Introduction of an Innovative approach for Bioanalytical Method Development and Validation of Febuxostat by using LC-ESI-MS/MS in Human Plasma

 

Dibya Das1*, Dhiman Halder2, Himangshu Sekhar Maji1, Pintu Kumar De1, Sudipta Saha3, Navjot Singh4, Tapan Kumar Pal2

1Department of Pharmaceutical Technology, JIS University, Kolkata, India.

2Department of Pharmaceutical Technology, Bioequivalence Study Centre, Jadavpur University, Kolkata, India.

3UCD School of Public Health, Physiotherapy and Sports Science, University College Dublin.

4NRI Institute of Pharmacy, Bhopal, India.

*Corresponding Author E-mail: dibyadas1990gmail.com

 

ABSTRACT:

The aim of the present work is to develop and validate an accurate, sensitive, rapid, precise, and simple bioanalytical method for estimation of Febuxostat in human plasma by using LC-ESI-MS/MS. The method was developed by gradient elution technique with the combination of the mobile phase as 0.1% Formic Acid in Milli-Q water and 0.1% Formic Acid in Acetonitrile at a flow rate of 0.5ml/min. The Analyte and IS (Tolbutamide) were separated by a C18 Phenomenex Kinetex (50x3mm, 5µ) column. The chromatographic run time was 7.0 minutes. The plasma extraction was done by a simple protein precipitation technique (PPT). The LOD and LLOQ were found to be 6.25ng/ml and 125ng/ml, respectively. The extraction recovery of the drug from plasma was more than 90%. The validation parameters were found within the range, as mentioned by USFDA and EMA guidelines.

 

KEYWORDS: Febuxostat, Human Plasma, LC-ESI-MS/MS, Method Development, and Validation.

 

 


INTRODUCTION:

Bioanalytical method development plays an essential role in preclinical and clinical studies. The major part of bioanalysis is mainly concerned with developing a suitable method to determine the concentration of drugs, their metabolites, and/or endogenous substances in the biological matrices such as blood, plasma, serum, cerebrospinal fluid, urine, and saliva1,2. The application of the bioanalytical method is mainly focused on the evaluation of bioavailability, bioequivalence, pharmacokinetic studies of investigating drug molecules and other biological entities in various biological matrixes.3,4,5,6.

 

Febuxostat is an orally administered non-purine xanthine oxidase selective inhibitor approved for chronic hyperuricemia management in gout patients.7 Xanthine oxidase is needed to oxidize hypoxanthine and xanthine successively to uric acid. Febuxostat, therefore, inhibits xanthine oxidase, reducing uric acid production. Because of its firm binding to the Molybdenum Pterin Site, Febuxostat inhibits both oxidized and reduced forms of xanthine oxidase.8

 

Fig. 1: Febuxostat Structure.

 

Tolbutamide (Used as Internal Standard) is an oral hypoglycemic sulfonylurea medicine for first-generation potassium channel blockers. If the diet alone is not adequate, this drug can be used for treating type 2 diabetes. Tolbutamide stimulates the pancreatic secretion of insulin.

 

Selective and sensitive bioanalytical methods for the quantitative evaluation of drugs and their metabolites are critical for the successful biopharmaceutics and clinical pharmacology studies9. The validation of the biological method comprising all methods that show that the specifically developed method for quantitative analyte measurement is reliable and reproductive for the intended use in a specific biological matrix10. The bioanalytical method undergoes many modifications during a drug development process. Each modification should be validated to ensure the proper performance of the bioanalytical method. The objective of method validation is to exhibit the reliability of a particular method developed for the quantitative determination of an analyte in a specific biological matrix11-16. Literature survey reveals that few analytical methods are done for the determination of Febuxostat by HPLC in blood plasma.17

 

MATERIALS AND METHODS:

Chemicals and Reagents:

The active pharmaceutical ingredient (API) of Febuxostat (Purity 98.33%) was obtained from AKUMS Drugs and Pharmaceutical Ltd... Moreover, Tolbutamide was obtained from Shubham Biopharma, which is used as an Internal Standard (IS). Acetonitrile (HPLC grade), Formic Acid (HPLC grade), DMSO (HPLC grade) were purchased from Merck Pvt. Ltd, Mumbai, India. Water was purified by a Milli-Q gradient system of Millipore (Elix, Milli-Q A10 Academic, Bedford, MA, USA) until a resistively of 18.2 MΩ.cm was achieved. The blank human plasma with EDTA-K3 anticoagulant was collected from the Clinical Pharmacological Unit (CPU) of TAAB Biostudy Services, Kolkata, India.

