Back to Journals » Drug Design, Development and Therapy » Volume 14

QbD-Based Investigation of Dermal Semisolid in situ Film-Forming Systems for Local Anaesthesia

Authors Kovács A, Kis N, Budai-Szűcs M, Gácsi A, Csányi E , Csóka I , Berkó S 

Received 23 September 2020

Accepted for publication 3 November 2020

Published 18 November 2020 Volume 2020:14 Pages 5059—5076

DOI https://doi.org/10.2147/DDDT.S279727

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 3

Editor who approved publication: Professor Anastasios Lymperopoulos



Anita Kovács, Nikolett Kis, Mária Budai-Szűcs, Attila Gácsi, Erzsébet Csányi, Ildikó Csóka, Szilvia Berkó

Institute of Pharmaceutical Technology and Regulatory Affairs, Faculty of Pharmacy, University of Szeged, Szeged 6720, Hungary

Correspondence: Szilvia Berkó
University of Szeged, Szeged 6720, Hungary
tpdel 44/ Tel/Fax +36-62-545-573
Email [email protected]

Purpose: The aim of our research work was to develop dermally applicable, lidocaine hydrochloride (LID-HCl)-containing semisolid in situ film-forming systems (FFSs) using the Quality by Design (QbD) approach to increase drug permeation into the skin.
Methods: Silicones were used to improve the properties of formulations and to increase the permeation through the skin. The QbD approach was applied to ensure quality-based development. With initial risk assessment, the critical material attributes (CMAs) and the critical process parameters (CPPs) were identified to ensure the required critical quality attributes (CQAs).
Results: During the initial risk assessment, four high-risk CQAs, namely in vitro drug release, in vitro drug permeation, drying properties, and mechanical properties, and three medium-risk CQAs, namely pH, viscosity, and film appearance were identified and investigated. Moreover, four high-risk CMAs were also considered during the formulation: permeation enhancing excipients, drying excipients, film-forming excipients, and emollients. During the experiments, LID-HCl influenced these critical parameters highly, thereby reducing the drying time. The formulation containing 25% silicone showed the best mechanical properties (49 mN skin adhesion, 20.3% film flexibility, 1.27 N film burst strength), which could predict better patient adherence. In addition, in vitro permeation studies showed that formulation containing 50% silicone has the fastest permeation rate. The flux of diffused API was 6.763 μg/cm2/h, which is much higher compared to the silicone-free formulation (1.5734 μg/cm2/h), and it can already be observed in the lower part of the dermis in 0.5 hour.
Conclusion: Our results show that LID-HCl has great influence on the critical parameters of FFSs. The silicone content can improve the applicability of formulations and has a favorable effect on the permeation rate of LID-HCl into the skin.

Keywords: in situ film-forming system, Quality by Design, formulation excipients, local anaesthesia

Introduction

Dermal local anaesthesia is a widely used method before a surgical procedure to decrease pain. Recently, research has focused on developing painless local anaesthetic treatment without needle insertion or injection. This non-invasive treatment can be used to decrease pain during a surgical intervention or chronic pain associated with neuralgia, arthritis or other severe medical diseases.1,2 Besides, it is an attractive alternative to injection in pediatrics.3

Lidocaine hydrochloride (LID-HCl) is one of the commonly used drugs during local anaesthesia.4 Lidocaine can relieve pain rapidly, but its effective time is short. Gels, sprays, creams, ointments, and patches are used as vehicles, but the barrier function of the skin limits the permeation of LID-HCl.5–7 It is a great challenge to reach the optimal permeation from these dermal preparations.8 The generally used dermal semisolid preparations, such as creams, ointments, and gels can be smeared and removed easily by movement and wetting.5 So a new drug delivery system may be required to enhance effectiveness and patient adherence.

The in situ film-forming system (FFS) is an innovative dermally applicable semisolid drug delivery system, which forms a film on the skin surface after application.9,10 FFSs contain non-volatile and volatile components. Non-volatile components include the active ingredients in matrix to provide the transport through the skin, while volatile components are responsible for film formation and enhanced drug permeation. Throughout the application process, volatile components evaporate, thereby an in situ formed film is left on the skin surface with a concentrated drug content, which promotes enhanced drug permeation.11 The application of the FFS is convenient, the appearance is attractive, and the film formed after fast drying has good mechanical properties. With optimal compositions, these advantages can be exploited,12 thus offering a promising choice in local anaesthesia.

To achieve optimal properties, volatile and non-volatile silicones are a good choice in FFSs.13–16 Silicones have a great effect on film-forming systems, for example, they ensure a “silky touch” feeling on the skin surface, have a softening feeling during the application, and they help the formulation to dry.17 The effects of silicones on critical attributes need to be considered during the formulation. Non-volatile silicones modify the mechanical attributes, thereby softer films can be formulated. Volatile silicones improve the film-forming ability, and more favorable drying time can be achieved.18 The optimal mechanical properties ensure that the films are retained on the skin surface for a longer time, thereby increasing exposure time. To improve patient adherence, films need to be removable painlessly, and the drying time needs to stay in a convenient range.

During the pharmaceutical development, it is essential to provide the appropriate quality of formulations. The International Council for Harmonization (ICH) guideline Q8 defines and sets the basis of the Quality by Design (QbD) approach.19 QbD is a systematic aspect, which ensures the optimal environment to reach the required properties. The method begins with identifying the quality target product profile (QTPP), then helps to understand the critical quality attributes (CQAs), organizes them and ensures the control of critical materials attributes (CMA) and critical process parameters (CPPs) based on the risk assessment.20–22 QbD tools are applied to examine the most critical parameters.23,24 These tools are summarized in the ICH guideline Q9.25

Based on our previous research, in situ FFSs could be a good choice to ensure painless local anaesthesia.18 The aim of this work was to develop LID-HCl-containing semisolid in situ FFSs using the QbD approach. The purpose of using QbD approach is to select the influencing attributes of the investigated system, which is essential for future development steps, such as defining product and process design space, and defining control strategy. Our study focuses on the formulation of three LID-HCl-containing compositions with or without silicones to investigate, on one hand, the effect of LID-HCl on the film-forming properties and on the other hand, the effect of formulations on the skin permeation of the active substance. The formulations were tested relative to each other and to blank FFSs.

