Department of Pharmaceutics, University Department of Chemical Technology Chatrapati Sambhajinagar, Maharashtra, India
Dexlansoprazole, a proton pump inhibitor widely used in the management of gastroesophageal reflux disease (GERD), belongs to the Biopharmaceutical Classification System (BCS) Class II, characterized by low aqueous solubility and high permeability. Its poor solubility and acid-labile nature significantly limit oral bioavailability and therapeutic effectiveness. The present study was aimed at the formulation and evaluation of Dexlansoprazole-loaded solid lipid nanoparticles (SLNs) to enhance solubility, stability, and dissolution performance. SLNs were prepared using a modified single-emulsion solvent evaporation method assisted by probe sonication. Glyceryl monostearate (GMS) was used as the lipid matrix, Tween 80 as the surfactant, and dimethylformamide as the organic solvent. Optimization of formulation variables was carried out using a factorial design approach to achieve minimal particle size and maximum entrapment efficiency. The optimized formulation exhibited a mean particle size of approximately 156 nm with a narrow polydispersity index, indicating uniform nanoparticle distribution. Fourier Transform Infrared (FTIR) analysis confirmed the absence of chemical interactions between Dexlansoprazole and formulation excipients, indicating physical entrapment of the drug. Scanning Electron Microscopy (SEM) revealed spherical nanoparticles with smooth surface morphology. Entrapment efficiency was found to be significantly high, demonstrating effective drug incorporation within the lipid matrix. In-vitro drug release studies showed a sustained release profile compared to pure Dexlansoprazole, suggesting improved dissolution characteristics. Stability studies conducted at refrigerated conditions confirmed the physical stability of the SLNs over the study period. Overall, the results indicate that solid lipid nanoparticles represent a promising carrier system for improving the solubility and oral delivery of Dexlansoprazole.
Poor aqueous solubility remains one of the most critical challenges in modern drug development, particularly for drugs belonging to the Biopharmaceutical Classification System (BCS) Class II, where solubility becomes the rate-limiting step for oral absorption. Approximately 40–60% of newly synthesized drug molecules exhibit poor water solubility, which directly impacts their oral bioavailability, therapeutic efficacy, and clinical translation. Dexlansoprazole, an acid-labile proton pump inhibitor (PPI) used for the treatment of gastroesophageal reflux disease (GERD), erosive esophagitis, and related acid-peptic disorders, is one such drug whose low solubility and poor stability in acidic medium limit its systemic exposure and overall therapeutic performance. Conventional approaches such as enteric-coated granules or delayed-release capsules have made significant strides in improving the pharmacokinetic behavior of Dexlansoprazole; however, they often suffer from issues such as inconsistent absorption, variable bioavailability, and limited dissolution in the intestinal environment. Therefore, advanced formulation strategies that can enhance the solubility, protect the drug from acidic degradation, and improve absorption are of paramount importance. Nanotechnology has emerged as a revolutionary tool in pharmaceutical sciences, particularly for formulating poorly water-soluble drugs. Among various nanocarrier systems, Solid Lipid Nanoparticles (SLNs) have attracted immense attention owing to their excellent biocompatibility, ability to encapsulate both hydrophilic and lipophilic molecules, controlled and sustained release behavior, and stability in physiological environments. SLNs are submicron colloidal carriers composed of physiological lipids that remain solid at both room and body temperatures. They are stabilized by surfactants such as Tween 80, Poloxamers, or phospholipids, enabling the formation of a stable dispersion. The high drug-loading capacity, protection of sensitive drug molecules from environmental degradation, improved solubility, and relatively simple manufacturing processes make SLNs an attractive alternative to conventional drug delivery systems such as emulsions, liposomes, and polymeric nanoparticles [4–7]. Dexlansoprazole is the R-enantiomer of lansoprazole and exhibits greater systemic exposure, longer residence time, and superior pharmacodynamic profile compared to its racemic counterpart. Despite these advantages, the drug suffers from extremely low aqueous solubility (~0.1 mg/mL at neutral pH), acid-labile nature, pH-dependent dissolution, and incomplete intestinal absorption. To address these limitations, Dexlansoprazole is commercially formulated using a dual delayed-release capsule technology wherein two types of enteric-coated granules dissolve at different intestinal pH values. This provides a biphasic drug release and prolonged plasma exposure [31–33]. While this system offers improved acid protection and extended action, challenges such as dose-related variability, delayed onset of therapeutic effect, and dissolution-limited absorption persist. Solid Lipid Nanoparticles provide an innovative drug delivery approach for Dexlansoprazole, offering crucial advantages beyond those of enteric-coated or conventional delayed-release formulations. By entrapping Dexlansoprazole within a solid lipid matrix such as Glyceryl Monostearate (GMS), the drug can be shielded from acidic degradation, improving its stability in gastric pH. The nanoscale size of SLNs significantly enhances surface area, facilitating improved dissolution and absorption across the gastrointestinal epithelium. Additionally, the lipid core slows down drug release, providing a sustained effect and potentially reducing dosing frequency. Surfactants such as Tween 80 not only stabilize the SLNs but also enhance solubility and modify membrane permeability, contributing to improved oral bioavailability [8–12]. The preparation of SLNs traditionally involves techniques such as high-pressure homogenization, solvent emulsification-diffusion, microemulsion techniques, ultrasonication, and spray drying. Among these, ultrasonication-assisted solvent evaporation stands out due to its simplicity, reproducibility, cost-effectiveness, and ability to