 

Instrumentation:

The liquid chromatographic system consists of Shimadzu series LC-20 AD Binary pump, CTO-10 AS VP Column oven, SIL 20 AC Autosampler, andCBM-20A Lite System Control Compartment. The LC-MS/MS system API 2000 with triple quadrupole tandem mass spectrometer (Applied Biosystems/MDS SCIEX, Instruments) was used for the quantitative determination of Febuxostat and Tolbutamide (IS) in human plasma. Data acquisition, processing, and quantitation were performed with Analyst 1.6.3 software (Applied Biosystems/MD SCIEX).

 

Chromatographic conditions:

Chromatographic separation was done on an Phenomenex Kinetex C18; 50x3mm, Particle Size-5µm column. The Mobile phase used for the elution of the analyte was Pump A: 0.1% Formic Acid in Water (Milli-Q) and Pump B: 0.1% Formic Acid in Acetonitrile. The flow rate was set at 0.5mL/min. The injection volume was 10µL and the total run time was 7.0 minutes. The column temperature was not applicable in this way, while the autosampler temperature was maintained at 15oC.

 

Mass spectrometry

Turbo electrospray ionization (ESI) interface with multiple reaction monitoring (MRM) was used to acquire the mass spectra of the compounds. Ions were measured in negative ionization mode. The tuning parameters were optimized by injecting 100 ng/mL of a standard solution containing Febuxostat and Tolbutamide (IS) at 10µL/min. through an external syringe pump directly connected to the mass spectrometer. The applied high voltage was -4.5 Kv to the spray needle. The instrument was programmed for a scan dwell time of 100.00 msec. The MRM transitions of m/z 315.100 ®271.100, m/z 269.200 ®169.700 were used to measure Febuxostat and Tolbutamide (IS), respectively (Table 1). The source temperature of turbo ion spray was set at 400oC, and the turbo ion spray voltage was set at -4500.00 V. The value set of nebulizer gas (GS1) was 45.00 p.s.i. Moreover, the turbo ion-spray gas (GS2) value was set at 45 p.s.i. The curtain gas (CUR) and the collision-associated dissociation (CAD) gas flow at instruments were 30.00 and 5.00 (arbitrary scale), respectively (Table 2). Figure 2.A: Q1 Scan and Figure 2.B: Q3 (MS2) Scan of the drug Febuxostat.

 

Table 1: Mass Spectrometric Conditions:

 

FEBUXOSTAT

TOLBUTAMIDE (IS)

Precursor (DA)

315.100

269.200

Product (DA)

271.100

169.700

Dwell (ms)

100.00

100.00

DP (V)

-13.00

-18.00

FP (V)

-394.00

-371.00

CEP

-16.40

-15.25

CE (Ev)

-12.00

-28.00

EP (V)

-6.80

-5.70

CXP (V)

-8.50

-11.00

 

Table 2: Gas/Source Parameters:

CUR (Psi)

30.00

CAD (Psi)

5.00

IS (V)

- 4500.00

TEM (oC)

400.00

GS1 (Psi)

45.00

GS2 (Psi)

45.00

 

Figure 2. A: Q1 Scan of Febuxostat

 

Figure 2. B: Q3 (MS2) Scan of Febuxostat

 

Preparations of Standard solutions, quality control samples and calibration curve:

The standard solutions were prepared using DMSO containing 1mg/mL of Febuxostat and IS. From this solution, we prepare a stock solution of 1µg/mL for Febuxostat and 15μg/mL for IS. From the drug stock solution, we prepare seven working solutions for calibration standards, they were 125ng/mL, 250ng/mL, 500ng/mL, 1000ng/mL, 2000ng/mL, 4000ng/ml, and 8000ng/mL. The solutions are stored at 20 – 80C.