Materials and Methods

Materials

Lidocaine hydrochloride was purchased from Hungaropharma Ltd. (Budapest, Hungary). Poly(vinyl alcohol) (87–90% hydrolyzed, average mol wt 30,000–70,000) was obtained from Sigma-Aldrich (Budapest, Hungary). Ethanol (96 per centum, Ph. Eur. 9.) was from Molar Chemicals Ltd. (Budapest, Hungary). Xantural ® 180 Xanthan Gum was a product sample from CP Kelco A Huber Company. ST – Cyclomethicone 5-NF, Dimethiconol Blend 20, ST Elastomer 10, and 7–3101 Elastomer Blend HIP Emulsion were kindly provided by Dow Corning (Midland, Michigan, USA). Purified and deionized water was used (Milli-Q system, Millipore, Milford, MA, USA). Methyl parahydroxybenzoate (Ph. Eur. 9.) was supplied by Molar Chemicals Ltd. (Budapest, Hungary). Salvequick® sticking plaster was from Orkla Care AB (Solna, Sweden). PVA-based artificial skin was obtained from © 2018 Tattoo machine Webshop (Mátészalka, Hungary). Cellulose acetate filter (Porafil membrane filter, cellulose acetate, pore diameter: 0.45 µm) was purchased from Macherey-Nagel GmbH & Co. KG (Düren, Germany).

Excised human skin was obtained from Caucasian female patient by routine plastic surgery procedure of the Department of Dermatology and Allergology, University of Szeged. Tissue samples to be destroyed are considered to be hazardous waste, so ethical permission is not required for research on these tissue samples in Hungary according to the Act CLIV of 1997 on Health Section 210/A. The local ethical committee (Regional Ethics Committee for Human Biomedical Research, University of Szeged) has to be informed about the in vitro skin penetration studies (Notification document number: 83/2008).

Methods

Quality by Design Methodology

Definition of TPP and QTPP

The definition of the Target Product Profile (TPP) is the first step during the product formulation. It defines the target of our work. After that, the Quality Target Product Profile (QTPP) summarizes the quality characteristics of the target product. It is the basis of design. QTPP parameters include, for example, therapeutic indication, route of administration, dosage form and strength, etc. according to the relevant guidelines.19,24,26

Definition of CQA, CMA, CPP

CQAs are derived from the QTPP. Based on prior knowledge, they potentially affect the quality of the product, for example, physical properties, solubility of active pharmaceutical ingredients (API) in the formulation, pH, viscosity, etc., which are clearly defined to provide the required product quality.19 After the determination of the quality attributes, the next step is to define the parameters which influence CQAs to provide the predefined quality. These are the critical material attributes (CMAs) and the critical process parameters (CPPs), which include properties of the API and excipients (eg, emollient, polymer), and properties of the applied process (eg, type of technology, homogenization time and rate).18,27

Risk Assessment: Quality Tools

Risk assessment is in the focus of the QbD approach. During risk management, the risks of the product quality are assessed, controlled, communicated, and reviewed. Quality tools such as the Ishikawa diagram, risk estimate matrix (REM) and Pareto analysis support and facilitate risk assessment. The Ishikawa diagram, which collects possible root causes and effects that have an influence on the quality. During the risk assessment, the next step was to select the critical parameters with Pareto analysis. This technique, which is also called ABC analysis, is used for the selection of risks that produce a significant effect. Categories A, B, and C of items have the highest, medium and the lowest influence on the quality of formulations, respectively. The REM presents the interdependence rating between QTPPs and quality attributes. Furthermore, the REM was used to define the connection between the quality attributes and CMAs, CPPs. The parameters were selected and the critical control points were defined with LeanQbD™ software (QbD Works LLC, Fremont, CA, USA).25,28

Preparation of FFSs

Three different types of formulations were examined. Blank formulations were also formulated and investigated for better understanding of the effect of LID-HCl on FFSs. The compositions and the functions of different ingredients can be found in Table 1. The blank film formulations without active substance (F1 - F3) and film formulations containing 5% LID-HC (LF1 - LF3) were prepared. The first type of formulations (F1 - LF1) did not contain silicones, while the others (F2 - LF2, F3 - LF3) contained different volatile and non-volatile components.

Table 1 Composition of Blank FFSs and FFSs Containing LID-HCl

ST – Cyclomethicone 5 – NF was the volatile silicone component to ensure the drying properties and to increase the permeation through the skin.18,29,30 Additional non-volatile silicones, Dimethiconol Blend 20, ST – Elastomer 10, and 7–3101 Elastomer Blend HIP Emulsion were added to F2, LF2 and F3, LF3, to modify film formation and rigidity of films.12 Furthermore, they modify the appearance of the film, provide a “silky touch” feeling, protect and soften the skin.17 F2, LF2 contained 25% of silicones, and F3, LF3 had 50% of silicones in the formulations. Volatile silicone containing was the same in these compositions. PVA and Xanthan gum also enhance film formation and have viscosity increasing properties.12,18 Additionally, ethanol is a drying excipient and a permeation enhancer excipient as well.30

LID-HCl was dissolved in Ethanol 96% and PVA was dissolved separately in purified water at 80°C under mixing. The two solutions were mixed at room temperature. Then, Xanthan gum was added to formulations F1, LF1 and F2, LF2 to ensure the required consistency. Finally, silicones were added slowly and mixed with a high shear mixer.

