You are on page 1of 10

Development and validation of LC‐MS/MS method for

simultaneous determination of sofosbuvir and daclatasvir in


human Plasma: Application to pharmacokinetic study
Ola M. Abdallah1,2 | Ahmed M. Abdel‐Megied3 | Amira S. Gouda4

1Analytical Chemistry Department, Faculty of


Pharmacy, Al‐Azhar University (Girls), Cairo,
Abstract
Egypt
A simple and highly sensitive liquid chromatography–tandem mass spectrometry (LC‐MS/MS)
2Pharmaceutical Chemistry Department,
bioanalytical method was developed and fully validated for the first time for the simultaneous
Faculty of Pharmacy, Badr University in Cairo
(BUC), Badr City, Cairo, Egypt determination of newly discovered antiviral drugs, namely sofosbuvir (SOF) and daclatasvir (DAC) in
3Pharmaceutical Analytical Chemistry human plasma. Tadalafil (TAD) was used as internal standard (IS). SOF, DAC and TAD (IS) were extracted
Department, Faculty of Pharmacy and from plasma using liquid–liquid extraction technique with methyl tert‐butyl ether. The chromatographic
Pharmaceutical Manufacturing, Kafrelsheikh
separation was carried out using ZorbaxSB‐C18 column (4.6 × 50 mm,5 μm) and 5 mM ammonium
University, Kafrelsheikh City, Egypt
4Zi‐diligence Research Center, Cairo, Egypt
formate buffer (pH 3.5)–acetonitrile (50:50, v/v) as mobile phase in an isocratic elution mode pumped

Correspondence
at a flow rate 0.7 mL min−1. The quantitation was performed on API4500 triple quadrupole tandem mass
Ahmed M. Abdel‐Megied, Pharmaceutical spectrometer with positive electrospray ionization interface in multiple reaction monitoring mode.
Analytical Chemistry Department, Faculty of Validation was applied according to US Food and Drug Administration guidelines for bio‐analytical
Pharmacy and Pharmaceutical Manufacturing,
Kafrelsheikh University, Kafrelsheikh City, Egypt. methodswith respect to linearity, precision, accuracy, selectivity, carry‐over, stability and dilution
Email: dr_ahmed80@hotmail.com, integrity. Linearity was obtained over concentration ranges of 0.3–3000 and 3–3000 ng mL−1 for SOF and
dr_ahmed80@pharm.kfs.edu.eg
DAC, respectively, by applying a weighted least‐squares linear regression method (1/x2). The proposed
1 | INTRODUCTION
method could be applied successfully in bioequivalence and/or clinical studies for therapeutic drug

Hepatitis C virus (HCV) infection and its monitoring of patients undergoing dual combination therapy as the latter combination proved more

complications affect more than 150 million efficacious and powerful tool for the complete treatment of hepatitis C genotype 3 within 16 weeks.

people worldwide, especially in Egypt, which The suggested method has been applied successfully to pharmacokinetic studies with excellent assay

has the highest prevalence of HCV in the ruggedness and reproducibility.

world, estimated nationally about 14.7%


KEYWORDS
(Kandeel et al., 2017; Souri, Jalalizadeh, &
Shafiee, 2007). Thus prevention of HCV must daclatasvir, hepatitis C, human plasma, LC‐MS/MS, pharmacokinetic study, sofosbuvir

become a national priority as it is considered the main cause of liver anti‐HCV activity in vitro (Product leaflet, 2013). Currently, SOF is the most

cirrhosis and hepatocellular carcinoma (Thein, Yi, Dore, & Krahn, 2008). Up used antiviral agent owing to its high pangenotypic effectiveness, alone or

until 2011, interferon (PEG‐α‐INF) with ribavirin was the only combination in combination with others such as daclatasvir (DAC), which inhibits the

available in global market. This protocol of treatment was effective against HCV nonstructural protein 5A (NS5A) by interfering with the replication and

