195
Views
19
CrossRef citations to date
0
Altmetric
Original Research

A novel transdermal nanoethosomal gel of betahistine dihydrochloride for weight gain control: in-vitro and in-vivo characterization

, , &
Pages 3377-3388 | Published online: 28 Nov 2017

Abstract

Background

Betahistine dihydrochloride (BDH) is a histamine analog used to control weight gain, with short elimination half-life and gastric irritation as side effects.

Objective

The aim of the current investigation is to formulate and optimize a topical BDH ethosomal gel for weight gain control.

Materials and methods

Box–Behnken design was applied to study the effect of independent variables: phosphatidylcholine (PC), propylene glycol (PG), and ethanol on vesicle size; entrapment efficiency; % drug release; and flux. The morphology and zeta potential of the optimized formulation were evaluated. The % drug release, flux, and pharmacodynamics of the optimized formulation gel were studied.

Results

The size and entrapment efficiency percent had a direct positive relationship with the concentration of PC and negative relationship with ethanol and PG. The % drug release and flux decreased with increasing PC and PG, while ethanol enhanced both responses. Regression modeling indicated a good correlation between dependent and independent variables, where F16 was chosen as the optimized formulation. F16 showed well-defined spherical vesicles and zeta potential of −24 mV, and % release from the gel exceeded 99.5% over 16 h with the flux of 0.28 mg/cm2/h. Food intake and weight gain of rats were significantly decreased after transdermal application of the BDH ethosomal gel when compared with control, placebo, and BDH gel. The histopathological findings proved the absence of inflammation and decrease in adipose tissue.

Conclusion

Results obtained showed a significant, sustained transdermal absorption of BDH ethosomal gel and, consequently, a decrease in food intake and weight gain.

Introduction

Obesity is a common metabolic disorder attributed to increased food intake and physical inactivity; it is considered as a serious risk factor for various diseases such as type II diabetes, coronary artery disease, atherosclerosis, hypertension, and hyperlipidemia.Citation1 Although the pathophysiology behind obesity is poorly understood, overeating is likely one of the most important factors. Thus, decreasing food intake might be effective in the treatment or prevention of obesity. Brain histamine has been shown to have a significant effect on weight gain and food intake. It has been suggested that H1 receptor activation and H3 inhibition decrease food intake.Citation2 Masaki et al have demonstrated that central infusion of histamine decreased fat accumulation and leptin, and enhanced insulin sensitivity in a leptin-resistant obese mouse.Citation3 Kasaoka reported that the histamine neurons system suppresses fat accumulation by regulating mRNA expression of uncoupling protein, which expends energy consumption in rats.Citation4 Yoshimatsu et al reported that histamine neurons downregulate ob gene expression in rat white adipose tissue.Citation5 These data suggest that drugs with H1 agonist and H3 antagonist properties would inhibit food intake.

Betahistine dihydrochloride (BDH), a centrally acting H3 antagonist and H1 agonist, is used for the management of Ménière’s disease due to its vasodilatation action in the cerebral vasculature region and reduction in food intake by increasing brain histamine.Citation6Citation8 Tighilet et al reported that BDH administration for 1 or 3 weeks induced an upregulation of histidine decarboxylase mRNA expression suggesting that histamine synthesis and release are increased under BDH treatment.Citation9 Barak et al reported that BDH, at doses ranging from 16 to 48 mg/day, did not have a significant effect on weight loss compared with a placebo over a 12 weeks interval in 281 adults. However, a subgroup post hoc analysis suggested that weight loss occurred in non-Hispanic women <50 years old, which was consistent with a potential sexual dimorphism.Citation10 The acute effect of BDH on food intake and appetite has been studied in obese women for 24 h, where the highest dose in that study was 1.3 mg/kg of body weight. Results showed no difference to the given BDH dose. However, longer period of administration or higher dose may show the effect of BDH on food intake.Citation11 In animal models, there is some evidence for sexual dimorphism in the histaminergic control of feeding. Female Wistar King rats exposed to dietary histidine decreased their food intake more than do male rats or ovariectomized female rats.Citation12 Two animal studies carried out by Rossi et al and Szelag et al have proved the effectiveness of BDH in reducing food intake.Citation13,Citation14 Unfortunately, BDH has a short plasma half-life (3–4 h), which necessitates its frequent dosing, and consequently, increases the risk of peptic ulcer and gastric irritation when taken orally.Citation15 Several studies have been carried out to control BDH release and explore a new route for its administration like the transdermal route and mucoadhesive tablets for buccal delivery.Citation8,Citation16,Citation17

Ethosomes are specially tailored vesicular carriers characterized by their ability to entrap both hydrophilic and lipophilic drugs. In addition, ethosomes could be applied topically to produce systemic action with high skin tolerability and safety.Citation18Citation21