produce nanocarriers with narrow particle size distribution without the need for high temperatures or harsh organic solvents [25–28]. In the present study, Dexlansoprazole-loaded SLNs were prepared using a modified single emulsification–solvent evaporation method assisted by probe sonication. GMS was used as the lipid matrix, Tween 80 as the surfactant, and dimethylformamide (DMF) as the organic solvent. During formulation, the lipid was melted, the drug was dissolved in DMF, and the aqueous phase containing Tween 80 was added dropwise to form a coarse emulsion. Ultrasonication was then applied to reduce particle size, and solvent traces were removed via rotary evaporation. This method allowed efficient entrapment of the drug in the lipid matrix while ensuring stability of the nanoparticulate dispersion. One of the critical aspects of SLN formulation is the evaluation of compatibility between the drug and excipients. Fourier Transform Infrared Spectroscopy (FTIR) serves as an essential tool for confirming the presence of characteristic functional groups and identifying potential interactions. FTIR studies conducted in the present research revealed the absence of significant drug–excipient interactions, as the characteristic peaks of Dexlansoprazole were retained in the SLN formulation. This confirms that the drug was physically entrapped without undergoing chemical modification. Moreover, particle size, polydispersity index (PDI), and zeta potential were assessed using Dynamic Light Scattering (DLS). The optimized formulation achieved a particle size of 156 nm with a low PDI, indicating a uniform and stable dispersion. Zeta potential measurements further confirmed the stability of the nanoparticles, as adequate charge repulsion minimizes aggregation. Entrapment efficiency and drug loading capacity are crucial parameters for evaluating the performance of SLNs. In the current work, formulations with optimized lipid-to-drug ratios demonstrated significantly higher entrapment efficiency compared to initial experimental batches, confirming the role of lipid and surfactant concentrations in determining the encapsulation potential of SLNs. In vitro release studies revealed a sustained drug-release pattern, which can greatly contribute to prolonged therapeutic efficacy of acid-labile drugs like Dexlansoprazole. Stability studies conducted at 4 °C further confirmed that the optimized formulation maintained its physicochemical integrity over the study period, demonstrating the suitability of SLNs for long-term storage and therapeutic use. The rising prevalence of GERD and other acid-related disorders further underscores the need for improved therapeutic formulations. Epidemiological studies across Asian populations indicate that the prevalence of GERD has nearly doubled in recent years, attributed largely to lifestyle modifications, dietary changes, and increased diagnostic awareness [29,30]. PPIs remain the cornerstone of GERD management, but therapeutic failures are not uncommon due to factors such as rapid metabolism, inconsistent absorption, and poor solubility of specific PPIs including Dexlansoprazole. Therefore, innovative formulation strategies such as SLNs provide a promising alternative to conventional oral drug delivery systems. SLNs offer multiple advantages in the context of Dexlansoprazole delivery. The lipid matrix not only enhances solubility and dissolution but also protects the drug from enzymatic and pH-induced degradation. The small particle size facilitates improved surface area, resulting in enhanced solubilization and absorption. Furthermore, the ability of SLNs to modulate drug release helps in maintaining therapeutic plasma levels for prolonged durations, reducing the dosing frequency and improving patient compliance. The use of physiological lipids such as GMS ensures biocompatibility, while surfactants like Tween 80 improve dispersion stability and prevent particle aggregation. Additionally, lipid-based nanocarriers are known to enhance lymphatic uptake, which may further improve the bioavailability of poorly soluble drugs [19]. Literature evidence strongly supports the use of SLNs for improving solubility and bioavailability of several water-insoluble drugs. Studies on PPIs such as omeprazole and lansoprazole have shown significant improvements in dissolution rate, stability, and pharmacokinetic behavior when formulated using SLNs. While extensive research exists for PPIs in nanoformulations, Dexlansoprazole-loaded SLNs remain largely unexplored, presenting a significant opportunity to investigate their potential for enhancing solubility and therapeutic efficacy. The present research addresses this gap by developing and evaluating Dexlansoprazole-loaded SLNs with optimized particle size, entrapment efficiency, stability, and dissolution characteristics [20]. An additional strength of the present study lies in the use of a factorial design approach for optimization. Statistical experimental design methods such as factorial design help in evaluating the effect of multiple formulation variables simultaneously while minimizing the number of experimental trials required. Factors such as lipid concentration, surfactant concentration, and sonication time were systematically varied to assess their impact on the particle size of SLNs. This approach allowed identification of the optimal formulation conditions that produced SLNs with desirable physicochemical properties. Mathematical modeling and response surface analysis generated during this process serve to improve understanding of interaction effects among variables, facilitating reproducible and efficient formulation development [21]. Evaluation of the formulated Dexlansoprazole SLNs included comprehensive physicochemical characterization involving FTIR, particle size analysis, zeta potential determination, scanning electron microscopy (SEM), entrapment efficiency assessment, and in vitro dissolution studies. The SEM micrographs revealed spherical particles with smooth surfaces, further validating uniformity of the formulation. Sustained drug release observed from the optimized SLN batch indicates potential for prolonged therapeutic effect and reduction in dosing frequency. The stability of the SLNs under refrigerated conditions further confirms robustness of the formulation approach [22].