genotype 1 and associated with severe side effects (Palumbo, 2011). A new impairing phosphotidylinositol‐4‐kinase III‐α activation. However, NS5A

antiviral class, sofosbuvir (SOF), is an oral nucleotide analog that inhibits has no known enzymatic functions and so it is difficult to understand its

the RNA polymerase of the HCV virus by being a defective substrate (Sofia mode of action (Belema et al., 2014; Smith, Regal, &

et al., 2010). The major metabolite of SOF, GS‐331007, was proven to lack
Mohammad, 2016; Figure 1). There are eight metabolites of DAC, one of which (BMS‐805215) has some activity; however, it is 100‐fold less potent than the
parent drug (Product monograph, 2016). Exposure of

Abbreviations: DAC, daclatasvir; HCV, hepatitis C virus; MRM, multiple reaction


monitoring; NS5A, nonstructural protein 5A; SOF, sofosbuvir; TAD, tadalafil.
Biomedical Chromatography. 2018;e4186. wileyonlinelibrary.com/journal/bmc
Copyright © 2018 John Wiley & Sons, Ltd. 1 of 9 https://doi.org/10.1002/bmc.4186
2 of 1 ABDALLAH ET AL .
0

FIGURE 1 Chemical structures for (a) sofosbuvir (SOF) and (b) daclatasvir (DAC)
the SOF metabolite (GS‐331007) was similar in the presence and absence Ultra‐pure water (resistivity > 18 MΩ cm−1 at 25°C and TOC < 5 ppb) was
of DAC, while SOF exposures were ~35% higher in the presence of DAC obtained from a MilliQ UF‐Plus system (Millipore, Bedford, MA, USA). Blank
(http://www.hcvdruginfo.ca/downloads/ plasma samples were obtained from (a) Vacsera Holding Co. (Giza, Egypt),
Hepatitis%20Cint_new%20NS5A%20inhibitors.pdf). The published (b) Dr Mohamed El Shabrawishy Hospital (Giza, Egypt), (c) Mataria Institute
methods for determination of SOF and/or DAC in human plasma are HPLC for Kidney and Urology (Cairo, Egypt), (d) El Demerdash Hospital (Cairo,
(Baker, El‐Kafrawy, Mahrous, & Belal, 2017; Hassib, Taha, Elkady, & Egypt) and (e) El‐Zeraien Hospital (Giza, Egypt) and El‐Helal Hospital (Cairo,
Barakat, 2017; Nannetti et al., 2017; Srinivasu, Kumar, & Thirupathi, 2016), Egypt).
LC‐MS/MS (Abdallah, Abdel‐Megied, & Gouda, 2017; Ariaudo et al., 2016;
Jiang et al., 2015; Nebsen & Elzanfaly, 2016; Qu, Wang, Zeng, Lub, & Yin,
2.2 | Pharmaceutical formulations
2015; Rezk et al., 2015, 2016, b; Shi et al., 2015) and electrochemical
Solvaldi® 400 mg tablets were manufactured by Gilead Sciences (Co. Cork,
detection (Azab & Fekry, 2017). To author's knowledge, this bio‐analytical
Ireland) for Gilead Sciences international (Cambridge, UK). Daklinza® 60 mg
method is the only LC‐MS/MS method for determination of SOF and DAC in
film‐coated tablets (Bristol‐Myers Squibb Pharma EEIG, UK) were labeled
human plasma with application to a pharmacokinetic study for both. Owing
to contain 65.92 mg daclatasvir dihydrochloride, equivalent to 60 mg DAC
to extensive use of these drugs in combination therapy, the monitoring of
per tablet.
SOF and DAC concentrations in patients undergoing HCV therapy is crucial,
especially to determine drug interaction potential. Treatment optimization
through therapeutic drug monitoring has already been reported to increase 2.3 | Instruments
the efficacy of the therapy, and could lead to a cost saving as well. Quantitative analysis was performed using an Exion LC™ chromatographic
The present work aimed to develop a simple and highly sensitive LC‐ system (ABSciex, USA). The detection was performed using a triple
MS/MS bio‐analytical method with high accuracy and precision for quadrupole mass spectrometry API4500 (ABSciex, Canada) operated in
simultaneous determination of SOF and DAC in human plasma. The method positive electrospray ionization using multiple reaction monitoring (MRM)
was thoroughly validated according to new World Health Organization bio‐ mode. The curtain, ion source gas 1 and ion source gas 2 pressures were
analytical guidelines (Bristol‐Myers‐Squibb, 2014; WHO Prequalification set at 30, 30 and 20 psi, respectively. The source temperature was 600°C
Team – Medicines, 2015). and the ion‐spray capillary voltage was 4000 V. The collision gas was set at
8 psi. The optimization of the analyte‐dependent MS/MS parameters was
performed via direct infusion of standards into the MS at a flow rate of 10
2 | EXPERIMENTAL
μL/min. Data acquisitions were performed with Analyst 1.6.3 software
Hotfixes® (ABSciex) to control all parameters of HPLC and MS.
2.1 | Chemicals and reagents
SOF was purchased from Optemus Ltd (Telangana, India). DAC was
2.4 | Liquid chromatographic and mass
purchased from Virupaksha Organics Ltd (Mumbai, India). Tadalafil (IS) was
spectrometric conditions
purchased from SMS Ltd (Hyderabad, India). Methanol, acetonitrile and
Chromatographic separations were achieved using a Zorbax SB‐C18 column
ammonium formate (HPLC grade) were obtained from Sigma‐Aldrich
(4.6 × 50 mm, 5 μm; Agilent Technologies, CA, USA) and the mobile phase
(Milwaukee, USA). Methyl tert‐butyl ether (HPLC grade) was purchased
was pumped with an isocratic elution program consisting of 5 m M
from Scharlab S.L. Chemicals (Barcelona, Spain).
ABDALLAH ET AL . 3 of 1
0

