5 research outputs found

    A SIMPLE LIQUID CHROMATOGRAPHIC METHOD FOR SIMULTANEOUS ESTIMATION OF AZITHROMYCIN, FLUCONAZOLE AND ORNIDAZOLE IN BULK AND PHARMACEUTICAL DOSAGE FORMS

    Get PDF
    Objective: The objective of the study was to develop and validate a new rapid and more sensitive Reverse Phase High-Performance Liquid Chromatography (RP-HPLC) method for the simultaneous estimation of azithromycin, fluconazole and ornidazole in bulk and pharmaceutical dosage forms. Methods: Separation was achieved with a cap cell pack C18 column (4.6 x 250 mm, 5μ) with an isocratic mobile phase containing a mixture of acetonitrile and phosphate buffer pH 4.8 [adjusted with ortho-phosphoric acid] (50:50 % v/v) at the flow rate of 1 ml/min and detection was monitored at 210 nm. Results: The retention time (Rt) of azithromycin, fluconazole and ornidazole were found to be 4.82±0.01, 5.25±0.01 and 6.33±0.01 min respectively. The precision was found with<1.5% of %RSD. The calibration curve was linear over the concentration ranging from 500-1000 µg/ml for azithromycin, 75-150 µg/ml for fluconazole and 375-750 µg/ml for ornidazole with the correlation coefficient (r2) of 0.999. The percentage recovery was found to be within the specified range i.e., 98-102 % for three drugs. Limit of detection (LOD) was found to be 5.810, 1.790 and 4.924 µg/ml, whereas Limit of quantification limits (LOQ) was found to be 9.834, 2.667 and 7.980 µg/ml, respectively. Conclusion: A simple isocratic liquid chromatographic method was developed and validated for simultaneous estimation of azithromycin, fluconazole and ornidazole in their formulations. Due to its simplicity, rapidness and specificity, it can be applied for routine quality control analysis of these drugs

    IMPLICATION OF CENTRAL COMPOSITE DESIGN IN THE DEVELOPMENT OF SIMVASTATIN-LOADED NANOSPONGES

    No full text
    Objective:  The present study’s objective was to apply a central composite design to develop the simvastatin-loaded nanosponge formulation to improve its oral bioavailability. Methods: With the help of a design expert (State-Ease version 13.0.1), a central composite design was selected for the formulation of simvastatin-loaded nanosponges by using a defined concentration of Eudragit L-100 (X1) and PVA (X2) as independent variables and particle size (Y1), percent (%) entrapment efficiency (EE) (Y2), in vitro drug release (Y3) as dependent variables. Fourteen (SF1-SF14) formulations were prepared using the emulsion solvent evaporation and evaluated for surface morphology, particle size, drug-excipient compatibility, %EE, and % drug release. The optimized model (SF14) obtained from a design expert was evaluated for in vivo pharmacokinetics in animal models. Results: SF14 was formulated and evaluated for morphology (shape and size) of the particle, % EE, in vitro % drug release, and its kinetics. The formulation showed particle size of 163±0.45nm, 80.54 %±0.57 of EE, and 97.13%±0.38 of drug release at 8h. The release kinetics followed the zero-order and Higuchi mechanisms with non-fickian diffusion. In vivo results showed Cmax, Tmax, AUC0-t, AUC0-α, and MRT0-α for nanosponges were 0.175 µg/ml, 6 h, 1.561 µg/ml*h, 1.755 µg/ml*h, 11.77 h, respectively. Conclusion: The results indicated a significant increase in the bioavailability of the drug in nanosponges compared with standard drugs. The experimentally designed nanosponge formulations have been successfully developed, and evaluated parameters show that the nanosponge formulation of Simvastatin is a promising delivery through the oral route

    Hepatoprotective activity of QBD-based optimized N-acetyl cysteine solid lipid nanoparticles against CCL4-induced liver injury in mice

    No full text
    Purpose: In the present study, N-acetyl cysteine (NAC)-Solid Lipid Nanoparticles (SLNs) were developed employing the Quality by Design (QBD) approach for the application of hepatoprotective activity. Methods: Using Box-Behnken Design (BBD) three independent variables (Soya lecithin, polysorbate content, and homogenization speed) and four dependent variables (% entrapment efficiency (EE), % drug release (DR), zeta potential (ZP), and particle size (PS)) were chosen for the study. The formulations were prepared by the hot homogenization method and characterized with SEM, FTIR, DSC, and XRD and evaluated their % EE, % DR, PS, and ZP. Developed SLNs were tested for their hepatoprotective activity by an in vivo mice model and compared the effectiveness with free NAC and Silymarin. Results: The optimized NAC-SLNs were found optimum with spherical and intact chemical structure (88.95% EE, 97.15% DR, -43.01 mv ZP, < 200 nm of PS) exhibiting Higuchi model of drug release. In terms of MDA levels, NAC-SLNs had a strong protective impact MDA level (23.09±0.01–21.84±0.01 u mole/mg protein) and were efficient in increasing GPx (16.89±0.01–20.71±0.02 unit/mg protein), GSH (18.94±0.57–24.21±1.00 unit/mg protein), which were reduced in the CCl4-intoxicated group. NAC-SLNs were more effective than NAC at inhibiting the liver enzymes SGOT (150.01±1.5–132.01±0.6 mg/dL), SGPT (100.73±1.1–91.98±2.8 mg/dL), ALP (147.07±0.8–124.79±0.5 mg/dL), and LDH (290.37±3.04–228.25±2.03U/L). Conclusion: The study concludes that NAC-SLNs therapy was not only substantially more effective than NAC, but it also had effects equivalent to a well-known hepatoprotective and antioxidant drug Silymarin

    Hepatoprotective activity of QBD-based optimized N-acetyl cysteine solid lipid nanoparticles against CCL4-induced liver injury in mice

    No full text
    Purpose: In the present study, N-acetyl cysteine (NAC)-Solid Lipid Nanoparticles (SLNs) were developed employing the Quality by Design (QBD) approach for the application of hepatoprotective activity. Methods: Using Box-Behnken Design (BBD) three independent variables (Soya lecithin, polysorbate content, and homogenization speed) and four dependent variables (% entrapment efficiency (EE), % drug release (DR), zeta potential (ZP), and particle size (PS)) were chosen for the study. The formulations were prepared by the hot homogenization method and characterized with SEM, FTIR, DSC, and XRD and evaluated their % EE, % DR, PS, and ZP. Developed SLNs were tested for their hepatoprotective activity by an in vivo mice model and compared the effectiveness with free NAC and Silymarin. Results: The optimized NAC-SLNs were found optimum with spherical and intact chemical structure (88.95% EE, 97.15% DR, -43.01 mv ZP, < 200 nm of PS) exhibiting Higuchi model of drug release. In terms of MDA levels, NAC-SLNs had a strong protective impact MDA level (23.09±0.01–21.84±0.01 u mole/mg protein) and were efficient in increasing GPx (16.89±0.01–20.71±0.02 unit/mg protein), GSH (18.94±0.57–24.21±1.00 unit/mg protein), which were reduced in the CCl4-intoxicated group. NAC-SLNs were more effective than NAC at inhibiting the liver enzymes SGOT (150.01±1.5–132.01±0.6 mg/dL), SGPT (100.73±1.1–91.98±2.8 mg/dL), ALP (147.07±0.8–124.79±0.5 mg/dL), and LDH (290.37±3.04–228.25±2.03U/L). Conclusion: The study concludes that NAC-SLNs therapy was not only substantially more effective than NAC, but it also had effects equivalent to a well-known hepatoprotective and antioxidant drug Silymarin
    corecore