2. MATERIALS AND METHODS
2.1 MATERIALS
2.1.1 Drug (Dexlansoprazole)
Dexlansoprazole, the active pharmaceutical ingredient used in this study, was received as a gift sample from a certified laboratory source. It is a well-known proton pump inhibitor (PPI) classified under the Biopharmaceutical Classification System (BCS) as a Class II drug, indicating low aqueous solubility but high permeability. Due to its poor solubility and susceptibility to degradation in acidic environments, enhancing its dissolution and stability is essential for improving its therapeutic performance. The drug sample obtained was of pharmaceutical grade quality and was used directly in the formulation process without any further purification.
2.1.2 Lipid (Glyceryl Monostearate, GMS)
Glyceryl monostearate (GMS) was selected as the lipid matrix for the preparation of solid lipid nanoparticles. GMS is a commonly used, biocompatible, and biodegradable lipid widely employed in nanoparticle formulations due to its excellent safety profile and ability to encapsulate lipophilic drugs. The lipid used in this study was of analytical grade and obtained from laboratory stock. Its physicochemical properties such as a suitable melting point, stability, and compatibility with Dexlansoprazole make it an ideal candidate for forming the solid lipid core required for SLN preparation.
2.1.3 Surfactant (Tween 80)
Tween 80 (Polysorbate 80) was employed as the surfactant in the formulation process. Being a nonionic surfactant with high solubilizing and stabilizing capabilities, Tween 80 plays a critical role in reducing interfacial tension during emulsification and ensuring the formation of uniform, stable nanoparticles. Its steric stabilization effect helps prevent aggregation of lipid particles, thereby enhancing the long-term physical stability of the SLN dispersion. Only analytical-grade Tween 80 was used in the study.
2.1.4 Solvent and Other Chemicals
Dimethylformamide (DMF) was used as the organic solvent to dissolve Dexlansoprazole during the formulation process. DMF’s strong solvent properties facilitate the complete dissolution of the lipophilic drug, enabling its efficient incorporation into the molten lipid phase. Distilled water of HPLC grade was used as the aqueous phase throughout the preparation steps to ensure purity and consistency. All chemicals and reagents used in the study were of analytical grade and were utilized as supplied, without the need for any additional purification steps.
2.2 Equipment
Table No.1: instruments were employed throughout the study
|
Instrument |
Model/Make |
Manufacturer |
|
UV–Visible Spectrophotometer |
V-530 |
JASCO, Japan |
|
Magnetic Stirrer |
Mini Mag Stirrer |
Remi, India |
|
Probe Sonicator |
Lab Model |
Rivotek |
|
Rotary Evaporator |
HB Digital |
Heidolph, Germany |
|
Cooling Centrifuge |
– |
Remi, India |
|
FTIR Spectrophotometer |
– |
Shimadzu, Japan |
|
Particle Size Analyzer (DLS) |
Zetasizer |
Malvern Instruments, UK |
|
Zeta Potential Analyzer |
Zetasizer |
Malvern Instruments, UK |
|
Scanning Electron Microscope (SEM) |
– |
– |
|
Vortex Mixer |
– |
Spinix/Equivalent |
|
Ultrasonic Bath Sonicator |
– |
Spectralab/Equivalent |
2.3 Preparation of Dexlansoprazole-Loaded Solid Lipid Nanoparticles (SLNs)
Dexlansoprazole-loaded solid lipid nanoparticles were formulated using a modified single-emulsion solvent evaporation technique, aided by probe sonication to achieve nanoscale particle size and uniform distribution. The method was optimized to ensure maximum drug entrapment, minimal particle size, and high formulation stability. The composition of the optimized batch consisted of 40 mg Dexlansoprazole, 400 mg Glyceryl Monostearate (GMS), 100 mg Tween 80 (0.5% w/v), 3 mL DMF, and 20 mL distilled water as the aqueous phase.
2.3.1 Preparation of the Lipid Phase
The lipid phase was prepared by accurately weighing 400 mg of GMS and melting it at 60 °C using a thermostatically controlled water bath. In a separate step, 40 mg of Dexlansoprazole was dissolved in 3 mL of dimethylformamide (DMF) to obtain a clear, homogeneous drug solution. This drug–solvent solution was then slowly introduced into the molten lipid under continuous stirring to ensure complete mixing and uniform dispersion of the drug within the lipid matrix. Throughout this process, the temperature was strictly maintained at 60 °C to prevent premature solidification of the lipid.
2.3.2 Preparation of the Aqueous Phase
The aqueous phase was prepared by dissolving 100 mg of Tween 80 in 20 mL of distilled water, yielding a 0.5% w/v surfactant solution. The surfactant plays a crucial role in stabilizing the emulsion and preventing particle aggregation. The aqueous phase was pre-heated to 60 °C to match the temperature of the lipid phase, ensuring seamless mixing during emulsification.