Characterization of the Critical Parameters of the Semisolid System

Investigation of Drying Properties

PVA-based artificial skin was used as model skin surface to detect the drying time of the preparations. When the formed film seemed to be dry, a microscope slide was used to touch the top of the film. If no marks were left on the slide, the film could be considered dry, and the elapsed time was the drying time. If the film left marks on the slide, the test was repeated until we got a dry film.18,31 Five parallel measurements were carried out. The results were calculated as means ± SD.

Investigation of pH

Each 10 g sample was placed in a beaker, and a Testo 206 pH meter (Testo SE & Co. KGaA, Lenzkirch, Germany) was used to measure the pH. Three parallel measurements were carried out.

Rheological Investigation

Rheological measurements were carried out with a Physica MCR101 rheometer (Anton Paar GmbH, Graz, Austria) to investigate the properties of FFSs before drying, and the effect of LID-HCl on the rheological parameters. Parallel plate geometry PP25 was applied with a measuring gap of 0.1 mm. The blank FFSs were also studied. Flow curves were obtained during the examination. The shear rate was raised from 0.1 to 100 1/s (up-curve) and then reduced from 100 to 0.1 1/s (down-curve) in CR mode. The shearing time was 300s and the measurements were made at 32°C.32

Characterization of the Critical Parameters of Films

Investigation of in vitro Drug Release

A Franz diffusion cell system (Logan Automated Dry heat sampling system) was applied to model drug release through a cellulose acetate synthetic membrane. Around 300 mg of each sample was used as the donor phase. The acceptor phase was thermostated phosphate buffer (PBS pH 7.4±0.15) at 32°C±0.5°C. Drug release lasted 8 hours (sampling times: 0.5; 1; 2; 3; 4; 5; 6; and 8 h). The concentration of the drug was examined by high-performance liquid chromatography (HPLC) (Shimadzu Nexera X2 UHPLC, Kyoto, Japan).

HPLC was equipped with a C18 reverse-phase column (ZORBAX Eclipse XDB-C18, Phenomenex, Torrance, CA, USA) with 5 µm, 4.6X150 mm dimension. The mobile phase was 0.1% phosphoric acid (solvent A):acetonitrile (solvent B) 90:10 in gradient mode. It was changed from 90:10 (A:B, v/v) to 40:60 (A:B, v/v) in 6 minutes then going back to 90:10 (v/v) between 6.1–10 minutes. The flow rate of 0.8 mL/min was set over 10 minutes, the column temperature and sample tray temperature were set to 25°C, and the detection was made at 230 nm. The injection volume was 5 µL. The time of analysis was 10 min, and the retention time was 4.2 min.32 Four parallel measurements were carried out. The results were calculated as means ± SD.

Investigation of in vitro Drug Permeation

Measurements of Drug Permeation with Franz Diffusion Cell System

A Franz diffusion cell system (Logan Automated Dry heat sampling system) was applied to model the permeation through ex vivo human heat-separated epidermis (HSE). The preparation of HSE was based on a procedure reported by Kligman and Christophers.33 Around 300 mg of each sample was used as the donor phase. The acceptor phase was thermostated phosphate buffer (PBS pH 7.4±0.15) at 32°C±0.5°C. Drug permeation through HSE lasted 24 hours (sampling times: 0.5; 1; 2; 4; 6; 12; 18; and 24 h). The concentration of the drug was examined by high-performance liquid chromatography (HPLC) (Shimadzu Nexera X2 UHPLC, Kyoto, Japan). The method of the HPLC measurement was the same as for the investigation of in vitro drug release. Four parallel examinations were carried out. The results were calculated as means ± SD.

Investigation of Drug Permeation with Raman Spectroscopy

Full thickness human subcutaneous fat free, abdominal skin was treated with LID-HCl-containing FFSs at room temperature. The skin sections were placed on filter papers that were soaked with phosphate-buffered saline solution. 200 mg of each preparation was placed on a 1x1 cm surface of the skin. The exposition time was 30 and 180 minutes. After the treatment, the films were pulled down and the residues were wiped. The treated skins were frozen and sectioned with a Leica CM1950 Cryostat (Leica Biosystems GmbH, Wetzlar, Germany). Aluminum-coated slides were used under the 15-μm-thick cross-sections. Raman spectroscopic analysis was carried out with a Thermo Fisher DXR Dispersive Raman Spectrometer (Thermo Fisher Scientific Inc., Waltham, MA, USA) equipped with a CCD camera and a diode laser operating at 780 nm. Microscopic lens with 50X magnification was used. Measurements were made with a laser power of 24 mW and a slit width of 25 μm. Skin mapping was captured of an area of 200X1,000 μm, with a step size of 50 μm vertically and horizontally. The OMNIC for Dispersive Raman 8.2 software package (Thermo Fisher Scientific) was used during instrument operation and evaluation. The untreated skin was also measured. The individual spectrum of each formulation was used as a reference when comparing the treated and non-treated skin.34

Investigation of the Mechanical Parameters of Films with Texture Analyzer

TA.XT plus Texture Analyzer (Stable Micro Systems Ltd, Vienna Court, Lammas Road, Godalming, Surrey, UK. GU7 1YL) was used to carry out the measurements. Skin adhesion, film burst strength and film flexibility were characterized with different accessories. Four parallel measurements were carried out during each examination. The results were calculated as means ± SD.

