored at 4 °C in light-protected glass vials, in the absence (A) or in the
presence (B) of Na Benzoate as preservative.

that of the corresponding solutions stored at 4 °C, maintaining residual


HCT concentration values clearly higher than 90%, up to 4 weeks
(Fig. 3A). Nonetheless, the presence of Na Benzoate was less effective in
prolonging the stability of the preparations, allowing an extension of
the stability period of only two further weeks. (Fig. 3B). Moreover,
differently from the case of the simple HCT solutions, no stability dif-
ferences were observed among the different syrup formulations by
varying the CD type (HPβCD or SBEβCD) or its concentration.
No modifications in organoleptic properties such as odour, clarity
and colour, or precipitation phenomena or mold formation, or pH
changes were observed in both the series of formulations (without or
Fig. 2. Stability of the different hydrochlorothiazide (HCT) pH 5.5 buffered
with preservative) during these storage periods (4o 6 weeks, respec-
solutions stored at room temperature (A) or at 4 °C in light-protected glass vials,
in the absence (B) or in the presence (C) of Na Benzoate as preservative. tively). Mold formation was instead detected in both cases after these
periods.

The obtained results are very satisfactory, considering that the sta-
bility of the different HCT suspensions previously developed varied 3.4. In vivo studies
from a minimum of 7 days (Mendes et al., 2013) up to a maximum of
60 days, but at pH 4.2–4.4 (Allen and Erickson, 2011) or 4.0 (Binson On the basis of the stability study results, the formulations con-
et al., 2019). taining 25 mM SBEβCD, both in form of solution and syrup, were se-
Visual inspection of the different solutions did not reveal any sig- lected for in vivo studies on rats, in order to evaluate the HCT diuretic
nificant modifications of their organoleptic properties such as odour, effect, in comparison to the effect evoked by a simple HCT suspension.
clarity and colour, or formation of precipitates during the considered All formulations were orally administered at the same dose of
period. Moreover, pH determinations showed that there was less than 10 mg kg−1. The induced diuretic effect was measured as total volume
0.5 pH unit change throughout the entire study period for all the pre- of excreted urine, diuretic activity (volume of excreted urine per 100 g
parations. body weight), and diuretic index, with respect to a control group
Storage stability studies of syrups (Fig. 3) were carried out only at treated with an equal volume of saline solution. The gastrointestinal
4 °C, since these formulations displayed problems of microbiological motility was also evaluated for safety assessment of the developed
stability within the first two weeks if stored at room temperature. formulations with regard to their potential side effects on gastro-
Syrups without preservative showed a stability substantially similar to intestinal motility, since one of the most common collateral reactions

6
M. Cirri, et al. International Journal of Pharmaceutics 587 (2020) 119692

caused by oral CD intake is an increase in gastrointestinal propulsions,


causing diarrhea (Lovatti Alves et al., 2019). The increased gastro-
intestinal motility could be due to the complex formation of bile salts
with CDs, leading to enhanced intestinal lipids concentrations (Stevens,
1999). Moreover, the concomitant presence of maltitol in the developed
syrups could exert a laxative function and emphasize such an effect. The
gastrointestinal motility was evaluated both assessing possible episodes
of diarrhea and measuring the number of faecal pellets produced by the
animals during the entire experiment, at the same time points con-
sidered for the diuresis.
Concerning the diuretic activity, the SBEβCD-based HTC solution
showed in the first 6 h a more pronounced effect compared to HCT
suspension, probably due to the increased drug solubility, and then
higher drug bioavailability, obtained by CD complexation. An analo-
gous result, in terms of faster effect and increased diuretic activity was
obtained after administration to rats of HCT as βCD complex rather
than as plain drug (Mendes et al., 2016). Surprisingly, the SBEβCD-
based HTC syrup showed, in the first 2 h after administration, values
analogous to those obtained with HTC suspension. This result could be
attributed to a slower drug absorption rate from the syrup, with respect
to the simple solution, due to its higher viscosity. Moreover, both so-
lution and syrup gave rise to more reproducible results than the sus-
pension, due to the better dose uniformity assured by the complete drug
dissolution obtained in such formulations (Fig. 4 A, B). Furthermore,
unlike the suspension, the diuretic activity values obtained by both
formulations containing the drug as SBEβCD complex were still sig-
nificantly different (p < 0.05) from the control up to 24 h.
As for possible gastrointestinal side effects of the formulations ex-
erted by the presence of CDs and/or maltitol, in vivo experiments re-
vealed that none of the tested treatments affected the gastrointestinal
motility with respect to the control group (Table 3). No episodes of
diarrhea occurred, and no increase in the faecal pellets number was
observed, thus allowing to rule out any possible gastrointestinal side
effect of the developed formulations due to the presence of SBEβCD
and/or the concomitant presence of maltitol.