Furthermore, application of various ethosomal systems containing acyclovir and salicylic acids and clindamycin or prostaglandin E1 to human skin in three clinical studies have not shown adverse skin reactions.Citation22 Paolino et al studied skin tolerability of ethosomes on a healthy human by reflectance spectrophotometry after topical application. The results showed that the ethosomal system did not induce skin erythema whereas the application of hydroethanolic solution with an equal water/ethanol ratio to that of ethosomes resulted in significant skin erythema, which indicate the safety of ethosomes.Citation23

The aim of the current study was to optimize ethosomal gel containing BDH and investigate its effect on decreasing food intake and weight gain. Several ethosomal formulations were prepared and characterized to select an optimum composition. The optimized formulation was subjected to both in-vitro and in-vivo characterizations.

Materials and methods

Materials

BDH was obtained as a gift sample from Pharco Pharmaceuticals (Amriya, Alexandria, Egypt), Soya beans phosphatidylcholine (PC) was purchased from Across Organics (Geel, Belgium), and propylene glycol (PG) was obtained as a gift from Arabcomed Company (Cairo, Egypt). Carbopol 934 (CP) was obtained from SERVA Electrophoresis GmbH (Heidelberg, Germany). Poloxamer 407 was obtained from Sigma-Aldrich (St Louis, MO, USA). The other ingredients used were of analytical grade.

Methods

Experimental design and data analysis

A three-factors, three-level Box–Behnken design was constructed using Design Expert® (Version 7.0.0, Stat-Ease Inc. Minneapolis, MN, USA), and a design matrix comprising 15 experimental runs () was used in ethosomes formulations. The concentration range and ingredient of independent variables were chosen based on preliminary research experiments and data collected from the literature review. The nonlinear computer-generated quadratic model was given as

Y=b0+b1X1+b2X2+b3X3+b12X1X2+b13X1X3+b23X2X3+b11X12+b22X22+b33X32
where Y is the measured response associated with each factor level combination; b0 is the intercept; b1 to b33 are the regression coefficients computed from the observed experimental values of Y; X1, X2, and X3 are the coded levels of independent variables. The terms X1X2 and Xi2 (i=1, 2, or 3) represent the interaction and quadratic terms, respectively.

Table 1 Observed responses in Box–Behnken design for BDH in different ethosomal formulations

Ethosomes preparation

BDH ethosomes were prepared by the method described by Touitou et al with a slight modification.Citation19 Briefly, the ethosomal suspension was prepared by dissolving PC and BDH in absolute alcohol using covered vessels under vigorous stirring at room temperature. PG was added, followed by water under constant mixing at 700 rpm. Mixing was continued for an extra 30 min. The system was maintained at 30°C during the preparation. The preparation was cooled at room temperature before sonication at 4°C for three cycles, 5 min each, and a 5 min rest between cycles.

Characterization of BDH vesicles

Determination of vesicle size

The vesicle size was determined by dynamic light scattering.Citation24 One-milliliter ethosomal suspension was diluted to 10 mL with distilled water and measured using Malvern Zeta Sizer (Zetasizer Ver. 7.11; Malvern Instruments, Malvern, UK) at 25°C. Three replicates of each sample were taken.

Entrapment efficiency percent

Ultracentrifugation method was used for the determination of entrapment efficiency percent (EE%). Vesicular preparations were centrifuged using a cooling centrifuge (Sigma Laborzentrifugen D-37520, Osterode, Germany) at 4°C and 15,000 rpm for 3 h with 15 min pauses after every 30 min. The supernatant was diluted with ethanol and measured with a UV spectrophotometer (Shimadzu UV-1800, Tokyo, Japan) at 261 nm. The mean of the three EE% measurements was then calculated based on the following equation:

EE%=DTDSDT×100(1)
where DT is the theoretical amount of BDH and DS is the detected amount.

In-vitro release studies

The drug released from the prepared vesicles was determined using the dialysis method. A glass cylinder open at both ends was attached to the shaft of the United States Pharmacopoeia (USP) dissolution apparatus (Erweka DT-720, Erweka GmbH, Heusenstamm, Germany), and dialysis membrane was tied at one end of the glass cylinder.Citation25 A volume of ethosomes formulations equivalent to 10 mg of the BDH was accurately pipetted into the cylinder. The cylinder was then suspended in 500 mL of phosphate buffer (pH 7.4) and the dissolution medium was maintained at 37°C±0.5°C. The speed of the device shaft was set at 50 rpm. The time for test run was 6 h and aliquots (2 mL) were withdrawn at intervals of 1, 2, 3, 4, 5, and 6 h and analyzed by UV spectrophotometer at 261 nm.Citation26Citation28 The cumulative amount of BDH released after 6 h (Q6 h) was calculated for each formulation and represented as mean ± SD.