A 10x2 cm film was measured under the 90 Degree Peel Rig during the skin adhesion test to determine the mean peeling force (Pa) which can separate the film from the artificial skin surface. The applied test speed was 5 mm/s, while the distance was 50 mm. The mean peel force was averaged from 10 mm to 45 mm during the evaluation. Sticking plaster was used as a reference.18,35

Film Support Rig was used during the film burst strength and film flexibility tests. Burst strength (Pa) and resilience (%) were measured. Films with a diameter of 22 cm were made and placed into the film support rig. The compression mode was used for measuring film burst strength. Test speed, force, and target distance were 0.5 mm/sec, 100 g, and 1 mm, respectively. Similarly, the compression mode was applied for film flexibility with a test speed of 0.5 mm/sec, a force of 100 g and a distance of 1 mm. In both cases, human HSE was used as reference.18,36 The preparation of heat-separated epidermis was based on a procedure reported by Kligman and Christophers.33

Investigation of Film Appearance

There was placed 0.2 g of each formulation on a 3x3-cm surface of the PVA-based artificial skin. After drying, the appearance was investigated visually.18

Statistical Analysis

The one-way ANOVA analysis of variance (Dunnett) with GraphPad Prism 8 for Windows software (GraphPad Software Inc., La Jolla, CA, USA) was applied to make the statistical analysis of results. More parallel measurements ± SD were carried out. The differences were significant if p< 0.0001****p< 0.001***p< 0.01**p< 0.05* versus the control.18,24

Results and Discussion

Determination of QTPP and CQAs

As regards the TPP, the target was to formulate a semisolid, in situ film-forming system containing LID-HCl, with required properties for local anaesthetic therapy. Based on these, the QTPP of the FFS-containing LID-HCl encompasses therapeutic indication, route of administration, dosage form, dosage strength, site of activity, release profile, appearance of semisolid system, physical, chemical and microbiological stability, patient adherence increasing effect, mechanical properties of film and packaging material type. The CQAs originate from the QTPPs. In the present case, the composition is a semisolid in situ FFS, so the properties of both the semisolid compositions and the film must be considered. The Ishikawa diagram shows the parameters influencing the quality of the semisolid in situ film-forming system containing LID-HCl (Figure 1).25,37

Figure 1 Parameters influencing the quality of FFSs containing LID-HCl.

The quality attributes of the semisolid system include physical properties, solubility of API in formulation, viscosity, pH, moisturizing effect, and drying properties. The quality attributes of the formed film include film appearance, integrity, mechanical properties, in vitro drug release and drug permeation. Tables 2 and 3 present the targets and justifications of QTPPs and CQAs.

Table 2 QTPPs of the FFS Containing LID-HCl

Table 3 CQAs of the FFS Containing LID-HCl

Initial Risk Assessment

After defining the QTPPs and CQAs, the next step is to determine the CMAs and CPPs of the FFSs containing LID-HCl with risk assessment. Risk assessment refers to the estimate of the risks, related to the investigated semisolid and film systems. Table 4 shows the influencing factors as well as their impacts and occurrences on the target product.

Table 4 Summary of All the Parameters That Affect the FFS Containing LID-HCl

After that, risk assessment tools were applied to determine the connection between the parameters. Firstly, the parameters were identified, and then we performed risk assessment and ranked the parameters based on their risk. The risk estimate matrix (REM) was used to evaluate the relationships between CQAs and QTPPs (Table 5).

Table 5 REM of QTPP and CQA Parameters, Low = Low Risk, Medium = Medium Risk, High= High-Risk Parameters

A three-step scale was applied to rank the relationship between the CQA and QTTP parameters for the FFS containing LID-HCl, by assigning low (low-risk parameters), medium (medium risk parameters) and high (high-risk parameters) values to each of them. A 1(low)-3(medium)-9(high) scale was used for the probability rating.

A Pareto chart (Figure 2) was constructed based on the results of the REM to rank and show the severity scores of CQAs, and to highlight the highly influenced parameters of product quality. The results show that four quality attributes have the highest influence on the quality of the target product with the highest severity score (>400), called Category A. These are in vitro drug release, in vitro drug permeation, drying properties, and film mechanical properties. Category B (severity score: 250–400) includes viscosity, pH, and film appearance. The last category of severity score is Category C (below 250), which has the quality attributes with the lowest influence on the target product.

Figure 2 Pareto chart of CQA parameters; (A) high-risk parameters, (B) medium-risk parameters, (C) low-risk parameters.

The next REM (Table 6) was established to show the connection between CQAs and CPPs, CMAs. The evaluation was made by using the same scale (1-low, 3-medium, 9-high). Eleven CMA and CPP parameters were screened to determine their influence on the CQAs during the formulation of FFSs containing LID-HCl. The influencing material parameters were viscosity and permeation enhancing excipients, drying excipients, film-forming excipients, preservatives, and emollients.

Table 6 REM of CPPs/CMAs and CQA Parameters, Low = Low Risk, Medium = Medium Risk, High= High-Risk Parameters

Mixing rate, mixing time, type of mixer, temperature of technology and type of technology were found to be the influencing process parameters. Based on the REM results, a Pareto chart (Figure 3) was generated showing the severity scores of CQAs. The most critical parameters (Category A) influencing the quality of FFS-containing LID-HCl have the highest severity scores (>20,000). These are the following parameters: permeation enhancing, drying, film-forming excipients, and emollients. Category B includes viscosity enhancer excipients and type of technology (severity scores are 10,000–20,000), these parameters have medium influence on the final product’s quality. Category C has a low impact on the quality of the FFS during the development (>10,000): mixing rate, temperature of technology, mixing time, type of mixer, and preservatives belong here.

Figure 3 Pareto chart of CPP and CMA parameters; (A) high-risk parameters, (B) medium-risk parameters, (C) low-risk parameters.

According to the initial risk assessment, four highly influencing CQA parameters were identified, namely in vitro drug release, in vitro drug permeation, drying properties, and film mechanical properties. Moreover, three medium critical CQA parameters were also defined: viscosity, pH, and film appearance. These were the investigated parameters during the research work. The initial risk assessment also showed that the following four material parameters of the defined CPPs and CMAs had the highest influence on the target product: permeation enhancing excipients, drying excipients, film-forming excipients, and emollients. These parameters were considered during the development.