 

The four types of quality control samples are LLOQ, LQC, MQC, and HQC were prepared from the seven-point standard calibration solutions of Febuxostat. The LLOQ was 125ng/mL., lowest calibration concentration. The LQC was 375ng/mL., middle concentration of second (250ng/mL) and third (500ng/mL) concentrations. The MQC was 3000ng/mL. middle concentration of fifth (2000ng/mL) and sixth (4000 ng/mL) concentrations. The HQC was 6000ng/mL., middle of concentration of sixth (4000ng/mL) and seventh (8000ng/mL) concentrations.

 

Standard calibration solutions of Febuxostat was prepared by spiking appropriate amounts of analyte and IS in blank human plasma to yield final concentrations. The quality control (QC) samples were prepared at three concentration levels of analyte. Calibration curves were plotted with a peak area ratio of drug and IS on Y-axis and concentration on X-axis.

 

Sample preparation:

Protein precipitation technique (PPT) is one of the best and simple techniques for plasma extraction. Only 100μl of blank plasma sample was transferred to a 2ml plastic Eppendorf tube. 50μl of drug and 50μl of IS working solutions were spiked into it and vortex for 1 minute. After adding 300μl cold Acetonitrile and vortexing for 10 minutes. After that centrifugation was done at 12000rpm for 5min by cold centrifuge at 40C Then 300µl of supernatant was taken and transferred to autosampler vials for injections.18

Bioanalytical Method Validation:

The method validation was executed as per USFDA and EMA guidelines.

 

Selectivity:

Blank plasma from the each healthy human volunteer was collected for establishing selectivity of the method. Selectivity was determined at the LLOQ level means a lower limit of quantification. The acceptance criteria as per the USFDA guideline for selectivity is the area response at the retention time (RT) of the analytes in blank should not be more than 20% compared to the response in LLOQ.19

 

Carryover:

The possibility of interference from a run to that of the immediately injected next run was analyzed by injecting blank samples after HQC. According to the USFDA guideline, the carryover from an injection of high concentrations should be < 20% of the area of LLOQ.19

 

Sensitivity:

The spiked plasma samples at LLOQ level were injected five times for confirmation of the sensitivity of the method. As per the guideline of USFDA, the area of the analyte peak at LLOQ should be higher than five times the blank's peak area. As per the guideline of USFDA, the accuracy of the injected LLOQ samples should be within ±20%.19

 

Linearity:

Linearity was evaluated through regression analysis for the seven-point (125, 250, 500, 1000, 2000, 4000, and 8000ng/mL) standard calibration curve. The standard calibration curve was constructed, taking the drug/IS ratio on Y-axis and concentration on X-axis to determine the coefficient of determination (R˛).

 

Accuracy:

Accuracy of the method was evaluated in five replicates at four different concentrations of LLOQ, LQC, MQC, and HQC. The drug to IS peak area ratio was determined, and the concentration was back-calculated from the line equation (y=mx+c). The deviation of the estimated back-calculated value from the theoretical concentration was determined to calculate the accuracy. As per the guideline of USFDA, the acceptance criteria for accuracy is±15% of the theoretical concentration except at LLOQ, where it should be within ±20%.19

 

Precision:

The precision of the analytical procedure was assessed after injecting the spiked plasma samples in five replicates at LLOQ, LQC, MQC, and HQC levels. The %CV of the back-calculated concentrations of the repeated injections was calculated. Intraday precision was assessed by determining %CV of the response of the injections injected on the same day. Additionally, inter-day precision was calculated after determining the %CV of the measured values of the samples injected on different days. As per the guideline of USFDA.19

 

Extraction Recovery:

The potential of the sample preparation technique to extract Febuxostat, including IS from the biological sample, was determined by comparing the chromatographic response found in the extracted samples at LQC, MQC, and HQC in three replicates to that of the unextracted samples of the same concentration which correspond 100% recovery.19,20

 

Matrix Effect:

Comparison of the chromatographic responses of the post-extracted HQC, MQC and LQC samples with the response of the analytes from neat samples at equal concentrations was done to get the effect of plasma constituents over analyte ionization to determine IS. Similar concentration of analyte and IS concerning the recovery experiment was used to determine the matrix effect.