Ex-vivo permeation studies

Permeation studies were performed using the dialysis method described above under the same experimental conditions for in-vitro release studies except for the use of freshly acquired abdominal skin from female albino rats instead of the dialysis membrane.Citation29 The whole abdominal region was carefully shaved and excised immediately before the start of experiment. The skin was later mounted in a receptor compartment with the stratum corneum side facing toward the donor compartment. The cumulative amount of drug permeated per unit area of skin during 6 h (Q6 in µg/cm2) is calculated as follows:

JSS=Amount of drug permeatedTime×area of the membrane(2)
where Jss is the BDH flux (µg/cm2/h), time is 6 h, and effective diffusion surface area is 4.6 cm2.Citation30,Citation31 All experiments were done in triplicate and mean values ± SD were determined.

Optimization and evaluation of the selected formulation

Experimental model evaluation

The coefficient of determination (R2), adjusted R2, predicted R2, and CV% values were used to evaluate the fitness of the model to experimental data. Furthermore, F test was conducted for the analysis of variance (ANOVA) to assess the statistical significance of the quadratic models. The percentage prediction error was calculated for the optimized formulation to verify the above findings.

Formulation and characterization of the optimized BDH ethosomal formulation

The optimized formulation was selected based on minimum particle size and minimum % released as well as maximum EE% and flux. The selected formula was characterized by transmission electron microscopy, zeta potential, in-vitro release study, and ex-vivo permeation study, and eventually incorporated into gel base.

Transmission electron microscopy

The morphology of optimized BDH ethosomal formulation (F16) was visualized by transmission electron microscopy. A droplet of ethosomes suspension after appropriate dilution was placed on carbon-coated copper grid till it gets dry. The formed film was then negatively stained with 1% phosphotungstic acid (PTA); the excess liquid was removed by filter paper and finally allowed to dry again. The air-dried sample was examined by a transmission electron microscopy (TEM) analyzer (FEI Tecnai G2 Spirit Twin, Brno, Czech Republic) at an accelerating voltage of 120 KV, conducted by a VELETA camera.Citation18,Citation32

Zeta potential

Zeta potential was measured for the optimized formulation (F16) using Malvern Zeta Sizer (Zetasizer Ver. 7.11, Malvern Instruments, Malvern, UK) and the mean of three replicates was taken.

Preparation of BDH ethosomal gel

BDH ethosomal gel of the optimized formulation was prepared using the cold method.Citation33 Briefly, carbopol was allowed to be hydrated overnight, and then poloxamer was slowly added to the solution with continuous agitation using a magnetic stirrer. The polymers were stored in a refrigerator overnight, and then the optimized BDH ethosomes (F16) were added to the mixture and probably stirred. After that, the mixture was neutralized by triethanolamine to pH 7.4. In the same manner, the gel of free BDH was prepared. The final concentration of drug was 10 mg BDH/g gel.

In-vitro release and ex-vivo permeation studies of BDH ethosomal gel

The in-vitro release profile of the optimized BDH ethosomal gel was conducted and compared to the aqueous solution of BDH. In addition, the ex-vivo permeation studies of both BDH ethosomal and free BDH gels were performed. As described before, a volume or amount of gel equivalent to 10 mg BDH was accurately transferred to the tester and at predetermined time intervals along 16 h test period, 2 mL samples were withdrawn from the receptor medium and replaced by an equal volume of freshly prepared buffer solution to maintain a constant volume. Samples were filtered through 0.45 µm millipore filter and analyzed spectrophotometrically at 261 nm for the cumulative amount of BDH released (% release). All experiments were performed in triplicate and mean values ± SD were determined.

Pharmacodynamics

Animals

The study was carried out on adult female albino rats. Animals were obtained from the Modern Veterinary Office for Laboratory Animals (Cairo, Egypt). Animals were housed individually in chambers under standard temperature conditions (25°C±0.5°C) and relative humidity (55%±1%) with a 12 h light/dark cycles for 2 weeks for adaptation before being subjected to the experiment and were allowed free access to commercial diet and water.

The animal housing and handling were conducted in compliance with Beni-Suef University guidelines and in accordance with the research protocols established by the Animal Care Committee of the National Research Center (Cairo, Egypt) which followed the recommendation of the National Institute of Health Guide for Care and Use of Laboratory Animal (Publication No 85-23, revised 1985).

Experiment

Food intake and weight gain were recorded according to the method described by Kasaoka et al with minor modification.Citation4,Citation12 Briefly, after 2 weeks of adaptation, the rats were assigned to one of the following four groups (six rats per group) on the basis of body weight (average initial body weight =285 g, range =270–300 g):

  • Group A: the control group, not subjected to any application.

  • Group B: the placebo group, subjected to plain ethosomal gel application.

  • Group C: the free drug group, subjected to free BDH in gel base application.

  • Group D: the ethosomal drug group, subjected to BDH ethosomal gel application.