2.3.3 Formation of Coarse Emulsion
The hot aqueous phase was added gradually in a dropwise manner to the lipid phase while continuously stirring at 1000 rpm for approximately 5–10 minutes. This controlled addition facilitated the formation of a milky oil-in-water (o/w) coarse emulsion, which served as the precursor for nanoemulsion formation.
2.3.4 Probe Sonication (Nanoemulsion Formation)
To reduce the droplet size and produce a fine nanoemulsion, the coarse emulsion was subjected to probe ultrasonication using a Rivotek sonicator. Sonication was carried out at 40% amplitude, operated in pulsed cycles of 30 seconds ON and 15 seconds OFF to avoid overheating. The total sonication time ranged between 120–150 seconds, and an ice bath was used throughout the process to maintain temperature stability. This step effectively reduced the emulsion droplet size, leading to the formation of nanoscale particles.
2.3.5 Solvent Removal and Solidification
Following sonication, the nanoemulsion was transferred to a rotary evaporator to remove DMF under reduced pressure at 40–45 °C. This step allowed the evaporation of organic solvent and facilitated the solidification of lipid droplets into solid lipid nanoparticles. The resulting dispersion was then cooled to room temperature and subsequently stored at 4 °C for 1 hour to ensure complete particle solidification.
2.3.6 Purification of SLNs
The SLN dispersion was purified by centrifugation at 15,000 rpm for 30 minutes at 4 °C using a Remi cooling centrifuge. The supernatant was collected to analyze the amount of unentrapped (free) drug, while the pellet containing the SLNs was washed with distilled water to remove residual surfactant or impurities. The purified nanoparticles were then re-suspended to obtain the final SLN formulation.
2.4 Optimization of Formulation Parameters
A factorial design approach using Design Expert® Version 12 was adopted to systematically optimize the formulation. Three critical independent variables were evaluated:
Lipid concentration: 80–400 mg
Surfactant concentration: 10–100 mg
Sonication time: 5–25 minutes
The dependent variable selected for optimization was particle size (nm). A total of eight experimental formulations were generated as per the factorial matrix. From these, the optimized batch was selected based on achieving the smallest particle size, lowest polydispersity index (PDI), and highest entrapment efficiency. This statistical approach enabled the identification of significant factors and their interactions affecting nanoparticle characteristics.
2.5 Evaluation of Dexlansoprazole-Loaded SLNs
2.5.1 Particle Size, Polydispersity Index (PDI), and Zeta Potential
The mean particle size, PDI, and zeta potential of the SLNs were measured using Dynamic Light Scattering (DLS) on a Malvern Zetasizer. Particle size and PDI provided insights into the uniformity and distribution of nanoparticles, whereas zeta potential indicated the electrostatic stability of the dispersion. All measurements were performed at room temperature, and samples were appropriately diluted to avoid multiple scattering effects.
2.5.2 Entrapment Efficiency (EE%)
Entrapment efficiency was determined by quantifying the amount of free drug present in the supernatant after centrifugation. The concentration of unentrapped drug was analyzed spectrophotometrically at 284 nm using a UV–Visible spectrophotometer. EE% was calculated using the following formula:
EE%=Total drugFree drugTotal drug×100
This parameter reflects the drug-loading capacity of the formulation and the affinity between the drug and lipid matrix.
2.5.3 FTIR Analysis
Fourier Transform Infrared (FTIR) spectroscopy was performed using a Shimadzu FTIR instrument to evaluate possible chemical interactions between Dexlansoprazole and formulation excipients. FTIR spectra of pure drug, blank SLNs, and drug-loaded SLNs were recorded within the range of 4000–400 cm?¹. Characteristic absorption peaks were assessed to confirm drug compatibility and retention of functional groups.
2.5.4 In-Vitro Drug Release Study
In-vitro drug release was studied using a dialysis membrane method in phosphate buffer (pH 6.8) to mimic intestinal conditions. The SLN dispersion was placed inside the dialysis bag and immersed in the dissolution medium maintained at 37 ± 0.5 °C. Samples were withdrawn at predetermined time intervals, filtered, and analyzed at 284 nm. The release profile of the SLN formulation was compared against that of pure Dexlansoprazole to evaluate enhancement in dissolution characteristics.
2.5.5 Stability Studies
Stability of the optimized SLN formulation was assessed by storing the sample at 4 °C for 30 days. Particle size, PDI, and entrapment efficiency were periodically evaluated to determine any physical changes over time. These observations provided insight into the long-term stability and shelf-life of the nanoparticles.
2.5.6 Surface Morphology (SEM Analysis)
Scanning Electron Microscopy (SEM) was used to visualize the shape and surface characteristics of the SLNs. A small quantity of lyophilized nanoparticles was mounted on an aluminum stub, sputter-coated with gold, and examined under SEM at appropriate accelerating voltages. This analysis confirmed the spherical shape, surface smoothness, and morphological uniformity of the nanoparticles.