ammonium formate buffer pH 3.5–acetonitrile (50:50, v/v) was delivered chromatographic interference from endogenous plasma components
at flow rate 0.7 mL min−1. Each component of the mobile phase was between the analytes and IS.
degassed before use in an ultrasonic bath for 10 min. The column 2.7.2 | Linearity and range
temperature was maintained at 25°C with an injection volume of 3 μL. The Calibration curves were constructed by plotting the ratio of peak area of
MRM mode with positive electrospray ionization interface was used by SOF and DAC with TAD (IS) against the nominal concentration of calibration
detection of the ions at transition pairs of m/z 530.0 → 243.3 for SOF, m/z standards. Concentrations used for SOF were 0.3, 3, 15, 30, 150, 300, 900,
739.5 → 565.3 for DAC and m/z 390.2 → 268.2 for tadalafil (TAD) (IS) which 1500 and 3000 ng mL−1 and 3, 15, 30, 150, 300, 900, 1500 and 3000 ng mL−1
are represented in Table 1. for DAC. Construction of calibration plots was performed using weighted
least‐squares linear regression method (1/x2) by measuring the peak area
ratio of the analytes to the IS. Linearity was achieved with correlation
2.5 | Standard solutions and calibration curves
coefficients 0.9996 and 0.9984 for SOF and DAC, respectively. The QC
The primary stock solutions (300 μg mL−1 of SOF and DAC, and 100 μg mL−1
samples concentrations and analytes under investigation were calculated
of TAD, IS) were completed with methanol and stored at −20°C for 2 weeks.
according to the calibration plots. The set of acceptance criteria deviated
Appropriate dilutions were then made in methanol to produce working
±15% from the nominal value for back‐calculated standard concentrations
solutions of 30,000 ng mL−1of SOF and DAC and 10 μg mL−1 of TAD (IS). For
except for LLOQ, which was set as ±20%.
each analyte, quality control (QC) samples were prepared at three levels
high (QCH), medium (QCM) and low (QCL) at concentrations 2400, 1200,
0.9 ng mL−1 for SOF and 2400, 1200, 9 ng mL−1 for DAC. 2.7.3 | Carry‐over
Blank plasma samples were injected after calibration standard at the ULOQ
to make sure that it did not affect the accuracy and the precision of the
2.6 | Sample preparation proposed method. For all three analytes, the carry‐over should be <20% of
Before analysis, the plasma samples were thawed at room temperature. the peak response of the LLOQ.
−1
Anliquot of 0.5 mL plasma was spiked with 50 μL of 10 μg mL IS then
vortex mixed for 30 s. Liquid–liquid extraction was carried out by adding
2.7.4 | Precision and accuracy
3.5 mL methyl tert‐butyl ether, and mixed for 4 min. Finally, samples were
Assay run precision was determined as intra‐ and inter‐day precision using
centrifuged at 4500 rpm at ambient temperature for another 5 min. The
six replicates (n = 6) for intra‐day and 18 replicates (n = 18) for inter‐day,
clear upper layer was separated carefully and evaporated at 45°C under a
expressed as relative standard deviation (RSD). The accuracy of the method
vacuum stream, then it was reconstituted with 300 μL of acetonitrile–water
was expressed as relative error (RE) and was within ±10.06% for the
in the proportion 40:60, v/v, and finally, 3 μL was injected into the LC‐
analytes. It was expressed with an acceptance criterion of ±15% except for
MS/MS system.
the LLOQ, which was set as ±20%.