Each group was allowed free access to experimental diet listed in based on the American Institute of Nutrition’s dietary allowance for rats (AIN-76) to maintain normal growth and weight gain.Citation34 The placebo and BDH ethosomal gel were spread on the animal dorsal skin. The dose was calculated as each animal receives 24 mg BDH/kg/d of body weight. The experiment was designed to investigate the effect of different treatments on food intake and weight gain of the animals.

Table 2 Composition of experimental diet

Food intake and weight gain studies

In the food intake study, each rat received 15 g of the experimental diet daily from the beginning of the experiment, and everyday food intake was recorded by subtracting the weight after feeding the experimental diet from that before feeding. The mean food intake per day for each group was calculated and presented as mean value ± SD. While in weight gain study, animal body weight was checked every 3 days and after 3 weeks, the final weights were measured. The difference in body weight for each animal was recorded and mean difference in body weight ± SD was expressed for each group.

Statistical analysis

All data were statistically analyzed by one-way ANOVA (P-value of ≤0.05) on GraphPad Prism software (version 6) followed by Tukey’s multiple comparison tests. The data were presented as a mean ± SD.

Histopathological study

For the histopathological study, the skin was cut out after animal scarification and kept in formalin solution (10% v/v). It was then fixed in paraffin and sections of 5 µm thickness were cut from each piece and stained with hematoxylin and eosin. The histological changes were examined under an optical microscope.Citation35,Citation36 The safety of BDH ethosomal gel on the skin was evaluated by naked eye observation.

Results and discussion

Preparation and evaluation of BDH ethosomes

Ethosomes are a vesicular system with hydrated bilayers. This system is mainly composed of phospholipid, ethanol, and water. The permeation property of ethosomes is attributed to the entire system.Citation22

Previous studies that had compared permeation enhancement of drugs from ethosomal systems with hydroethanolic solutions showed that permeation enhancement from ethosomes was much greater than would be expected from ethanol alone.Citation31,Citation37Citation39

In our study, BDH-loaded ethosomes were prepared using various concentrations of PC, PG, and ethanol and characterized for appropriate physicochemical attributes. The Box–Behnken design was constructed to study the effect of independent variables on dependent ones (). The relationship between the dependent and the independent variables was further elucidated using 3D-response surface plots and regression equations ().

Table 3 Data of regression analysis for responses Y1, Y2, Y3, and Y4 for fitting to quadratic model

A positive sign in the regression equation represents a synergistic effect, while a negative sign indicates an antagonistic one. The 3D-response surface plots exhibit the effect of two independent variables after fixing the third one at the medium-level value.

The vesicle size and EE% of various BDH ethosomes are presented in . Being hydrophilic, BDH would be incorporated into the internal aqueous core of vesicles as suggested by Lopes et al.Citation37

The vesicle size range was 55.48–291.40 nm, while the EE% of BDH in different ethosomal formulations was in the range 82.31%–83.72% as shown in . An initial decrease in vesicle size and EE% was observed with increasing concentration of PG and ethanol, while there was a marked increase in the vesicle size and EE% with increasing PC concentration. These results were in agreement with the regression equations in .

The reason for size reduction with increasing ethanol and PG concentration could be attributed to the negative charge gained by both ethanol and PG, which caused electrostatic repulsion and some degree of steric stabilization.Citation29

Another explanation can be due to the interaction of ethanol and PG with lipid bilayers, which reduced membrane thickness.Citation38Citation41 The reduction in EE% with the increase in ethanol and PG content could be attributed to increasing the fluidity of vesicles membrane leading to drug leakage.Citation41,Citation42 The surface plots showing the effects of independent variables on vesicle size and EE% are illustrated in .

Figure 1 3D-response surface plots showing the effect of independent variables on vesicle size and EE%.

Abbreviations: EE, entrapment efficiency; PC, phosphatidylcholine; PG, propylene glycol.
Figure 1 3D-response surface plots showing the effect of independent variables on vesicle size and EE%.

The Q6 h of BDH released from the different formulations lies in the range from 42.59% to 72.35%, while the results of drug flux ranged from 0.16 to 0.26 mg/cm2/h. It was also observed that increased concentrations of PC and PG had a negative relationship with % BDH released and flux, while ethanol showed a positive relationship with both responses. These findings may be attributed to the dual effect of ethanol on both the lipid bilayer in the stratum corneum and the vesicle lipid bilayer. Ethanol has a fluidizing effect on phospholipid bilayers, producing compact, soft, deformable vesicles, along with the interaction between ethanol and the polar head group region of the lipid molecule resulting in a decrease in transition temperature of stratum corneum lipids with a consequent increase in membrane fluidity and BDH ethosomes partitioning.Citation21,Citation43Citation47

As PG concentration increased the % released and flux were decreased due to increased viscosity of the ethosomal suspension, reducing vesicles fluidity and decreasing skin penetration; however, the use of PG with ethanol might improve ethosomes stability.Citation48 The effects of the independent variables on % released and flux are illustrated in the surface plots in and in .