 

Robustness:

The robustness of the method was established by making deliberate minor variations in the composition of flow rate and mobile phase.21-25

 

Limit of Detection (LOD) and Lower Limit of Quantification (LLOQ):

LOD and LLOQ they were determined at signal to noise ratios by injecting series of dilute solutions with known concentrations.26,27,28

Stability:

The stability of the drug in plasma was estimated by performing different stability testing like short-term stability, freeze and thaw cycle stability, autosampler stability, benchtop stability, and long-term stability. Blank Human plasma was spiked with the analytes at the concentration of three quality control (LQC, MQC, and HQC) in five replicates followed by extraction and analyzed after completing the suitable storage condition for individual stability test. The stability of the analytes in stock solution was assessed by freshly preparing the QC samples from the stock solution before the study. Short-term stability study was spiked plasma samples were stored at -20°C for 24 hours, followed by extraction and analysis. Freeze and thaw stability study was done by storing samples at -20°C and exposed to three repeated freeze and thaw cycles by freezing at -20°C and thawing at normal laboratory conditions. Autosampler stability study was done by processed samples which were analyzed by injecting the samples after keeping 24 hours inside the autosampler maintained at 15°C. Benchtop stability study was performed by analyzing the plasma samples spiked with the analytes after keeping 24 hours on bench top at room temperature. Long-term stability study was spiked plasma samples were stored at -20°C for 45 days, followed by extraction and analysis. All the stability samples were compared against freshly spiked QC samples at LQC, MQC, and HQC level. As per the guideline of USFDA the stability studies, the accuracy at each level should be within ±15%.18

 

RESULTS AND DISCUSSIONS:

Selectivity:

The chromatograms showed no response in any of the blank samples, indicating an absence of any interference of plasma components at the similar RT of Febuxostat. For this reason, the developed method met the selectivity requirement as the RT in blank samples' peak area was ≤20% compared to the area in LLOQ for the analyte. Figure 3.A is the representative chromatograms of blank with IS. Figure 3.B represents a chromatogram of LLOQ with IS.

 


Figure 3. A: blank with IS

 

Figure 3. B: LLOQ with IS

 

Carryover:

There was no peak response observed at the RT of Febuxostat in a blank chromatogram when injected after HQC. The developed method is thus free from the carryover problem as analyte peak response in blank was less than 20% of LLOQ.

 

Sensitivity:

The analyte’s peak area of LLOQ was found five times higher than the blank's peak area. The precision (%CV) values for Febuxostat was 4.67-9.65%. For this reason, the developed method was sensitive as low as the concentration level of 125 ng/mL (LLOQ).

 

Linearity:

The R˛ value of the seven-point calibration curve was 0.9957 for Febuxostat. The mean accuracy for Febuxostat for the five LLOQ injections was 96.86-98.41%. Hence, the developed method was linear over the calibration range of 125-8000 ng/mL for the analyte. Figure 4 show the Calibration Curve of Febuxostat.


 

Figure 4: Calibration Curve of Febuxostat

 


Accuracy:

The accuracy determined from the back-calculated concentrations of QC samples, including LLOQ for five replicates was ranged from 92.47-109.20% for Febuxostat. The results showed that accuracy values were within the acceptance limit of ±15% for QC samples for LQC, MQC, and HQC. And ±20% for LLOQ. Intra-day and interday-accuracy study results have been shown in Table-3 and Table-4, respectively.


 

Table 3: Inter-day-accuracy:

Quality control

Concentration (ng/mL)

Run

Mean found (ng/mL)

SD

CV (%)

Accuracy (%)

 

LLOQ

 

 

125

1

115.59

4.70

4.07

92.47

2

136.51

10.66

7.81

109.20

3

116.92

3.87

3.31

93.54

 

LQC

 

 

375

1

375.51

11.10

2.96

100.14

2

411.43

14.10

3.43

109.72

3

395.69

6.43

1.63

105.50

 

MQC

 

 

3000

1

2905.57

45.49

1.57

96.85

2

3097.87

89.77

2.90

103.26

3

3058.09

54.72

1.79

101.94

 

HQC

 

 

6000

1

5372.53

92.55

1.72

89.54

2

5724.87

130.04

2.27

95.41

3

5603.54

120.21

2.15

93.39

 

Table 4: Intraday-accuracy:

Quality control

Concentration (ng/mL)

Mean found (ng/mL)

SD

CV (%)

Accuracy (%)