Figure 4 shows the result of the risk assessment during the formulation of FFSs containing LID-HCl based on the QbD approach.

Figure 4 Process of research work.

Results of the Critical Parameters of the Semisolid System

Drying Properties

As our previous work described, silicone content significantly decreases drying time.18 Additionally, a further examination was carried out to observe the effect of LID-HCl on this parameter. LID-HCl significantly decreased the drying time of all compositions, so it has favorable effect on the formulations. It was the most noticeable in the case of formulation LF2, where the values dropped from 10 minutes to 8.35 minutes (Figure 5). Formulations LF2 and LF3 met the requirement (10 minutes).

Figure 5 Comparison of the drying times of blank and LID-HCl-containing formulations; Mean ± SD (n=5), (p < 0.05 * and p < 0.0001 **** vs blank formulation).

Results of pH of FFSs

The pH is an important attribute to provide safety and efficacy of use, and the stability of formulations. The results showed that all the formulations were in the optimal range of pH, thereby these are appropriate for dermal application. The LID-HCl content decreased the pH of the formulations. The values are presented in Table 7. However, the LID-HCl is a weak base, with 7.75 pKa. Below this pKa the ionized form of the drug is higher, than the base form, which predicts lower permeation rate through the SC, so the permeation enhancer silicones could be favorable for increasing permeability of SC.48,49

Table 7 pH Data of FFSs

Rheological Properties of FFSs

The three different types of formulations showed different rheological properties. In the flow curves, thixotropy was observed in all cases. Thixotropy was the lowest in the case of F1 and LF1. Based on the flow curves (Figure 6), both the LID-HCl content and the silicon amount increased the shear stress of the systems.

Figure 6 Flow curves of FFSs.

The viscosity value of F1 (608 mPas) and LF1 (718 mPas) was the lowest at 100 1/s at 32°C. F2 was 2070 mPas and LF2 was 2560 mPas. The highest viscosity was detected for F3 with 3540 mPas and for LF3 with 4770 mPas. The rheological measurements showed that the formulations met the requirements (Table 3), they are suitable for dermal use.

Results of the Critical Parameters of Films

In vitro Drug Release Studies

The in vitro drug release study was one of the most critical parameters in the case of risk assessment (Figure 3). The measurement was performed to examine the release profile of the active ingredient from the formulations. The release of LID-HCl was measured through a synthetic membrane, which was an artificial mixed cellulose ester membrane, for 8 hours (Figure 7). According to our target (Table 3), 30% of the API had to be released in 0.5 h. The results showed that the diffused API was lower than 30% in 0.5 hour but more than 30% of LID-HCl was released in the case of LF2 and LF3 after 1 hour. After 3 hours, the diffused amount of API was more than 60% from all formulations (Table 8).

Table 8 Mean Cumulative Amount of Diffused API (%) ± SD (n=4) After 0.5; 1 and 3 Hours, (p < 0.05 *)

Figure 7 In vitro drug release through cellulose acetate membrane; Mean ± SD (n=4), (p < 0.05 *, LF3 vs LF1 and LF2).

In the case of LF3, the diffusion of API significantly increased in 0.5 hour. Later, the release profiles through this synthetic membrane were similar in all cases, so the different excipients have a similar effect on the drug release. However, the API released form the LF3 formulation faster, than from the other formulations, so it could be explained by the favourable effect of the 7–3101 Elastomer Blend HIP Emulsion in the composition.

In vitro Drug Permeation Studies

The in vitro drug permeation study consisted of two measurements. The permeation of LID-HCl was measured through HSE during 24 h in a Franz cell and completed with Raman spectroscopy, where the location of the API was detected in the skin layers.

Then, the mathematical evaluation was carried out. The rate of the permeation (flux) was calculated from the slope of the cumulative amounts of LID-HCl (µg/cm2) permeated versus time (h) profiles (Table 9). LF1 and LF2 contained volatile silicone to increase the permeation rate through the skin. The results showed that ST-Cyclomethicone 5NF increased the permeation rate compared to LF1. In addition, the amount of volatile silicone was the same in both formulations, but the highest permeation rate was detected in the case of LF3. This could be explained by the additional effect of 7–3101 Elastomer Blend HIP Emulsion, and the fast drying of the formulation with prolonged drying mechanism.18

Table 9 Fluxes J (µg/cm2/h) of Diffused API

The in vitro skin permeation studies were also carried out with Raman spectroscopy. The correlation maps were established, which showed the distribution of LID-HCl-containing FFSs by using the appropriate spectra that fit to the treated skin spectra. The spectra of LID-HCl, blank formulations, and API-containing formulations were recorded. In this case, the spectra of API-containing formulations were used to make the distribution correlation maps, because these spectra indicate the presence of LID-HCl. The spectra of pure API could not be used because of the low intensity of LID-HCl in the formulations. A non-treated correlation map was used as a control during the evaluation. Figure 8 shows the recorded correlation maps of LF1 (A), LF2 (B), LF3 (C). The warmer color indicates a higher amount of the preparations in the different skin layers.

Figure 8 Raman correlation maps for the distribution of FFSs of (A) LF1, (B) LF2, and (C) LF3.

The results showed that the LID-HCl-containing preparations were present in the different regions of the human skin. Formulation LF1 was found in the upper region of the human skin and formulation LF2 permeated into the epidermis and the upper part of the dermis after 3 hours. Formulation LF3 could already be observed in the lower part of the dermis after 0.5 hour. After 3 h the distribution of the formulation could be found in all skin layers as well.

Based on the results, the LF3 showed the fastest permeation rate, so the results of Raman mapping correlated with the measurements of the in vitro permeation study in a Franz cell.