4. Conclusions

The objective of developing an oral pediatric liquid HCT formula-


tion at the target concentration of 2 mg/mL has been successfully
reached. Complexation with CD, in the presence of PVP as ternary
component, not only allowed to increase HCT solubility up to the de-
sired target value, but also to improve its stability in solution against
Fig. 4. Diuretic effect in rats as excreted urine total volume (A), diuretic ac-
hydrolysis, enabling to obtain a solution stable up to 3-months at pH
tivity (excreted urine volume /100 g body weight) (B) and diuretic index (mean
5.5, in the presence of a safe preservative such ad 0.05 %w/v Na urine volume treated group/mean urine volume control group) (C) after a
Benzoate. single oral administration (10 mg/Kg−1) of hydrochlorothiazide (HCT) as pH
The developed oral pediatric HCT solution offers numerous benefits 5.5 buffered solutions or syrups, both in the presence of 25 mM SBEβCD, or as
with respect to the previously developed HCT suspensions (Tagliari aqueous suspension, compared to a control group treated with a same volume
et al., 2009; Allen and Erickson, 2011, Santoveña et al., 2012; Mendes of physiological saline (0.9% NaCl). *P < 0.05 and **P < 0.01 vs control
et al., 2013; Binson et al., 2019), in terms of easier administration (also group; ^P < 0.05 vs HCT suspension (values shown are the mean ± SEM of 5
by enteral feeding tubes), better patient acceptability, lower dosing rats for group).
errors (due to possible not-well re-dispersion of the sediment) and then
more reproducible therapeutic effect, more accurate dose dilution, Table 3
better tolerability in terms of pH (5.5 vs 3.0–3.5), and, additionally, Effects of a single oral administration (10 mg kg−1) in rats of HCT as pH 5.5
higher stability on storage (3 months vs maximum 2 months), despite buffered solution or syrup, both in the presence of 25 mm SBEβCD, or as
the higher pH. aqueous suspension on the gastrointestinal motility (measured as number of
Moreover, the developed HCT solution provides several advantages pellets) vs a control group treated with a same volume of physiological saline
also compared to the 0.5 mg/mL solution present in the European (0.9% NaCl) (Values shown are the mean ± SEM of 5 rats for group).
Paediatric Formulary, such as higher drug concentration in solution Treatments time (h)
(and then lower dose volume to be administered), higher versatility,
allowing accurate dose adjustments by simple dilution, better patient 1 2 4 6

tolerability, due to the less acidic pH (5.5 vs 3.2), high safety, due to the Control 0.0 ± 0.0 0.0 ± 0.0 0.7 ± 0.6 1.0 ± 0.9
absence of potential harmful co-solvents such as propylene glycol HTC + SBEβCD solution 0.0 ± 0.0 1.3 ± 1.6 1.7 ± 1.4 3.0 ± 2.2
(EMA, 2006), as well as of parabens (considering the potential toxic HTC + SBEβCD syrup 0.3 ± 0.3 0.3 ± 0.3 0.3 ± 0.3 2.0 ± 1.2
effects of such preservatives (Nowak et al., 2018). Furthermore, it is HTC suspension 0.3 ± 0.3 0.3 ± 0.3 0.3 ± 0.3 2.0 ± 1.2
suitable also for children with diabetic diseases and does not present