Figure 2 3D-response surface plots showing the effect of independent variables on % cumulative release (Q6 h) and flux.

Abbreviations: PC, phosphatidylcholine; PG, propylene glycol.
Figure 2 3D-response surface plots showing the effect of independent variables on % cumulative release (Q6 h) and flux.

Our results agree with Dayan and Touitou who had studied the skin permeation of minoxidil and testosterone from ethosomal formulations.Citation18 Also, they agree with Paolino et al, who had studied the inflammatory activity of ethosomal ammonium glycyrrhizinate for transdermal delivery.Citation23 Also, they agree with the results of Dayan and others who studied the efficacy of ethosomes as carriers for skin delivery of trihexyphenidyl HCl.Citation18

It was found that the experimental values fell close to unity lines when plotted against predicted dependent variables, indicating a good correlation between dependent and independent variables. Furthermore, the three independent variables were significant model terms.

The optimization process of the design was based on minimizing vesicle size and the release rate and, on the other hand, maximizing flux and EE% to ensure permeability of vesicles through the skin and longer stability. F16 was selected as optimized formulation, and comparing experimental and predicted values of F16 showed close resemblance with small relative prediction error (<11%), suggesting the validity of the generated regression equations. The values of size, EE%, Q6 h, and flux of the optimized formulation were 55.28±2.25 nm, 85.46%±0.50%, 46.50%±0.79%, and 0.21±0.02 mg/cm2/h, respectively.

As shown in , the TEM images of the optimized BDH ethosomes formulation showed well-identified spherical vesicles with sealed nanovesicular structure. The zeta potential of the optimized formulation was −24 mV, which indicated stability of vesicles against aggregation and confirmed the effect of ethanol on vesicles repulsion.Citation39

Figure 3 TEM photomicrograph of the optimized BDH ethosomes (F16).

Abbreviations: BDH, betahistine dihydrochloride; TEM, transmission electron microscopy.
Figure 3 TEM photomicrograph of the optimized BDH ethosomes (F16).

Evaluation of BDH ethosomal gel

The in-vitro release profiles of BDH from BDH ethosomal gel in comparison with that of an aqueous solution as a control are represented graphically in . About 37.5% was released from BDH ethosomal gel, compared to 99% released from an aqueous solution of BDH within 7 h.

Figure 4 Cumulative % released from aqueous solution of BDH and BDH ethosomal gel in phosphate buffer pH 7.4.

Abbreviation: BDH, betahistine dihydrochloride.
Figure 4 Cumulative % released from aqueous solution of BDH and BDH ethosomal gel in phosphate buffer pH 7.4.

The in-vitro release of BDH ethosomal gel followed zero-order kinetics (R2 =0.99) indicating a controlled release dosage form. This might be due to the surface active properties of poloxamer gel and would add further advantage to this system that is capable of delivering BDH at a controlled release rate.Citation49

The ex-vivo permeation results of both free BDH in gel and BDH ethosomal gel in phosphate buffer pH 7.4 are shown in . The retardation of drug release from BDH ethosomal gel compared to free BDH in gel could attribute to two main effects. Firstly, the viscosity of the gel and the reduction in the size of the extramicellar aqueous channels of poloxamer matrix through which the drug diffuses.Citation50,Citation51 Secondly, combining ethanol and PG in ethosomes can significantly enhance deposition of drugs on the skin.Citation48 The cumulative % released and the cumulative amount permeated of the drug from the BDH ethosomal gel exceeded 99.5% over 16 h with a flux of 0.28 mg/cm2/h.

Figure 5 Ex-vivo permeation of both free BDH in gel and BDH ethosomal gel in phosphate buffer pH 7.4.

Abbreviation: BDH, betahistine dihydrochloride.
Figure 5 Ex-vivo permeation of both free BDH in gel and BDH ethosomal gel in phosphate buffer pH 7.4.

Pharmacodynamic activity

With regard to weight gain results (), free BDH gel showed a significant difference from control and placebo (P<0.05), while ethosomal gel application showed a significant difference from control, placebo, and free BDH gel with P<0.05. This could be attributed to permeation enhancer effect of ethosomes, which delivers more BDH to systemic circulation than that delivered from BDH gel alone. The results obtained were in agreement with the two animal studies carried out by Rossi et al and Szelag et al, which have evaluated the effect of BDH on food intake in animals.Citation13,Citation14

Figure 6 Results of pharmacodynamic activity of BDH.