Mechanical Properties of Films with Texture Analyzer

According to the risk assessment, the film mechanical properties are also critical parameters, which give important information about the usability of formulations (Figure 2). The optimal mechanical properties ensure patient adherence as well as long and durable application. Skin adhesion is required to increase the exposure time of API; however, if the skin adhesion value is too high, the removal of the film can be painful for the patients. Flexibility ensures that the film can imitate the movement of the skin, which is also required for durable use. If film burst strength is high, the structure of the film can be too rigid, but if the value is low, the film is not able to keep its integrity. During the measurements, we used sticking plaster as reference in the case of skin adhesion, and heat-separated epidermis in the case of flexibility and film burst strength.

In our previous study, it was obtained, that silicone content makes the films softer but less resistant.18 In this research, we examined the effect of API on the FFSs. As the results show (Figure 9A), skin adhesion highly increased due to the LID-HCl content in formulations LF1 and LF2. When these preparations were used, the parameters met our targets (Table 3). In the case LF3 the adhesion parameter was lower than expected.

Figure 9 Mechanical properties of FFSs: (A) Skin adhesion, (B) film flexibility, (C) film burst strength; Mean ± SD (n=4), (p < 0.05 *, p < 0.01 **, p < 0.001 ***, and p < 0.0001 **** vs blank formulation).

The flexibility of films can be seen in Figure 9B. LF1 (27.2%) and LF2 (38.3%) met the requirement, while LF3 (20.3%) showed lower flexibility. In the case of LF1 and LF3, the API decreased the flexibility of the film; however, the flexibility of LF2 increased.

Film burst strength showed a significant increase when the formulation contained LID-HCl (Figure 9C). The burst strength of LF1 jumped from 4.91 N to 8.12 N, and thus its values were too high for our target. The other two films, LF2 (2.96 N) and LF3 (1.27 N) had optimal properties.

In conclusion, the API increased skin adhesion and film burst strength. The film flexibility of LF2 increased, while the flexibility of LF1 and LF3 decreased due to the API content. Based on the results, LF2 had the best mechanical properties, which could predict higher effect and better patient adherence.

Appearance of Films

The film appearance met our requirements (Table 3). After drying, translucent, slightly white, homogenous, and compact films were formed, which are appropriate for convenient application. There were no differences between the films formed by blank or LID-HCl-containing formulations.18

Conclusion

In this research work semisolid in situ FFSs containing LID-HCl were formulated using the QbD approach. Initial risk assessment was applied to evaluate the critical parameters during the development. Based on the results of initial risk assessment, three compositions with or without API were formed and investigated. Initial risk assessment defined the CMAs and the CPPs to ensure the required CQAs. During the initial risk assessment, four CQAs, namely in vitro drug release, in vitro drug permeation, drying properties, and film mechanical properties were found to be highly critical attributes, and three CQAs, namely viscosity, pH, and film appearance were found to be medium critical attributes in the formulation of FFSs. These parameters were investigated. Furthermore, the effects of LID-HCl on these parameters were studied. The initial risk assessment also revealed that four factors, namely permeation enhancing excipients, drying excipients, film-forming excipients, and emollients were highly critical parameters for the CQAs.

The results of in vitro drug release showed, there were no significant differences between the diffusion of the formulations after 3 hours, so the formulations have the same drug release profile. However, the LF3 showed significant increase in the diffusion of API in 0.5 hour, so it was released from the LF3 formulation faster, than from the other formulations. The results of in vitro permeation through the HSE showed that the silicone content increased the permeation rate through the skin. The LF3 formulation had the highest permeation rate, which could be explained by the additional effect of non-volatile silicone in the composition, and the fast drying of the formulation with prolonged drying mechanism.18 The measurements of Raman spectroscopy revealed that the LID-HCl-containing preparations were present in the different regions of the human skin. LF3 showed deeper permeation into the skin and the rate of permeation was higher. The measurements of drying properties showed that LID-HCl decreased the drying time significantly. Mechanical properties give important information about the usability of films. Silicone content makes the films softer and less resistant; however, the API content modifies the mechanical properties of FFSs. The API increased the skin adhesion and film burst strength, and in the case of LF2, the film flexibility increased, too. Based on these, the formulation containing 25% silicone (LF2) showed the best mechanical properties, which could predict better patient adherence. In respect of pH, rheological parameters, and film appearance, all the three formulations met our requirement. Table 10 summarizes the applicability of FFSs considering our initial expectations (Tables 2 and 3).

Table 10 Summary of Applicability of FFSs Containing LID-HCl: Optimal: Exceptionally Good Result, Adequate: The Result Meets Our Requirement of Table 3, Inadequate: The Result Does Not Meet the Requirement of Table 3

In summary, LID-HCl has high influencing effect on the formulation properties. The silicone content has improved the effect on the permeation rate of API through the skin. Based on our results, formulations F2-LF2 have good mechanical properties, but the permeation rate need to be increased with addition of other permeation enhancers, and F3 - LF3 are appropriate delivery systems for LID-HCl, but the mechanical properties of film are required to improve.

Funding

This research was funded by the Gedeon Richter’s Talentum Foundation (Gyömrői Street 19-21), Budapest, Hungary, 1103) and project EFOP 3.6.3-VEKOP-16-2017-00009. The article publishing charge was funded by The University of Szeged Open Access Fund (FundRef, Grant No. 4943).

Disclosure

The authors report no conflicts of interest in this work.

References

1. Lin H, Zhang Y, Hou M, Wang Y. Transdermal local anaesthetic films of tetracaine in comparison to the conventional gel in effective management of pain. Int J Pharmacol. 2016;12(6):669–674. doi:10.3923/ijp.2016.669.674

2. Uziel Y, Berkovitch M, Gazarian M, et al. Evaluation of eutectic lidocaine/prilocaine cream (EMLA®) for steroid joint injection in children with juvenile rheumatoid arthritis: a double blind, randomized, placebo controlled trial. J Rheumatol. 2003;30:594–596.