7
M. Cirri, et al. International Journal of Pharmaceutics 587 (2020) 119692

cariogenicity problems, due to the absence of sucrose (EMA, 2013). Cirri, M., Maestrelli, F., Mura, P., Ghelardini, C., Di Cesare Mannelli, L., 2018a. Combined
Moreover, the lack of side effects on the gastrointestinal motility was approach of cyclodextrin complexation and nanostructured lipid carriers for the
development of a pediatric liquid oral dosage form of hydrochlorothiazide.
also proved. Pharmaceutics 10, 287.
Furthermore, the simple developed preparation method could be Cirri, M., Maestrini, L., Maestrelli, F., Mennini, N., Mura, P., Ghelardini, C., Di Cesare
employed to draw up a Standard Operating Procedure (SOP) easy to use Mannelli, L., 2018b. Design, characterization and in vivo evaluation of
Nanostructured Lipid Carriers as a new drug delivery system for hydrochlorothiazide
by both community and hospital pharmacies to prepare a safe HCT oral oral administration in pediatric therapy. Drug Deliv. 25, 1910–1921.
pediatric solution, able to assure the correct dose administering, im- Compaore, M., Lamien-Meda, A., Mogoşan, C., Lamien, C.E., Kiendrebeogo, M.,
prove the efficiency of the treatment and guarantee the formulation Voştinaru, O., Vlase, L., Ionescu, C., Nacoulma, O.G., 2011. Antioxidant: diuretic
activities and polyphenol content of Stereospermum kunthianum Cham. (bigno-
stability up to 3 months. niacee). Nat. Prod. Res. 25, 1777–1788.
Danamma, K.A.K., Jayasimha, B.G., Basha, S.N., 2011. Diuretic activity and study of
Funding biochemical parameters in the methanol extract of Hibiscus esculentus (Okra) fresh
fruits. Int. J. Pharm. Biol. Sci. 1, 160–169.
Denadai, A.M.L., Santoro, M.M., Da Silva, L.H., Viana, A.T., Santos, R.A.S., Sinisterra,
This research did not receive any specific grant from funding R.D., 2006. Self-assembly Characterization of the β-cyclodextrin and
agencies in the public, commercial or not-for-profit sectors. Hydrochlorothiazide System: NMR, Phase Solubility, ITC and QELS. J. Incl. Phenom.
Macro. 55, 41–49.
Embrechts, R., Van Assche, I., De Schaepdrijver, L., Arien, A., 2010. Use and safety of
CRediT authorship contribution statement benzoic acid and sodium benzoate as antimicrobial preservative in oral pediatric
formulations. 3rd Annual Conference, EuPFi, Berlin.
Marzia Cirri: Conceptualization, Data curation, Formal analysis, EMA (European Medicines Agency), 2006. Pre-authorisation of Medicines for human USE.
Reflection paper: Formulations of choice for the paediatric population. 28 July 2006
Investigation, Methodology, Project administration, Supervision, (accessed 2 May 2020).
Writing - original draft, Writing - review & editing. Francesca EMA (European Medicines Agency), 2013. Guideline on pharmaceutical development of
Maestrelli: Data curation, Formal analysis, Writing - review & editing. medicines for paediatric use 1 August 2013. https://www.ema.europa.eu (accessed 2
May 2020). European Paediatric Formulary, 2019. 12/2019:F0001.
Natascia Mennini: Data curation, Formal analysis, Investigation. European Paediatric Formulary, https://paedform.edqm.eu. 2019. (Accessed 14 May
Lorenzo Di Cesare Mannelli: Data curation, Formal analysis, 2020).
Investigation, Methodology. Laura Micheli: Formal analysis, Fabiano, V., Mameli, C., Zuccotti, G.V., 2011. Paediatric pharmacology: Remember the
excipients. Pharmacol. Res 63, 362–365.
Investigation. Carla Ghelardini: Project administration, Resources, Franco, P., De Marco, I., 2020. The Use of Poly(N-vinyl pyrrolidone) in the Delivery of