RESULTS & DISCUSSION:
Physicochemical Characterization of Dexlansoprazole
1. Chemical Identity
Dexlansoprazole is chemically identified as (R)-2-([3-methyl-4-(2,2,2-trifluoroethoxy) pyridin-2-yl] methyl sulfinyl)-1H-benzimidazole. It has a molecular formula of C??H??F?N?O?S with a molecular weight of 369.36 g/mol. The drug belongs to the class of substituted benzimidazole derivatives and represents the (R)-enantiomer of lansoprazole, which is known to exhibit improved pharmacokinetic performance and prolonged acid suppression compared to the racemic form.
2. Appearance
Dexlansoprazole was observed as a white to off-white crystalline powder, indicating its well-defined solid-state structure. This crystalline nature plays a critical role in its dissolution behavior and formulation stability.
3. Solubility
Dexlansoprazole exhibits very low aqueous solubility, approximately 0.1 mg/mL at pH 7, which significantly limits its oral bioavailability. The drug shows marked pH-dependent solubility, being freely soluble in acidic conditions (pH < 4) while remaining practically insoluble in neutral to alkaline environments (pH 6–8). However, it is readily soluble in organic solvents such as methanol, ethanol, dichloromethane, and dimethylformamide, confirming its lipophilic character. Based on these solubility and permeability properties, Dexlansoprazole is classified as a BCS Class II drug.
4. Partition Coefficient (log P)
The partition coefficient (log P) of Dexlansoprazole ranges between 2.0 and 2.3, indicating moderate lipophilicity. This property facilitates passive diffusion across biological membranes, enhancing permeability, but simultaneously contributes to poor dissolution in aqueous gastrointestinal fluids.
5. pKa Values
Dexlansoprazole exhibits two characteristic pKa values, with a primary pKa of approximately 4.0 attributed to the pyridine nitrogen and a secondary pKa of about 1.1 associated with the benzimidazole moiety. These ionization constants significantly influence the drug’s pH-dependent solubility, chemical stability, and absorption behavior.
6. Melting Point
The reported melting point of Dexlansoprazole lies between 138–140 °C, with slight variations depending on the purity and crystalline form of the drug. This thermal property is important for assessing processing conditions during formulation development.
7. Stability
Dexlansoprazole is acid-labile and undergoes rapid degradation in gastric acidic conditions, which poses a major challenge for oral delivery. In contrast, it remains relatively stable in neutral and alkaline media. Additionally, the drug is sensitive to light, heat, and moisture, necessitating appropriate protection during formulation, packaging, and storage.
8. Crystallinity
Dexlansoprazole exists predominantly in a crystalline form. However, it is susceptible to polymorphic transformations under stress conditions, which can affect its dissolution rate, stability, and overall performance in pharmaceutical formulations.
9. UV Absorption
The drug exhibits a characteristic UV absorption maximum (λmax) at approximately 284 nm in methanol–water systems. This property is commonly utilized for UV spectrophotometric estimation of Dexlansoprazole in analytical and formulation studies.
10. Pharmacokinetic Relevance of Physicochemical Properties
The physicochemical properties of Dexlansoprazole have a direct impact on its pharmacokinetic behavior. Despite its high membrane permeability, poor aqueous solubility limits its oral absorption. Furthermore, its acid-labile nature necessitates protective delivery approaches such as enteric coating or advanced nanocarrier systems, including solid lipid nanoparticles (SLNs) and nanostructured lipid carriers (NLCs). The development of dual delayed-release capsule technology specifically addresses these limitations by enhancing stability, prolonging drug release, and improving therapeutic efficacy.
UV–Visible Spectrophotometer Analysis of Dexlansoprazole
Principle
The UV–Visible spectrophotometric analysis is based on the Beer–Lambert law, which establishes a direct relationship between the absorbance of light and the concentration of the absorbing species in a solution. According to this law, the absorbance of a compound is directly proportional to its concentration and the path length of the cell, provided the system follows linearity within a specific concentration range. Dexlansoprazole exhibits strong absorption in the ultraviolet region due to the presence of conjugated aromatic and heterocyclic structures in its chemical composition. When scanned in a suitable solvent system, Dexlansoprazole shows a sharp and well-defined absorption maximum (λmax) at 282 nm. This characteristic absorption peak enables the accurate and reliable quantitative estimation of Dexlansoprazole using UV–Visible spectrophotometry.
Figure 1: UV–Visible Spectrophotometer Analysis of Dexlansoprazol
Instrumentation
The UV–Visible spectrophotometric analysis was carried out using a standard double-beam UV–Visible spectrophotometer equipped with appropriate optical and electronic components to ensure accurate and reproducible measurements.
Light Source:
The instrument employs two complementary light sources to cover the required spectral range. A deuterium lamp is used for the ultraviolet region (200–400 nm), providing a stable and continuous UV output. For the visible region (400–800 nm), a tungsten–halogen lamp is utilized, which ensures consistent intensity and reliability across the visible spectrum.
Monochromator:
A diffraction grating–based monochromator is incorporated to disperse the polychromatic radiation emitted from the light source into its constituent wavelengths. The monochromator selectively isolates the desired single wavelength required for analysis, thereby enhancing spectral resolution and accuracy.
Sample Holder:
The samples are placed in quartz cuvettes, which are transparent to ultraviolet radiation. Quartz is preferred over glass or plastic cuvettes, as the latter materials absorb UV light and may interfere with accurate absorbance measurements in the UV region.