Notes: (A) Weight gain study. (B) Food intake study. *Significant difference from control and placebo. **Significant difference from control, placebo, and BDH gel with P<0.05.
Abbreviation: BDH, betahistine dihydrochloride.
Figure 6 Results of pharmacodynamic activity of BDH.

Rossi investigated the effect of different doses of BDH on water and food intake in 12 pygmy goats and concluded that the injection of BDH (4 mg/kg body weight in power of 0.75) significantly reduced food intake at 1 h or during 6 h (8 mg/kg body weight in power of 0.75) post injection. Szelag studied the effect of systemic BDH on food intake of rats. Animals were divided into three experimental groups. The first was on a normal diet, the second one was deprived of food, and the third group was accustomed to “tasty” food. The results of the three experimental groups concluded that BDH at the dose of 24 mg/kg revealed stronger activity, reducing food intake.

Barak et al had studied the effect of BDH orally on reducing weight gain associated with olanzapine.Citation52 In this study, 36 schizophrenic patients treated with olanzapine were randomized to BDH and matching placebo. The patients treated with BDH had less weight gain compared with the placebo group, but due to small sample size, the effect did not reach statistical significance. Furthermore, a study carried out by Zheng et al evaluated the effect of BDH on olanzapine-induced intestinal fat deposition using Caenorhabditis elegans as a model for obesity.Citation53 This study proved that the combination of olanzapine with BDH reduced intestinal fat deposition compared with the olanzapine-treated group. Furthermore, the study indicated that the appropriate BDH dose to be used for this indication should be tripled due to the short half-life of BDH, so a sustained release formulation may have approved the effect.

Regarding food intake, application of BDH ethosomal gel significantly decreased the food intake of rats as compared with control and placebo and free BDH gel groups ().

The decrease in weight gain and food intake observed with BDH is due to the H3 antagonist action and H1 agonist action of BDH, which increase brain histamine concentration, inhibiting food intake and thus decreasing weight gain.Citation3

Histopathological investigation

The histopathological investigation of skin and adipose tissue for the BDH ethosomal gel compared with control and placebo and free BDH gel are shown in . The structure and the thickness of the horny layer of the BDH ethosomal gel-treated group did not show any differences compared with control and placebo. Additionally, no infiltration of inflammatory cells to the skin treated with BDH ethosomal gel was observed. These outcomes are in line with the safety and tolerability of the ethosomal system reported in the literature.Citation22

Figure 7 Light photomicrographs of morphology and adipose tissue of rat’s skin after (A) control, (B) placebo, (C) BDH gel, and (D) BDH ethosomal gel applications at a magnification of 40× and 100× respectively.

Abbreviation: BDH, betahistine dihydrochloride.
Figure 7 Light photomicrographs of morphology and adipose tissue of rat’s skin after (A) control, (B) placebo, (C) BDH gel, and (D) BDH ethosomal gel applications at a magnification of 40× and 100× respectively.

A study carried out by Shumilov and Touitou,Citation54 evaluated the safety of transdermal ethosomal system containing buspirone. The results showed no infiltration of inflammatory cells to the skin, and also no changes in the structure and thickness of the horny layer.

Regarding the adipose histopathological investigation, a marked decrease in adipose tissue with the use of a BDH ethosomal gel for 3 weeks was observed; however, large fat cells of adipose tissue were seen with both the control and the placebo groups with no significant difference in size.

Conclusion

A novel ethosomal gel of BDH with satisfactory release characteristics was successfully prepared. A mathematical model was developed connecting the independent variables with the measured responses. The quadratic regression modeling was a proper way for selecting the best drug ethosomal formulation based on minimizing vesicle size as well as the release rate and maximizing flux and EE% to ensure permeability of vesicles through the skin and longer stability. Poloxamer gel had offered a further advantage in delivering BDH at a controlled release rate. BDH ethosomal gel showed effective, sustained transdermal absorption and central action in decreasing food intake and, consequently, the weight gain.

Disclosure

The authors report no conflicts of interest in this work.