2.7 | Method validation


2.7.5 | Extraction recovery and matrix effect
Validation of the proposed method was performed with respect to The relative recoveries (RE) of SOF, DAC and TAD (IS) from real human
selectivity, precision, accuracy, matrix effect, stability, dilution integrity, plasma were determined by comparing the peak area before extraction and

TABLE 1 LC‐MS/MS parameters selected for the quantification of SOF, DAC and TAD (IS)
Analyte Q1a (m/z) Q3b (m/z) Declustering potential (V) Entrance potential (V) Collision energy (V) Cell exit potential (V)

SOF 530.0 243.3 80.0 10.0 26.2 9.0

DAC 739.5 565.3 80.0 10.0 63.7 13.5

TAD (IS) 390.2 268.2 59.0 10.0 16.0 9.0


DAC, daclatasvir; SOF, sofosbuvir; TAD, tadalafil.
a
Q1, precursor ion.
b Q3, product ion.
carry‐over and application to incurred plasma samples as per the guidelines the response of unextracted standards at equivalent concentrations. The IS
of the US Food and Drug Administration (2001; Zimmer, 2014). recovery was similarly assessed. The matrix effect (ME) was demonstrated
by applying a comparison between the peak areas obtained from samples
2.7.1 | Selectivity with those obtained from the pure standard solution at equivalent

The selectivity of the proposed method was evaluated by analyzing six concentrations. The process efficiency was estimated as PE = [ME × RE/100]

different sources of human plasma to demonstrate the lack of (Matuszewski, Constanzer, & Chavez‐Eng, 2003). All parameters were
evaluated at three QC levels in six replicates (n = 6). The estimation of IS‐
4 of 1 ABDALLAH ET AL .
0

normalized matrix factor was obtained by dividing the matrix factor of each and Daklinza® 60 mg tablets. The samples were immediately centrifuged at
analyte by the matrix factor of the IS. 3500 rpm for 10 min. The plasma obtained was stored at −80°C until
2.7.6 | Stability analysis. All pharmacokinetic parameters were estimated using the

Analyte stability in human plasma was evaluated by analyzing three validated Kinetica® 5.1 SP1software.