LLOQ

125

121.08

5.66

4.67

96.86

LQC

375

390.78

5.20

1.33

104.21

MQC

3000

3129.48

120.17

3.84

104.32

HQC

6000

5549.49

84.08

1.52

92.49

 


Precision:

The %CV determined from the back-calculated concentrations QC samples, including LLOQ for the five replicates in the intraday precision study, were 1.57-7.81% for Febuxostat. In an inter-day precision study, the %CV of back-calculated concentrations for the QC samples in three different days were ranged from 1.33-4.67% for Febuxostat. The precision experiment results satisfied the required criteria for the establishment of repeatability of the method. In this study, all the precision results met the acceptance criteria. Table-3 and Table-4 show the accuracy-precision study results in intra-day and inter-day experiments, respectively.

 

Extraction Recovery:

Mean extraction recovery for Febuxostat at the QC levels was 97.60-99.63%. The developed method of sample preparation is perfect for this drug analysis; hence, it shows high extraction efficiency for the analyte. Table-5 contains the data of extraction recovery.

 

Matrix effect:

In the study, evaluation of the matrix effect was done by the analysis of three batches of QC samples HQC, MQC, and LQC for the analyte. ±15% was the average matrix effect value obtained for the analyte. Table-6 contains the data of the matrix effects of analytes. The changes seen were insignificant.

 

Robustness:

The developed method was found robust.

 

Limit of Detection (LOD) and Lower Limit of Quantification (LLOQ):

LOD and LLOQ of Febuxostat were 62.50 ng/ml and 125 ng/mL, respectively. Since the LOD and LOQ values of the drug was achieved at a very low level, this method can be suitable for validation.

 

Stability:

The mean accuracy observed for the back-calculated concentration of the stability samples of all the stability experiment for Febuxostat was passed as per guideline. The analyte found to be stable in all tested stability conditions, including 24 hours on bench top, 24 hours in autosampler, three freeze-thaw cycles, 24 hours short term stability, and 45 days long term stability. The samples accuracy of all the run at LQC, MQC and HQC levels was within ±15% of the analyte. The results of all the stability study experiments of the method validation summarized in Table-7.


 

Table 5: Extraction Recovery:

 

Diluent Sample

In Plasma Sample

Quality Control

LQC

MQC

HQC

LQC

MQC

HQC

Concentration (ng/mL)

375

3000

6000

375

3000

6000

Mean found (ng/mL)

411.16

3104.23

5472.40

402.68

3029.61

5452.22

% Recovery

97.94

97.60

99.63



Table 6: Matrix effect (Analyte):

 

Extracted Blank Plasma Sample

Aqueous Sample

 

% OF ME

Quality Control

Concentration (ng/mL)

Mean found (ng/mL)

SD

CV (%)

Mean found (ng/mL)

SD

CV (%)

LQC

375

374.56

18.73

5.04

404.40

4.17

1.03

92.60

MQC

3000

3017.20

25.43

0.84

3202.72

20.52

0.64

94.21

HQC

6000

5481.16

130.42

2.38

5996.19

45.01

0.75

91.42


Table 7: Stability Studies:

Stability

Quality control

Concentration (ng/mL)

Mean found (ng/mL)

Accuracy (%)

 

Freshly Thawed

LQC

375

379.18

 

MQC

3000

3095.64

HQC

6000

5672.33

 

Bench top

(24 Hours)

LQC

375

400.90

105.73

MQC

3000

3109.27

100.44

HQC

6000

5709.30

100.65

 

Autosampler

(24 Hours)

LQC

375

394.69

104.09

MQC

3000

3043.63

98.32

HQC

6000

5956.82

105.02

 

Freeze-thaw

(3 Cycles)

LQC

375

409.24

107.93

MQC

3000

3038.02

98.14

HQC

6000

5416.12

95.48

 

Short term

 (24 Hours)

LQC

375

386.29

101.88

MQC

3000

2980.53

96.28

HQC

6000

5493.94

96.86

 

Long term

(45 Days)

LQC

375

383.06

101.02

MQC

3000

3218.80

103.98

HQC

6000

5956.80

105.02

 


CONCLUSION:

In this present study, a simple bioanalytical method was developed for the detection and quantification of Febuxostat in human plasma by using LC-ESI-MS/MS. The sample preparation procedure consists of a simple protein precipitation technique (PPT) using cold Acetonitrile. The developed method is very cost-effective and also was found to have accepted in terms of various parameters like selectivity, accuracy, precision, and stability performed as per the guideline that is USFDA bioanalytical method validation. Therefore, this study definitely will have a significant contribution to the field of bioanalytical research and will be superior enough to apply in comparative pharmacokinetic studies in the future. The entire study to analyses plasma drug concentration was designed with proper validation to find the vital pharmacokinetic parameters of the drug Febuxostat. The method can be suitably used by the researchers to calculate pharmacokinetic parameters and as well as bioavailability of the drugs in human volunteers. And the routine analysis of Febuxostat in pharmaceutical dosage forms for routine application in quality control laboratories.

 

ACKNOWLEDGEMENT:

The authors want to acknowledge M/S, TAAB Biostudy Services, Kolkata-700032, India, for providing blank human plasma samples and the necessary instrumental facilities to complete the work.

 

CONFLICT OF INTEREST:

The authors declare that there is no conflict of interest.

 

REFERENCES:

1.      FDA US. Bioanalytical method validation guidance for industry. US Department of Health and Human Services Food and Drug Administration Center for Drug Evaluation and Research and Center for Veterinary Medicine. 2001.

2.      Pulido A, Ruisánchez I, Boqué R, Rius FX. Uncertainty of results in routine qualitative analysis. TrAC Trends in Analytical Chemistry. 2003 Oct 1; 22(9): 647-54.

3.      Murugan S, Pravallika N, Sirisha P, Chandrakala K. A review on bioanalytical method development and validation by using LC-MS/MS. Journal of Chemical and Pharmaceutical Sciences. 2013 Mar; 6(1): 41-5

4.      Bressolle F, Bromet-Petit M, Audran M. Validation of liquid chromatographic and gas chromatographic methods Applications to pharmacokinetics. Journal of Chromatography B: Biomedical Sciences and Applications. 1996 Nov 8; 686(1): 3-10.

5.      Hartmann C, Smeyers-Verbeke J, Massart DL, McDowall RD. Validation of bioanalytical chromatographic methods. Journal of Pharmaceutical and Biomedical Analysis. 1998 Jun 1; 17(2): 193-218.

6.      James CA, Breda M, Frigerio E. Bioanalytical method validation: a risk-based approach? Journal of Pharmaceutical and Biomedical Analysis. 2004 Jun 29; 35(4): 887-93.

7.      Hair PI, McCormack PL, Keating GM. Febuxostat. Drugs. 2008; 68(13): 1865-74.

8.      Love BL, Barrons R, Veverka A, Snider KM. Urate‐lowering therapy for gout: focus on febuxostat. Pharmacotherapy: The Journal of Human Pharmacology and Drug Therapy. 2010 Jun; 30(6): 594-608.

9.      Rocci ML, Devanarayan V, Haughey DB, Jardieu P. Confirmatory reanalysis of incurred bioanalytical samples. The AAPS journal. 2007 Sep 1; 9(3): E336-43

10.    Viswanathan CT, Bansal S, Booth B, DeStefano AJ, Rose MJ, Sailstad J, Shah VP, Skelly JP, Swann PG, Weiner R. Workshop/conference report—quantitative bioanalytical methods validation and implementation: best practices for chromatographic and ligand binding assays. The AAPS Journal. 2007 Mar 1; 9(1): E30-42.

11.    Pandey S, Pandey P, Tiwari G, Tiwari R. Bioanalysis in drug discovery and development. Pharmaceutical methods. 2010 Oct 1; 1(1): 14-24.

12.    Shah VP. The history of bioanalytical method validation and regulation: evolution of a guidance document on bioanalytical methods validation. The AAPS Journal. 2007 Mar; 9(1): E43.

13.    Garofolo F. The 1st Canadian Workshop on the Crystal City AAPS/FDA White Paper. In Workshop Report. http://www. Canadianl CMS group. com/docs/B-2007WorkshopWhitePaper. pdf. Accessed on 2010 Jul (Vol. 29).

14.    Savoie N, Booth BP, Bradley T, et al. The 2nd calibration and validation group workshop on recent issues in good laboratory practice bioanalysis.