Detector:
The transmitted light passing through the sample is measured using a sensitive detector such as a photodiode array or a photomultiplier tube. These detectors convert the light signal into an electrical signal proportional to the intensity of the transmitted radiation, enabling precise quantification of absorbance.
Readout and Software:
The instrument is integrated with advanced software and digital readout systems that display absorbance and transmittance values. The software also facilitates spectral scanning, data processing, and the generation of calibration curves, which are essential for quantitative analysis and method validation.
Procedure for Dexlansoprazole
Preparation of Stock Solution:
A primary stock solution of Dexlansoprazole was prepared by accurately weighing the required quantity of the drug and dissolving it in a suitable solvent system, such as DMF: water (2:8). This solvent mixture was selected to ensure complete solubilization of the drug and to obtain a clear, homogeneous solution suitable for spectrophotometric analysis.
Preparation of Working Solutions:
From the stock solution, a series of working standard solutions were prepared by appropriate dilution to obtain concentrations in the range of 1–25 µg/mL. These concentrations were chosen to cover the linear range required for quantitative analysis in accordance with Beer–Lambert’s law.
Spectral Scanning:
The prepared solutions were scanned in the UV–Visible spectrophotometer over the wavelength range of 200–400 nm using quartz cuvettes. This scanning range was selected to identify the maximum absorbance of Dexlansoprazole in the ultraviolet region.
Determination of λmax:
During spectral scanning, Dexlansoprazole exhibited a sharp and well-defined absorbance peak at 282 nm. This wavelength was considered as the λmax of Dexlansoprazole and was selected for further quantitative and analytical studies.
Calibration Curve and Linearity:
The absorbance values obtained at 282 nm were plotted against the corresponding concentrations to construct a calibration curve. The plot showed good linearity within the concentration range of 1–25 µg/mL, confirming the applicability of Beer–Lambert’s law and the suitability of the method for quantitative estimation.
Applications
Analytical Method Development:
The determination of the λmax of Dexlansoprazole provides a fundamental basis for the development and optimization of UV spectrophotometric analytical methods, ensuring specificity and sensitivity.
Quantitative Estimation:
The established method can be effectively applied for the quantitative estimation of Dexlansoprazole in bulk drug and pharmaceutical formulations, offering a simple, rapid, and cost-effective analytical approach.
Evaluation of Solid Lipid Nanoparticles:
The method serves as a reliable analytical tool for assessing entrapment efficiency and in vitro drug release studies of Dexlansoprazole-loaded solid lipid nanoparticles (SLNs), thereby supporting formulation development and performance evaluation.
Optimization of Variables by Using Factorial Design
In the present study, the formulation variables of Solid Lipid Nanoparticles (SLNs) were optimized using a factorial design approach. Factorial design is a systematic statistical method that allows simultaneous evaluation of the effect of multiple independent variables and their interactions on the dependent responses. This design provides a scientific and efficient way to identify the most significant factors influencing the formulation performance. For SLN optimization, variables such as lipid concentration, surfactant concentration, and solvent ratio were considered as independent factors, while particle size, polydispersity index (PDI), and entrapment efficiency (EE%) were selected as critical response parameters. The factorial design helped to generate mathematical models and response surface plots, which revealed the influence of each variable and their interactions on the quality of the nanoparticles. The optimization process enabled the selection of the most suitable formulation conditions that produced SLNs with minimum particle size, narrow size distribution (PDI < 0.3), and high entrapment efficiency. Thus, factorial design not only reduced the number of experimental trials but also ensured systematic optimization of the formulation for achieving reproducible and stable nanoparticles.
UV- Analysis:
Table 1: UV spectrophotometric calibration data of Dexlansoprazole showing the relationship between concentration (µg/mL) and absorbance at 282 nm
|
Concentration (µg/mL) |
Absorbance (AU) |
|
1 |
0.0287 |
|
2 |
0.0744 |
|
4 |
0.1598 |
|
8 |
0.3271 |
|
16 |
0.6570 |
|
20 |
0.8411 |
|
25 |
1.0397 |
Calibration curve:
Figure 2: Standard curve obtained for Dex lansoprazole
Calibration Curve (Paragraph for Thesis)
For the quantitative estimation of Dexlansoprazole, a calibration curve was constructed using UV–Visible spectrophotometer at λmax of 282 nm. A stock solution of the drug was prepared in DMF:Water (2:8), and serial dilutions were made in the concentration range of 1–25 µg/mL. The absorbance of each solution was measured at 282 nm against blank, and the obtained values showed a linear increase with concentration. The calibration curve plotted between concentration and absorbance obeyed Beer–Lambert’s law within the studied range, confirming the suitability of the method for analysis. The correlation coefficient (R²) was found to be close to unity, indicating excellent linearity of the method. This standard curve was further used for determination of drug content, entrapment efficiency, and in-vitro release studies of Dexlansoprazole-loaded Solid Lipid Nanoparticles (SLNs).