3. Houck CS, Sethna NF. Transdermal analgesia with local anesthetics in children: review, update and future directions. Expert Rev Neurother. 2005;5(5):625–634. doi:10.1586/14737175.5.5.625

4. Chun M-K, Hossain M, Choi S-H, Ban S-J, Moon H, Choi H-K. Development of cataplasmic transdermal drug delivery system containing eutectic mixture of lidocaine and prilocaine. J Pharm Invest. 2012;42:139–146. doi:10.1007/s40005-012-0023-6

5. Omar MM, Hasan OA, El Sisi AM. Preparation and optimization of lidocaine transferosomal gel containing permeation enhancers: a promising approach for enhancement of skin permeation. IJN. 2019;14:1551–1562. doi:10.2147/IJN.S201356

6. Shipton EA. New delivery systems for local anaesthetics—Part 2. Eisenkraft JB, ed. Anesthesiol Res Pract. 2011;2012:289373. doi:10.1155/2012/289373

7. Alster T. Review of lidocaine/tetracaine cream as a topical anesthetic for dermatologic laser procedures. Pain Ther. 2013;2(1):11–19. doi:10.1007/s40122-013-0010-2

8. Topical Anesthesia: Overview, Mechanism of Action, Absorption. Available from: https://reference.medscape.com/article/109673-overview. Accessed October 23, 2020.

9. Tran TTD, Tran PHL. Controlled release film forming systems in drug delivery: the potential for efficient drug delivery. Pharmaceutics. 2019;11(6):290. doi:10.3390/pharmaceutics11060290

10. Zurdo Schröder I. Film forming polymeric solutions as drug delivery systems for the skin. 2007. doi:10.22028/D291-22361.

11. McAuley WJ, Caserta F. Film-Forming and Heated Systems. In: Donnelly RF, Singh TRR, eds. Novel Delivery Systems for Transdermal and Intradermal Drug Delivery. Chichester, UK: John Wiley & Sons, Ltd; 2015:97–124. doi:10.1002/9781118734506.ch5

12. Kathe K, Kathpalia H. Film forming systems for topical and transdermal drug delivery. Asian J Pharm Sci. 2017;12(6):487–497. doi:10.1016/j.ajps.2017.07.004

13. Houben R Silicone crosspolymers: customising sensory feel. . 2014paper/Silicone-crosspolymers%3A-customising-sensory-feel-Houben/f99423c80c3207568d9ecf6b878bd67541f93d20. Available from: Accessed October 19, 2020.

14. Aliyar H, Schalau G. Recent developments in silicones for topical and transdermal drug delivery. Ther Deliv. 2015;6(7):827–839. doi:10.4155/tde.15.39

15. Aliyar H, Huber R, Loubert G, Schalau G. Efficient ibuprofen delivery from anhydrous semisolid formulation based on a novel cross-linked silicone polymer network: an in vitro and in vivo study. J Pharm Sci. 2014;103(7):2005–2011. doi:10.1002/jps.23990

16. Pieńkowska K. Review of Current Pharmaceutical Applications of Polysiloxanes (Silicones). In: Thakur VK, Thakur MK, eds. Handbook of Polymers for Pharmaceutical Technologies. Vol 2. Hoboken: John Wiley & Sons, Inc.; 2015:363–381. doi:10.1002/9781119041412.ch13

17. DiSapio AJ Silicones as alternatives to hydrocarbons in personal care formulations. Available from: https://www.yumpu.com/en/document/read/30821322/silicones-as-alternatives-to-hydrocarbons-in-personal-care-. Accessed November 3, 2020.

18. Kis N, Kovács A, Budai-Szűcs M, et al. Investigation of silicone-containing semisolid in situ film-forming systems using QbD tools. Pharmaceutics. 2019;11(12):660. doi:10.3390/pharmaceutics11120660

19. European Medicines Agency. ICH guideline Q8 (R2) on pharmaceutical development. Available from: https://www.ema.europa.eu/en/documents/scientific-guideline/international-conference-harmonisation-technical-requirements-registration-pharmaceuticals-human-use_en-11.pdf. Accessed November 3, 2020.

20. Visser JC, Dohmen WMC, Hinrichs WLJ, Breitkreutz J, Frijlink HW, Woerdenbag HJ. Quality by design approach for optimizing the formulation and physical properties of extemporaneously prepared orodispersible films. Int J Pharm. 2015;485(1–2):70–76. doi:10.1016/j.ijpharm.2015.03.005

21. Yu LX. Pharmaceutical quality by design: product and process development, understanding, and control. Pharm Res. 2008;25(4):781–791. doi:10.1007/s11095-007-9511-1

22. Gyulai O, Kovács A, Sovány T, Csóka I, Aigner Z. Optimization of the critical parameters of the spherical agglomeration crystallization method by the application of the quality by design approach. Materials. 2018;11(4):635. doi:10.3390/ma11040635

23. Radhakrishnan V, Davis P, Hiebert D. Scientific approaches for the application of QbD principles in lyophilization process development. In: Warne NW, Mahler H-C, editors. Challenges in Protein Product Development. Vol 38. AAPS Advances in the Pharmaceutical Sciences Series. Cham: Springer International Publishing; 2018:441–471. doi:10.1007/978-3-319-90603-4_20

24. Bakonyi M, Berkó S, Kovács A, et al. Application of quality by design principles in the development and evaluation of semisolid drug carrier systems for the transdermal delivery of lidocaine. J Drug Deliv Sci Technol. 2018;44:136–145. doi:10.1016/j.jddst.2017.12.001

25. European Medicines Agency. ICH guideline Q9 on quality risk management. Available from: https://www.ema.europa.eu/en/documents/scientific-guideline/international-conference-harmonisation-technical-requirements-registration-pharmaceuticals-human-use_en-3.pdf. Accessed November 3, 2020.