Supervision. Paola Mura: Conceptualization, Data curation, Drugs: A Review. Polymers 12, 1114.
Supervision, Project administration, Resources, Writing - original draft, Glass, B.D., Haywood, A., 2006. Stability considerations in liquid dosage forms ex-
temporaneously prepared from commercially available products. J. Pharm.
Writing - review & editing.
Pharmaceut. Sci. 9, 398–426.
Gumieniczek, A., Galeza, J., Mroczek, T., Wojtanowski, K., Lipska, K., Pietras, R., 2018.
Declaration of Competing Interest Kinetics and characterization of degradation products of dihydralazine and hydro-
chlorothiazide in binary mixture by HPLCUV, LC-DAD and LC–MS methods.
Chromatographia 81, 1147–1162.
The authors declare that they have no known competing financial Higuchi, T., Connors, K.A., 1965. Phase-solubility techniques. Adv. Anal. Chem. Instr. 4,
interests or personal relationships that could have appeared to influ- 117–122.
ICH Q2(R1), 2005. Validation of analytical procedures: Text and Methodology. https://
ence the work reported in this paper. www.gmp-compliance.org/guidemgr/files/Q2(R1).pdf.
Ivanovska, V., Rademaker, C.M.A., van Dijk, L., Mantel-Teeuwisse, A.K., 2014. Pediatric
References drug formulations: A Review of challenges and progress. Pediatrics 134, 361–372.
Jansook, P., Ogawa, N., Loftsson, T., 2018. Cyclodextrins: structure, physico-chemical
properties and pharmaceutical applications. Int. J. Pharm. 535, 272–284.
Allen, L.V., Erickson, M.A.J., 2011. Stability of extemporaneously prepared pediatric Kurkov, S.V., Loftsson, T., 2011. Cyclodextrins. Int. J. Pharm 453, 167–180.
formulations using Ora-Plus with Ora-Sweet and Ora-Sweet - SF-Part II. Secundum Li, Y.Y. 2013. Development of a hydrochlorothiazide 0.5 mg/mL oral solution for chil-
Artem 6, N. 1. dren. Eur. J. Hosp. Pharm. 20, A-71 (Downloaded May 11, 2020).
Altamimi, M.A., Elzayat, E.M., Alhowyan, A.A., Alshehri, S., Shakeel, F., 2018. Effect of β- Loftsson, T., Fridriksdottir, H., Sigurardottir, A.M., Ueda, H., 1994. The effect of water-
cyclodextrin and different surfactants on solubility, stability, and permeability of soluble polymers on drug-cyclodextrin complexation. Int. J. Pharm. 110, 169–177.
hydrochlorothiazide. J. Molec. Liq. 250, 323–328. Loftsson, T., Fridriksdottir, H., 1998. The effect of water-soluble polymers on the aqueous
American Academy of Pediatrics, 2014. Off-Label use of drugs in children. Pediatrics 133, solubility and complexing abilities of β-cyclodextrin. Int. J. Pharm. 163, 115–121.
563-567 (accessed 4 May 2020). Loftsson, T., Másson, M., 2004. The effects of water-soluble polymers on cyclodextrins
Ammar, H.O., Salama, H.A., Ghorab, M., Mahmoud, A.A., 2006. Implication of inclusion and cyclodextrin solubilization of drugs. J. Drug Deliv. Sci. Technol. 14, 35–43.
complexation of glimepiride in cyclodextrin-polymer systems on its dissolution, sta- Lovatti Alves, Q., Barbosa Camargo, S., Cruz de Jesus, R.L., Silva, D.F., 2019. Drug-β-
bility and therapeutic efficacy. Int. J. Pharm. 320, 53–57. Cyclodextrin inclusion complex: Would be a new strategy to improve
Asif, M., Jabeen, Q., Abdul-Majid, A.M., Atif, M., 2014. Diuretic activity of Boswellia Antihypertensive Therapy? Clin. Res. Trials 5, 1–3.
serrata Roxb oleo gum extract in albino rats. Pak. J. Pharm. Sci. 27, 1811–1817. Mahajan, A.A., Thaker, A.K., Mohanraj, K., 2012. LC, LC-MS/MS studies for the identi-