Partical size analyser:
The particle size of the developed Solid Lipid Nanoparticles (SLNs) was determined using Dynamic Light Scattering (DLS). The analysis showed that the formulation had an average particle size of 156 nm, which confirms the successful preparation of nanoparticles in the desired nanometric range. Nanoparticles with a size below 200 nm are advantageous for drug delivery, as they provide a larger surface area for dissolution, enhance solubility, and improve drug absorption. The obtained particle size indicates that the Dexlansoprazole-loaded SLNs are suitable for enhancing bioavailability, prolonging circulation time, and achieving controlled drug release. This optimized size range also contributes to better stability of the dispersion and efficient cellular uptake, which are crucial for improving therapeutic performance.
Fig No 3: Particle size distribution profile of Dexlansoprazole-loaded solid lipid nanoparticles showing differential intensity (%) and cumulative intensity (%) as a function of particle diameter (nm), as determined by dynamic light scattering (DLS).
1). FTIR of dexlanzoprazole API:
The FTIR spectrum of pure Dexlansoprazole was recorded to identify its characteristic functional groups. The observed absorption bands correspond to the principal functional groups present in the structure of Dexlansoprazole. These peaks serve as reference markers to compare with the FTIR spectrum of the Solid Lipid Nanoparticles (SLNs) in order to assess possible drug–excipient interactions. The results confirm the presence of hydroxyl, aromatic, amine, and carbonyl groups, which are characteristic of Dexlansoprazole.
Table No 2: FTIR Characteristic Peaks of Dexlansoprazole (API)
|
Functional Group |
Observed Frequency (cm?¹) |
Assignment / Nature |
|
O–H / N–H Stretch |
~3400–3200 |
Hydrogen bonded hydroxyl / amine |
|
Aromatic C–H Stretch |
~3100–3000 |
Aromatic ring C–H stretching |
|
Aliphatic C–H Stretch |
~2950–2850 |
Symmetric and asymmetric C–H stretch |
|
C=O Stretch (sulfoxide/ester/amide) |
~1730–1650 |
Carbonyl stretching vibration |
|
N–H Bending (amines) |
~1600–1550 |
Primary/secondary amine bending |
|
C=C Aromatic Stretch |
~1500–1450 |
Aromatic skeletal vibrations |
|
C–O Stretch (alcohol/ester) |
~1250–1050 |
C–O–C and C–O stretching |
|
Out-of-plane Aromatic C–H bend |
~900–700 |
Aromatic substitution vibrations |
Figure 4: Dexlanzoprazole API
2) FTIR Analysis of Blank Solid Lipid Nanoparticles
The FTIR spectrum of the blank Solid Lipid Nanoparticles (SLNs) was recorded to identify the characteristic functional groups of the excipients used, such as Glyceryl Monostearate (GMS), Lecithin, and Tween 80. The spectrum showed absorption peaks corresponding mainly to aliphatic C–H stretching, ester carbonyl (C=O) stretching, and C–O stretching vibrations of the lipid and surfactants. These peaks serve as reference values for comparison with drug-loaded formulations. The absence of characteristic Dexlansoprazole peaks in the blank SLN confirms that the formulation spectrum represents only excipients.
Table No 3: FTIR Peaks of Blank SLN (Without Drug)
|
Functional Group |
Observed Frequency (cm?¹) |
Assignment / Nature |
|
O–H Stretch (hydroxyl) |
~3400–3300 |
Broad –OH stretching from surfactant/lipid |
|
Aliphatic C–H Stretch |
~2950–2850 |
–CH? and –CH? symmetric & asymmetric stretch |
|
C=O Stretch (ester/lipid) |
~1750–1730 |
Ester carbonyl stretching (from GMS, lecithin) |
|
C–O Stretch (ester) |
~1250–1050 |
C–O–C stretching in lipids/surfactants |
|
CH? Bending (lipid chain) |
~1465–1380 |
Bending vibrations of lipid alkyl chains |
|
C–H Rocking |
~720–700 |
Long-chain hydrocarbon vibrations (lipid matrix) |
Figure 5: Blank Solid Lipid Nanoparticles
3) FTIR Analysis of Drug-Loaded Solid Lipid Nanoparticles
The FTIR spectrum of Dexlansoprazole-loaded Solid Lipid Nanoparticles (SLNs) was recorded to confirm the entrapment of the drug in the lipid matrix and to check for possible drug–excipient interactions. The spectrum of drug-loaded SLNs exhibited the characteristic peaks of both Dexlansoprazole (N–H stretching, aromatic C–H stretching, C=O stretching, C–O stretching) and the lipid excipients (GMS, lecithin, Tween 80). Importantly, no significant peak disappearance or shifting was observed, indicating that there is no strong chemical interaction between Dexlansoprazole and the lipid/surfactant. This confirms that the drug is physically entrapped within the SLN system and remains chemically stable.