replicates (n = 3) at the low and high concentrations. The samples were 3 | RESULTS AND DISCUSSION
exposed to different conditions of temperature with different time
intervals and the results were compared with those obtained from freshly 3.1 | Development of sample preparation
prepared samples. Stability of analytes in plasma was evaluated at room
Development of a procedure for plasma extraction is considered the
temperature after 24 h. The extracted samples were kept in the
cornerstone of any bio‐analytical study in order to reach the maximum
autosampler at 4°C and evaluated after 72 h. The QC samples were frozen
extraction recovery with minimum matrix effects, thus enhancing the
and stored at −80°C for 100 days. The samples were evaluated after three
sensitivity and accuracy of LC‐MS/MS analysis. The procedure used for the
freeze–thaw cycles, stored at −80°C for 12 h and thawed unassisted at
extraction of any drug from biological matrices usually requires the removal
room temperature. The results should be within 15% of nominal
of proteins and any interfering species prior to the HPLC analysis.
concentrations.
Therefore, many trials were performed to achieve the best recovery with
the lowest matrix effect. For simplicity and time‐saving, a protein

2.7.7 | Dilution integrity precipitation technique using methanol, acetonitrile was tried but the
results were unsatisfactory owing to high matrix effects and low recovery
Dilution integrity could be demonstrated through spiking the matrix with
at low concentrations. Therefore, liquid–liquid extraction using methyl tert‐
an analyte concentration above the LLOQ and diluting this sample with the
butyl ether, dichloromethane and diethyl ether was also applied. It was
same blank matrix (at least five determinations per dilution factor). SOF and
found that methyl tert‐butyl ether is the most reproducible solvent with
DAC were spiked separately into real human plasma samples. Furthermore,
the best recovery and minimal matrix effect.
these solutions were diluted in six replicates (n = 6) by 2‐ and 4‐fold with
human plasma. The acceptance criteria must have accuracy of 100 ± 15%
and precision ≤15%.
3.2 | Optimization of the chromatographic
conditions
Optimization of the proposed method was required several trials using
2.7.8 | Incurred plasma samples reanalysis
different mobile phases and stationary phases. Columns such as Kinetex®
The use of calibration standards and QC samples during validation may not
C8 column (100 × 4.6 mm, 5 μm), Kinetex® C8 column (50 × 4.6 mm, 5 μm),
mimic the actual study samples. Therefore reanalysis of study samples was
Zorbax Eclipse XDB‐C18 column (4.6 × 100 mm, 3.5 μm) and Zorbax SB‐C18
done to evaluate the accuracy of incurred samples. The percentage
column (4.6 × 50 mm, 5 μm) were tried. The C8 columns resulted in low
difference between the initial concentration and the concentration
resolution between SOF and matrix peaks or long retention time for DAC.
measured during the repeat analysis should not be more than 20% of their
Using Zorbax Eclipse XDB‐C18 column (4.6 × 100 mm, 3.5 μm) resulted in
mean.
long run time while the Zorbax SB‐C18 column (4.6 × 50 mm, 5 μm) was the
most suitable one since it produced symmetrical peaks with high resolution
and high sensitivity within a reasonable time of analysis of <2.5 min.
2.8 | Pharmacokinetic study Preliminary trials with mobile phase compositions containing different
The aim of this study is to investigate the pharmacokinetic parameters of ratios of organic and aqueous phases were tested in an isocratic mode.
SOF and DAC in three healthy male subjects. The study was conducted Different trials were carried out at different values of pH, 2–7, of the mobile
according to the Declaration of Helsinki for biomedical research involving phase, but all attempts were futile. A 0.1% aqueous solution of formic acid,
human subjects (https://www.wma.net/ policies‐post/wma‐declaration‐ ammonium acetate pH 2–6 with different ratios of acetonitrile and
of‐helsinki‐ethical‐principles‐for‐medical‐research‐involving‐human‐ methanol was tested; the results were unsatisfactory owing to high matrix
subjects/, last accessed May 2017), which indicates that all volunteers interference that leads to relative low responses, especially for SOF. It was
should be informed about the protocol, purpose and risk of the study then a challenge to find a suitable LC‐MS/MS method for both analytes owing to
give written consent to participate. In addition, Zi Diligence Research different physiochemical properties where they behave in different ways in
Center (Cairo, Egypt) reviewed and approved all experimental procedures. plasma extraction and chromatographic separation. The best conditions
Volunteers were prohibited from eating for 12 h before the study but water were achieved with isocratic elution using a ZorbaxSB‐C18 column (4.6 × 50
was freely available. Blood samples (3.0 mL) were collected from a forearm mm, 5 μm). A mixture of ammonium formate buffer (pH 3.5; 5 m M) and
vein into vacutainers containing K3EDTA at (pre‐dose) 0.00, 0.25, 0.50, 0.75, acetonitrile (50:50, v/v) was used as the mobile phase pumped at a flow
1.00, 1.25, 1.50, 1.75, 2.00, 2.50, 3, 4, 5, 6,8, 10, 12, 24, 36, 48 and finally rate of 0.7 mL min−1. The peaks achieved were well defined and resolved
72 h after simultaneous co‐administration of a single dose Sovaldi® 400 mg
ABDALLAH ET AL . 5 of 1
0