References

  • KaplanNMThe deadly quartet: upper-body obesity, glucose intolerance, hypertriglyceridemia, and hypertensionArch Intern Med19891497 1514 15202662932
  • SakataTYoshimatsuHKurokawaMHypothalamic neuronal histamine: implications of its homeostatic control of energy metabolismNutrition1997135 403 4119225331
  • MasakiTYoshimatsuHChibaSWatanabeTSakataTCentral infusion of histamine reduces fat accumulation and upregulates UCP family in leptin-resistant obese miceDiabetes2001502 376 38411272150
  • KasaokaSTsuboyama-KasaokaNKawaharaYHistidine supplementation suppresses food intake and fat accumulation in ratsNutrition20042011 991 99615561489
  • YoshimatsuHHidakaSNiijimaASakataTHistamine neurons down-regulate ob gene expression in rat white adipose tissueInflamm Res200150Suppl 2 S72 S7311411610
  • TighiletBTrottierSLacourMDose- and duration-dependent effects of betahistine dihydrochloride treatment on histamine turnover in the catEur J Pharmacol20055231–3 54 6316226741
  • JainRYadavRKRatherJAVoltammetric assay of anti-vertigo drug betahistine hydrochloride in sodium lauryl sulphateColloids Surf A Physicochem Eng Asp20103661–3 63 67
  • HathoutRMNasrMTransdermal delivery of betahistine hydrochloride using microemulsions: physical characterization, biophysical assessment, confocal imaging and permeation studiesColloids Surf B Biointerfaces2013110 254 26023732802
  • TighiletBTrottierSMourreCChotardCLacourMBetahistine dihydrochloride interaction with the histaminergic system in the cat: neurochemical and molecular mechanismsEur J Pharmacol20024461–3 63 7312098586
  • BarakNGreenwayFLFujiokaKAronneLJKushnerRFEffect of histaminergic manipulation on weight in obese adults: a randomized placebo controlled trialInt J Obes20083210 1559 1565
  • AliAHYanoffLBSternEAAcute effects of betahistine hydrochloride on food intake and appetite in obese women: a randomized, placebo-controlled trialAm J Clin Nutr2010926 1290 129720881066
  • KasaokaSKawaharaYInoueSGender effects in dietary histidine-induced anorexiaNutrition2005217–8 855 85815975494
  • RossiRDel PreteEScharrerEEffect of the H 1-histamine receptor agonist betahistine on drinking and eating behavior in pygmy goatsPhysiol Behav1999663 517 52110357443
  • SzelgATrochaMMerwid-LadABetahistine inhibits food intake in ratsPol J Pharmacol2001536 701 70711985350
  • SweetmanSCMartindale The Complete Drug ReferenceThirty-sixth editionPharmaceutical Press2009
  • HedaAASonawaneARNaranjeGHSomaniVGPuranikPKDevelopment and in vitro evaluation of betahistine adhesive-type transdermal delivery systemTrop J Pharm Res201096 516 524
  • El-NabarawiMAAliAAAboudHMHassanAHGodahAHTransbuccal delivery of betahistine dihydrochloride from mucoadhesive tablets with a unidirectional drug flow: in vitro, ex vivo and in vivo evaluationDrug Des Devel Ther201610 4031 4045
  • DayanNTouitouECarriers for skin delivery of trihexyphenidyl HCl: ethosomes vs. liposomesBiomaterials20002118 1879 188510919691
  • TouitouEGodinBDayanNWeissCPiliponskyALevi-SchafferFIntracellular delivery mediated by an ethosomal carrierBiomaterials20012222 3053 305911575480
  • TouitouEGodinBEthosomes for skin deliveryJ Drug Deliv Sci Technol2007175 303 308
  • ScognamiglioIDe StefanoDCampaniVNanocarriers for topical administration of resveratrol: a comparative studyInt J Pharm20134402 179 18722909994
  • AinbinderDPaolinoDFrestaMTouitouEDrug delivery applications with ethosomesJ Biomed Nanotechnol201065 558 56821329048
  • PaolinoDLucaniaGMardenteDAlhaiqueFFrestaMEthosomes for skin delivery of ammonium glycyrrhizinate: in vitro percutaneous permeation through human skin and in vivo anti-inflammatory activity on human volunteersJ Control Release20051061–2 99 11015935505
  • AboudHMAliAAEl-MenshaweSFElbaryAANanotransfersomes of carvedilol for intranasal delivery: formulation, characterization and in vivo evaluationDrug Deliv2016237 2471 248125715807
  • ZakiRMAliAAEl MenshawiSFBaryAAEffect of binary and ternary solid dispersions prepared by fusion method on the dissolution of poorly water soluble diacereinInt J Drug Deliv201351 99 109
  • ShammaRNBasaliousEBShoukriRDevelopment of novel sustained release matrix pellets of betahistine dihydrochloride: effect of lipophilic surfactants and co-surfactantsPharm Dev Technol2012175 583 59321770719
  • MishraGPKinserRWierzbickiIHAlanyRGAlaniAWIn situ gelling polyvalerolactone-based thermosensitive hydrogel for sustained drug deliveryEur J Pharm Biopharm2014882 397 40524931340