26. Yu LX, Amidon G, Khan MA, et al. Understanding pharmaceutical quality by design. AAPS J. 2014;16(4):771–783. doi:10.1208/s12248-014-9598-3

27. Nadpara NP, Thumar RV, Kalola VN, Patel PB Quality by design (QBD): a complete review 9.

28. Kovács A, Berkó S, Csányi E, Csóka I. Development of nanostructured lipid carriers containing salicyclic acid for dermal use based on the Quality by Design method. Eur J Pharm Sci. 2017;99:246–257. doi:10.1016/j.ejps.2016.12.020

29. DUPONT Transportation & Industrial. Dow Corning® ST-Cyclomethicone 5-NF. Available from: https://www.dupont.com/products/DowCorningSTCyclomethicone5NF.html. Accessed May 26, 2020.

30. Ahad A, Aqil M, Kohli K, et al. Chemical penetration enhancers: a patent review. Expert Opin Ther Pat. 2009;19(7):969–988. doi:10.1517/13543770902989983

31. Saravanan J, Elango K, Chellakumari SD, Arulkumar K. Formulation and evaluation of film forming gel of diclofenac diethylamine. World J Pharm Pharm Sci. 7(6):12. doi:10.20959/wjpps20186-11757

32. Zsikó S, Cutcher K, Kovács A, et al. Nanostructured lipid carrier gel for the dermal application of lidocaine: comparison of skin penetration testing methods. Pharmaceutics. 2019;11:7. doi:10.3390/pharmaceutics11070310

33. Kligman AM. Preparation of isolated sheets of human stratum corneum. Arch Dermatol. 1963;88(6):702. doi:10.1001/archderm.1963.01590240026005

34. Bakonyi M, Gácsi A, Kovács A, M-B S, Berkó S, Csányi E. Following-up skin penetration of lidocaine from different vehicles by Raman spectroscopic mapping. J Pharm Biomed Anal. 2018;154:1–6. doi:10.1016/j.jpba.2018.02.056

35. Professionals TA. Texture analysis professionals blog: texture analysis in action: peel testing. Texture Analysis Professionals Blog. 2015. Available from: https://textureanalysisprofessionals.blogspot.com/2015/10/texture-analysis-in-action-peel-testing.html. Accessed May 26, 2020.

36. Texture Analysis Professionals Blog: Texture Analysis in Action: the Film Support Rig. https://textureanalysisprofessionals.blogspot.com/2015/02/texture-analysis-in-action-film-support.html. Accessed May. 26, 2020.

37. Singh L, Sharma V. Quality by Design (QbD) approach in pharmaceuticals: status, challenges and next steps. DDL. 2015;5(1):2–8. doi:10.2174/2210303104666141112220253

38. Kumar M, Chawla R, Goyal M. Topical anesthesia. J Anaesthesiol Clin Pharmacol. 2015;31(4):450–456. doi:10.4103/0970-9185.16\9049

39. Paudel KS, Milewski M, Swadley CL, Brogden NK, Ghosh P, Stinchcomb AL. Challenges and opportunities in dermal/transdermal delivery. Ther Deliv. 2010;1(1):109–131. doi:10.4155/tde.10.16

40. Ngo HV, Tran PHL, Lee B-J, Tran TTD. Development of film-forming gel containing nanoparticles for transdermal drug delivery. Nanotechnology. 2019;30(41):415102. doi:10.1088/1361-6528/ab2e29

41. Weilbach C, Hoppe C, Karst M, et al. Effectiveness of various formulations of local anesthetics and additives for topical anesthesia – a prospective, randomized, double-blind, placebo-controlled study. JPR. 2017;10:1105–1109. doi:10.2147/JPR.S131029

42. Klykken P, Servinski M, Thomas X. Silicone film-forming technologies for health care applications. 2009. Available from: /paper/Silicone-Film-Forming-Technologies-for-Health-Care-Klykken-Servinski/53f4f961044f0a08c2425d187f1a08eba874f124. Accessed October 23, 2020.

43. Savjani KT, Gajjar AK, Savjani JK. Drug solubility: importance and enhancement techniques. ISRN Pharm. 2012;2012:1–10. doi:10.5402/2012/195727

44. Lambers H, Piessens S, Bloem A, Pronk H, Finkel P. Natural skin surface pH is on average below 5, which is beneficial for its resident flora. Int J Cosmet Sci. 2006;28(5):359–370. doi:10.1111/j.1467-2494.2006.00344.x

45. Ali S, Yosipovitch G. Skin pH: From basic science to basic skin care. Acta Derm Venerol. 2013;93(3):261–267. doi:10.2340/00015555-1531

46. Shah DK, Khandavilli S, Panchagnula R. Alteration of skin hydration and its barrier function by vehicle and permeation enhancers: a study using TGA, FTIR, TEWL and drug permeation as markers. Methods Find Exp Clin Pharmacol. 2008;30(7):499. doi:10.1358/mf.2008.30.7.1159653

47. Berkó S, Zsikó S, Deák G, et al. Papaverine hydrochloride containing nanostructured lyotropic liquid crystal formulation as a potential drug delivery system for the treatment of erectile dysfunction. DDDT. 2018;12:2923–2931. doi:10.2147/DDDT.S168218

48. PubChem. Lidocaine hydrochloride. 2009. Available from: https://pubchem.ncbi.nlm.nih.gov/compound/6314. Accessed November 3, 2020.

49. Østergaard Knudsen N, Pommergaard Pedersen G. pH and Drug Delivery. In: Surber C, ed. Current Problems in Dermatology. Vol 54. Basel: Karger; 2018:143–151. doi:10.1159/000489528

Creative Commons License © 2020 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, v3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.