Table No 4: FTIR Peaks of Drug-Loaded SLNs (Dexlansoprazole + Lipid Matrix)
|
Functional Group |
Observed Frequency (cm?¹) |
Assignment / Nature |
|
O–H / N–H Stretch |
~3400–3200 |
Broad hydroxyl / amine stretching (drug + lipid) |
|
Aromatic C–H Stretch |
~3100–3000 |
Aromatic ring vibrations (Dexlansoprazole) |
|
Aliphatic C–H Stretch |
~2950–2850 |
Lipid –CH?/–CH? stretching |
|
C=O Stretch (drug + lipid) |
~1730–1650 |
Ester (lipid) and amide/sulfoxide (drug) |
|
N–H Bending (drug) |
~1600–1550 |
Secondary amine bending |
|
C=C Aromatic Stretch |
~1500–1450 |
Aromatic skeletal vibration |
|
C–O Stretch |
~1250–1050 |
Ester/lipid + C–O of Dexlansoprazole |
|
C–H Rocking |
~720–700 |
Long-chain lipid hydrocarbon chains |
Drug-Loaded SLNs (Dexlansoprazole + Lipid Matrix)
Figure 6: Drug-Loaded SLNs (Dexlansoprazole + Lipid Matrix)
FTIR Analysis
The FTIR spectrum of the prepared formulation was recorded to identify the characteristic functional groups and to confirm drug–excipient compatibility. The graph (Figure X) shows distinct absorption bands in the range of 400–2000 cm?¹, which correspond to the principal vibrational frequencies of functional groups present in the formulation. Major peaks were observed in the regions around ~3400 cm?¹ (O–H/N–H stretching), 2950–2850 cm?¹ (aliphatic C–H stretching), 1730–1650 cm?¹ (C=O stretching), 1600–1550 cm?¹ (N–H bending), 1500–1450 cm?¹ (C=C aromatic stretching), and 1250–1050 cm?¹ (C–O stretching).The presence of these characteristic peaks in the FTIR spectra of the drug-loaded Solid Lipid Nanoparticles (SLNs) confirms that the functional groups of Dexlansoprazole remained intact and there was no significant chemical interaction with the lipid or surfactant. Thus, the FTIR analysis verifies successful encapsulation and compatibility of the drug with the excipients.
Figure 7: Dexlanzoprazole
Figure 8: blank sln
Figure 9: Solid Lipid Nanoparticals of Dexlanzoprazole
1.Scanning Electron Microscopy (SEM) Analysis
Scanning Electron Microscopy (SEM) is a powerful technique used to study the surface morphology, size, and shape of nanoparticles. In the present study, SEM analysis was performed to confirm the structural characteristics of the prepared Solid Lipid Nanoparticles (SLNs). The images obtained showed that the SLNs were predominantly spherical in shape with a smooth surface and uniform distribution, without significant aggregation. The particle size observed under SEM was in agreement with the results from Dynamic Light Scattering (DLS), further validating the nanoscale dimensions of the formulation. The spherical morphology of the nanoparticles is desirable because it provides a larger surface area, promotes better drug entrapment efficiency, and contributes to sustained release properties. Moreover, the absence of surface irregularities or cracks suggests good formulation stability. The SEM analysis thus confirms that the developed SLNs possess the appropriate morphology and physical characteristics required for efficient drug delivery.
Figure 10: SEM micrograph of best formulation.
DISCUSSION
The present investigation successfully demonstrated the potential of solid lipid nanoparticles as an effective carrier system for improving the solubility and stability of Dexlansoprazole. The nanoscale particle size achieved through probe sonication significantly increased surface area, which directly contributed to enhanced dissolution behavior. The use of glyceryl monostearate provided a stable lipid matrix capable of protecting the acid-labile drug from degradation, while Tween 80 ensured dispersion stability and uniform particle formation. FTIR studies confirmed drug–excipient compatibility, indicating that the formulation process did not alter the chemical integrity of Dexlansoprazole. SEM images supported the DLS findings by revealing spherical, smooth-surfaced nanoparticles, which are desirable for controlled drug release. The sustained in-vitro release profile observed for the SLNs suggests prolonged drug availability and potential reduction in dosing frequency. These findings collectively highlight the suitability of SLNs in overcoming formulation challenges associated with poorly soluble proton pump inhibitors
CONCLUSION
The study concludes that Dexlansoprazole-loaded solid lipid nanoparticles were successfully formulated using a modified emulsification-solvent evaporation method with probe sonication. The optimized SLN formulation exhibited desirable physicochemical characteristics, including nanoscale particle size, high entrapment efficiency, uniform morphology, and good stability. The formulation effectively addressed the major limitations of Dexlansoprazole, namely poor aqueous solubility and acid instability. FTIR and SEM analyses confirmed drug compatibility and successful encapsulation within the lipid matrix. The sustained drug release pattern observed in vitro indicates the potential of SLNs to enhance therapeutic efficacy and improve patient compliance. Therefore, solid lipid nanoparticles represent a promising and efficient delivery system for the oral administration of Dexlansoprazole. Further in-vivo and pharmacokinetic studies are recommended to establish the clinical relevance of this nanocarrier system.
REFERENCE
Aashwini Ghuge*, Dr. Pravin Wakte, Dr. Sachin Bhusari, Renuka Dhakne, Dr. Preeti Sable, Formulation and Evaluation of Dexlansoprazole-Loaded Solid Lipid Nanoparticles for Solubility Enhancement, Int. J. Sci. R. Tech., 2025, 2 (12), 284-301. https://doi.org/10.5281/zenodo.17984224
10.5281/zenodo.17984224