  • QianSWongYCZuoZDevelopment, characterization and application of in situ gel systems for intranasal delivery of tacrineInt J Pharm20144681–2 272 28224709220
  • AhadAAqilMKohliKSultanaYMujeebMEnhanced transdermal delivery of an anti-hypertensive agent via nanoethosomes: statistical optimization, characterization and pharmacokinetic assessmentInt J Pharm20134431–2 26 3823313344
  • GuptaAAggarwalGSinglaSAroraRTransfersomes: a novel vesicular carrier for enhanced transdermal delivery of sertraline: development, characterization, and performance evaluationSci Pharm2012804 1061 108023264950
  • NipunTSAshraful IslamSMSEDDS of gliclazide: preparation and characterization by in-vitro, ex-vivo and in-vivo techniquesSaudi Pharm J2014224 343 34825161379
  • TrompetaAFKokliotiMAPerivoliotisDKLynchICharitidisCATowards a holistic environmental impact assessment of carbon nanotube growth through chemical vapour depositionJ Clean Prod2016129 384 394
  • MoawadFAAliAASalemHFNanotransfersomes-loaded thermosensitive in situ gel as a rectal delivery system of tizanidine HCl: preparation, in vitro and in vivo performanceDrug Deliv2017241 252 26028156169
  • DietCBDietUDietNPReport of the American Institute of Nutrition Ad Hoc Committee on Standards for Nutritional StudiesJ Nutr1977107 1340874577
  • SinghRMKumarAPathakKThermally triggered mucoadhesive in situ gel of loratadine: β-cyclodextrin complex for nasal deliveryAAPS Pharm Sci Tech2013141 412 424
  • TuntiyasawasdikulSLimpongsaEJaipakdeeNSripanidkulchaiBA monolithic drug-in-adhesive patch of methoxyflavones from Kaempferia parviflora: in vitro and in vivo evaluationInt J Pharm20154782 486 49525482264
  • LopesLBScarpaMVSilvaGVRodriguesDCSantilliCVOliveiraAGStudies on the encapsulation of diclofenac in small unilamellar liposomes of soya phosphatidylcholineColloids Surf B Biointerfaces2004394 151 15815555896
  • DubeyVMishraDDuttaTNaharMSarafDKJainNKDermal and transdermal delivery of an anti-psoriatic agent via ethanolic liposomesJ Control Release20071232 148 15417884226
  • VermaPPathakKNanosized ethanolic vesicles loaded with econazole nitrate for the treatment of deep fungal infections through topical gel formulationNanomedicine201284 489 49621839053
  • TanrıverdiSTÖzerÖNovel topical formulations of Terbinafine-HCl for treatment of onychomycosisEur J Pharm Sci2013484–5 628 63623295582
  • AhadARaishMAl-MohizeaAMAl-JenoobiFIAlamMAEnhanced anti-inflammatory activity of carbopol loaded meloxicam nanoethosomes gelInt J Biol Macromol201467 99 10424657163
  • López-PintoJMGonzález-RodríguezMLRabascoAMEffect of cholesterol and ethanol on dermal delivery from DPPC liposomesInt J Pharm20052981 1 1215896932
  • GodinBTouitouEMechanism of bacitracin permeation enhancement through the skin and cellular membranes from an ethosomal carrierJ Control Release2004942–3 365 37914744487
  • ElsayedMMAbdallahOYNaggarVFKhalafallahNMLipid vesicles for skin delivery of drugs: reviewing three decades of researchInt J Pharm20073321–2 1 1617222523
  • FanCLiXZhouYEnhanced topical delivery of tetrandrine by ethosomes for treatment of arthritisBiomed Res Int20132013 16194324062995
  • ZhangYTShenLNWuZHZhaoJHFengNPComparison of ethosomes and liposomes for skin delivery of psoralen for psoriasis therapyInt J Pharm20144711–2 449 45224907596
  • YuXDuLLiYFuGJinYImproved anti-melanoma effect of a transdermal mitoxantrone ethosome gelBiomed Pharmacother201573 6 1126211575
  • ShenLNZhangYTWangQXuLFengNPEnhanced in vitro and in vivo skin deposition of apigenin delivered using ethosomesInt J Pharm20144601–2 280 28824269286
  • BendasERTadrosMIEnhanced transdermal delivery of salbutamol sulfate via ethosomesAAPS Pharm Sci Tech200784 E107
  • AbdelkaderHPierscionekBAlanyRGNovel in situ gelling ocular films for the opioid growth factor-receptor antagonist-naltrexone hydrochloride: fabrication, mechanical properties, mucoadhesion, tolerability and stability studiesInt J Pharm20144771–2 631 64225445974
  • HaniUShivakumarHDevelopment of miconazole nitrate thermosensitive bioadhesive vaignal gel for vaginal candidiasisAm J Adv Drug Deliv201313 358 368
  • BarakNBeckYAlbeckJHA randomized, double-blind, placebo-controlled pilot study of betahistine to counteract olanzapine-associated weight gainJ Clin Psychopharmacol2016363 253 25627028981
  • ZhengJVasselliJRKingJFUsing caenorhabditis elegans as a model for obesity pharmacology developmentAm J Ther2016236 e1363 e137024786852
  • ShumilovMTouitouEBuspirone transdermal administration for menopausal syndromes, in vitro and in animal model studiesInt J Pharm20103871–2 26 3319961912