15.    Trullols E, Ruisánchez I, Rius FX. Validation of qualitative analytical methods. TrAC Trends in Analytical Chemistry. 2004 Feb 1;23(2):137-45.

16.    Buick AR, Doig MV, Jeal SC, Land GS, McDowall RD. Method validation in the bioanalytical laboratory. Journal of Pharmaceutical and Biomedical Analysis. 1990 Jan 1; 8(8-12): 629-37.

17.    Shiv Kumar Gupta, Babita Kumar, Pramod Kumar Sharma. Development and validation of a RP-HPLC method for estimation of Thalidomide in solid dosage form. Asian J. Pharm. Ana. 3(1): Jan.-Mar. 2013; Page 17-19.

18.    Halder D, Dan S, Sarkar P, Das D, Chandra Halder U, Kumar Pal T. LC-MS/MS determination of 4-hydroxynimesulide, an active metabolite of nimesulide and application to bioequivalence study in Indian subjects. European Journal of Mass Spectrometry. 2019;25(5): 399-411.

19.    FDA US. Bioanalytical method validation guidance for industry. US Department of Health and Human Services Food and Drug Administration Center for Drug Evaluation and Research and Center for Veterinary Medicine. 2018 May.

20.    Jumle RS, Mundhey AS, Wate SP, et al. UV-spectrophotometric method development and validation for estimation of tizanidine and aceclofenac in tablet formulation. Asian Journal of Pharmaceutical Analysis. 2012; 2(4): 101-3.

21.    Akhilesh Gupta, Swati Rawat, Arun Pandey. Method Development and Photolytic Degradation Study of Doxofylline by RP-HPLC and LC-MS/MS. Asian J. Pharm. Ana. 1(2): April-June 2011; Page 29-33.

22.    Swapna J, Madhu C, Srivani M, et al. Analytical method development and method validation for the simultaneous estimation of metformin hydrochloride and pioglitazone hydrochloride in tablet dosage form by RP-HPLC. Asian Journal of Pharmaceutical Analysis. 2012; 2(3): 85-9.

23.    Renu Solanki, Badri Prakash Nagori, Mahendra Kumar Naval, Joytosh Banerjee. Development and Validation of Simultaneous Estimation Method for Amoxycillin Trihydrate and Tinidazole in Tablet Dosage Form by RP-HPLC. Asian J. Pharm. Ana. 3(2): April- June 2013; Page 66-71.

24.    D. Narmada, P.V. Murali Krishna, Shaik Mohammad Yusuf, B. Ranganayakulu, K. Uday Praveen, P. Raja Abhilash. RP-HPLC method development and validation for estimation of Glibenclamide in tablet dosage form. Asian J. Pharm. Ana. 4(3): July-Sept 2014; Page 125-128.

25.    Govinda Rao Kamala, Sowjanya Vadrevu, Malipeddi Haripriya. Method Development and Validation for Simultaneous Estimation of Omeprazole and Domperidone by RP-HPLC. Asian J. Pharm. Ana. 5(4): 2015; 195-205. doi: 10.5958/2231-5675.2015.00031.9.

26.    Sumithra, M., Yuvanesh, P. and Mistry, A., 2016. Analytical method development and validation of ambroxol hydrochloride by UV spectroscopy and forced degradation study and detection of stability. Research Journal of Pharmacy and Technology, 9(7), pp.794-800.

27.    K.S. Nataraj, P. Sivalingachari, S. Sai Naveen, K. Alekhya. Method Development and Validation for the Estimation of Dasatinib Monohydrate Tablets by RP-HPLC. Asian J. Research Chem. 6(9): September 2013; Page 859-862.

28.    D.M.Darandale, K.B. Erande, S.R. Tambe, R.S. Bhamber. Development and Validation of RP-HPLC Method for the Determination of Febuxostat in Bulk and Pharmaceutical Dosage Form. Asian J. Research Chem. 2017; 10(6): 713-718. doi: 10.5958/0974-4150.2017.00121.3

 

 

 

Received on 23.07.2020           Modified on 19.08.2020

Accepted on 11.09.2020         © RJPT All right reserved

Research J. Pharm. and Tech. 2021; 14(8):4060-4066.

DOI: 10.52711/0974-360X.2021.00703