Baka, E., Comer, J.E.A., Takacs-Novak, K., 2008. Study of equilibrium solubility mea- fication and characterization of degradation products of hydrochlorothiazide and
surement by saturation shake-flask method using hydrochlorothiazide as model establishment of mechanistic approach towards degradation. J. Braz. Chem. Soc. 23,
compound. J. Pharm. Biomed. Anal. 46, 335–341. 445–452.
Bellis, J.R., Kirkham, J.J., Thiesen, S., Conroy, E.J., Bracken, L.E., Mannix, H.L., Bird, Maestrelli, F., Cirri, M., García-Villén, F., Borrego-Sánchez, A., Viseras Iborra, C., Mura,
K.A., Duncan, J.C., Peak, M., Turner, M.A., Smyth, R.L., Nunn, A.J., Pirmohamed, M., P., 2020. Tablets of “Hydrochlorothiazide in Cyclodextrin in Nanoclay”: A New
2013. Adverse drug reactions and off-label and unlicensed medicines in children: a Nanohybrid System with Enhanced Dissolution Properties. Pharmaceutics 12, 104.
nested case-control study of inpatients in a pediatric hospital. BMC Med. 11, 238. https://doi.org/10.3390/pharmaceutics12020104.
Binson, G., Beuzit, K., Migeot, V., Marco, L., Troussier, B., Venisse, N., Dupuis, A., 2019. McGrath, J.C., Lilley, E., 2015. Implementing guidelines on reporting research using
Preparation and physicochemical stability of liquid oral dosage forms free of po- animals (ARRIVE etc.): new requirements for publication in BJP. Br. J. Pharmacol.
tentially harmful excipient designed for pediatric patients. Pharmaceutics 11, 190. 172, 3189–3193.
Brewster, M.E., Loftsson, T., 2007. Cyclodextrins as pharmaceutical solubilizers. Adv. Mendes, C., Costa, A.P., Oliveira, P.R., Tagliari, M.P., Silva, M.A., 2013. Physicochemical
Drug Deliv. Rev. 59, 645–666. and microbiological stability studies of extemporaneous antihypertensive pediatric
Bucci-Rechtweg, C., 2017. Enhancing the pediatric drug development framework to de- suspensions for hospital use. Pharm. Dev. Technol. 18, 813–820.
liver better pediatric therapies tomorrow. Clin. Ther. 39, 1920–1932. Mendes, C., Buttchevitz, A., Kruger, J.H., Müller Kratz, J., Oliveira Simões, C.M., de
Chua, S.S., Chua, H.M., Omar, A., 2010. Drug administration errors in paediatric wards: a Oliveira Benedet, P., Oliveira, P.R., Segatto Silva, M.A., 2016. Inclusion complexes of
direct observation approach. Eur. J. Pediatr. 169, 603–611. hydrochlorothiazide and β-cyclodextrin: Physicochemical characteristics, in vitro and
Cirri, M., Maestrelli, F., Corti, G., Furlanetto, S., Mura, P., 2006. Simultaneous effect of in vivo studies. Eur. J. Pharm. Sci. 83, 71–78.
cyclodextrin complexation, pH, and hydrophilic polymers on naproxen solubilisation. Mollica, J.A., Rehm, C.R., Smith, J.B., Govan, H.K., 1971. Hydrolysis of benzothiadia-
J. Pharm. Biomed. Anal. 42, 126–131. zines. J. Pharmacol. Sci. 60, 1380–1384.
Cirri, M., Mennini, N., Maestrelli, F., Mura, P., Ghelardini, C., Di Cesare Mannelli, L., Mosher, G.L., Pipkin, J.D., Zimmerer, R.O., Fulk, C.F., Thompson, D.O., 2005. Use of
2017. Development and in vivo evaluation of an innovative “Hydrochlorothiazide-in sulfoalkyl ether cyclodextrin as a preservative. Patent Application Publication US
Cyclodextrins-in Solid Lipid Nanoparticles” formulation with sustained release and 2005/0164986 A1, July 28, 2005.
enhanced oral bioavailability for potential hypertension treatment in pediatrics. Int. Mura, P., Faucci, M.T., Bettinetti, G.P., 2001. The influence of poly-vinylpyrrolidone on
J. Pharm. 521, 73–83. naproxen complexation with hydroxypropyl β-cyclodextrin. Eur. J. Pharm. Sci. 13,

8
M. Cirri, et al. International Journal of Pharmaceutics 587 (2020) 119692

187–194. quantify hydrochlorothiazide in oral suspensions for pediatric use. Chromatographia


Nguyen, T.T.P., Bhandari, B., Cichero, J., Prakash, S., 2015. A comprehensive review on 67, 647–652.
in vitro digestion of infant formula. Food Res. Int. 76, 373–386. Tagliari, M.P., Stuzler, S.K., Assreuy, J., Bresolin, T.M.B., Silva, M.A.S., 2009. Evaluation
Nowak, K., Ratajczak-Wrona, W., Górska, M., Jabłonska, E., 2018. Parabens and their of physicochemical characteristics of suspensions containing hydrochlorotiazide de-
effects on the endocrine system. Mol. Cell. Endocrinol. 2018 (474), 238–251. veloped for pediatric use. Lat. Am. J. Pharm. 28, 734–740.
Onnainty, R., Shenfeld, E.M., Quevedo, M.A., Fernández, M.A., Longhi, M.R., Granero, Taupitz, T., Dressman, J., Buchanan, C.M., Klein, S., 2013. Cyclodextrin-water soluble
G.E., 2013. Characterization of the hydrochlorothiazide:β-cyclodextrin inclusion polymer ternary complexes enhance the solubility and dissolution behaviour of
complex. Experimental and theoretical methods. J. Phys. Chem. B 117, 206–217. poorly soluble drugs. Case example: Itraconazole. Eur. J. Pharm Biopharm. 83,
O'Neil, M.J. (Ed.), 2013. The Merck Index - An Encyclopedia of Chemicals, Drugs, and 378–387.
Biologicals. Cambridge, UK, Royal Society of Chemistry, p. 885. Tomasi, P.A., Egger, G.F., Pallidis, C., Saint-Raymon, A., 2017. Enabling development of
Pires, M.A.S., Souza dos Santos, R.A., Sinisterra, R.D., 2011. Pharmaceutical composition pediatric medicines in Europe: 10 years of the EU Pediatric Regulation. Pediatric
of hydrochlorothiazide:β-cyclodextrin Preparation by Three Different Methods, Drugs 19, 505–513.
Physico-Chemical Characterization and In Vivo Diuretic Activity Evaluation. Van der Vossen, A.C., van der Velde, I., Smeets, O.S.N.M., Postma, D.J., Vermes, A., Koch,
Molecules 16, 4482–4499. B.C.P., Vulto, A.G., Hanff, L.M., 2016. Design and stability study of an oral solution of
Ribeiro, L.S.S., Ferreira, D.C., Veiga, F.J.B., 2003. Physicochemical investigation of the amlodipine besylate for pediatric patients Eur. J. Pharm. Sci. 92, 220–223.
effects of water-soluble polymers on vipocetine complexation with β-cyclodextrin Van der Vossen, A.C., van der Velde, I., Smeets, O.S.N.M., Postma, D.J., Eckhardt, M.,
and its sulfobutyl-ether derivative in solution and solid state. Eur. J. Pharm. Sci. 20, Vermes, A., Koch, B.C.P., Vulto, A.G., Hanff, L.M., 2017. Formulating a poorly water
253–266. soluble drug into an oral solution suitable for paediatric patients; lorazepam as a
Rose, K., 2008. Ethical, regulatory and scientific challenges in pediatric drug develop- model drug Eur. J. Pharm. Sci. 100 (2017), 205–210.
ment. Pharmaceutical Medicine. 22, 221–234. Wang, D., Li, H., Gu, J., Guo, T., Yang, S., Guo, Z., Zhang, X., Zhu, W., Zhang, J., 2013.
Rowe, R.R., Sheskey, P.J., Quinn, M.E. (Eds), 2009. Handbook of Pharmaceutical ex- Ternary system of dihydroartemisinin with hydroxypropyl-β-cyclodextrin and le-
cipients, 6th edition, Pharmaceutical Press, London. cithin: Simultaneous enhancement of drug solubility and stability in aqueous solu-
Santoveña, A., Hernández-Paiz, Z., Fariña, J.B., 2012. Design of a pediatric oral for- tions. J. Pharm. Biomed. Anal. 83, 141–148.
mulation with a low proportion of hydrochlorothiazide. Int. J. Pharm. 423, 360–364. Ward, R.M., Benjamin Jr., D.K., Davis, J.M., Gorman, R.L., Kauffman, R., Kearns, G.L.,
Schwarz, W., 1990. A critical review of the kinetics and toxicology of poly- Murphy, M.D., Sherwin, C.M.T., 2017. The Need for Pediatric Drug Development. J.
vinylpyrrolidone (Povidone). CRC Press, Taylor & Francis Group. Pediatr.
Stevens, D.A., 1999. Itraconazole in cyclodextrin solution. Pharmacotherapy 19, Wiebelhaus, V.D., Weinstock, J., Maass, A.R., Brennan, F.T., Sosnowski, G., Larsen, T.,
603–611. 1965. The diuretic and natriuretic activity of triamterene and several related pter-
Shah, S.S., Hall, M., Goodman, D.M., Feuer, P., Sharma, V., Fargason Jr., C., Hyman, D., idines in the rat. J. Pharmacol. Exp. Ther. 149, 397–403.
Jenkins, K., et al., 2007. Off-label drug use in hospitalized children. Arch. Pediatr. WHO (World Health Organization), 2011. Development of pediatric medicines: points to
Adolesc. Med. 161 (3), 282–290. consider in pharmaceutical development. Working document QAS/08.257/Rev.3,
Skwierczynski, C., Conroy, S., 2008. How long does it take to administer oral medicines to August 2011.
children. Paediatr. Perinat. Drug Ther. 8, 145–149. WHO (World Health Organization), 2013. Model List of Essential Medicines for Children,
Tagliari, M.P., Stuzler, S.K., Murakami, F.S., Kuminek, G., Valente, B., Oliveira, P.R., April 2013.https://www.who.int/medicines/publications/essentialmedicines/en/
Silva, M.A.S., 2008. Development and validation of a stability-indicating method to index.html (accessed 4 May 2020).

You might also like