free from tailing with retention times 1.23, 1.09 and 1.72 min for SOF, DAC 3.4.3 | Carry‐over
and TAD (IS), respectively, as shown in (Figure 2). Blank plasma samples were injected after calibration standards at ULOQ
(3000 ng mL−1) for SOF and DAC. The carry‐over in the blank samples was
found to be <20% of LLOQ.
3.3 | Optimization of mass parameters
Both analytes and internal standard produced strong signals in the positive‐ 3.4.4 | Precision and accuracy

FIGURE 2 Mass chromatograms of (a) blank plasma, (b) blank plasma spiked with LLOQ and (c) plasma samples of subject at peak concentration
ion mode rather than the negative‐ion mode. Therefore, ESI+ (Cmax) after Assay run precision was determined as intra‐ and inter‐day precision, which
oral co‐administration of SOF/DAC, 400/60 tablets was performed using six replicates (n = 6) for intra‐day and 18 replicates (n
= 18) for inter‐day, expressed as RSD. The intra‐day and inter‐day precisions
ranged from 1.54 to 15.97% for the analytes.

mode was appropriate for the detection both analytes and IS. The The accuracy of the method expressed as RE was within ±10.06% for the

predominant protonated ions [M + H]+ of SOF, DAC and TAD (IS) in the Q1 analytes. The results are summarized in Table 3.

full‐scan MS spectra were at m/z 530.0, 739.5 and 390.2, respectively, as


shown in Figure 3. The collision energy and other MRM parameters were 3.4.5 | Extraction recovery and matrix effect
optimized. The following product ions were selected: 243.3, 565.3 and Matrix effect was evaluated using blank plasma samples obtained from six
268.2 for SOF, DAC and TAD (IS), respectively. different batches. The matrix factor for each lot was calculated by
comparing the analytes and (IS) peak areas in extracted plasma samples

3.4 | Method validation with those in the neat equivalent standard concentration. The IS‐
normalized matrix factor, CV %, was found not to exceed10.38% for both
3.4.1 | Selectivity
analytes, indicating that any enhancement from the human plasma should
Representative chromatograms are shown in Figure 3 and indicate that
be negligible. The relative recovery is the real recovery, which could be
there were no interference peaks observed from human plasma at the
calculated by comparing the area ratio response of extracted plasma
retention times of the analytes and IS. The retention times of SOF, DAC and
samples (spiked before extraction) with the post‐extraction (spiked after
TAD (IS) were 1.23, 1.09 and 1.72 min, respectively. Six plasma (n = 6)
extraction) samples. The recoveries across QCs ranged from 83.13 to
samples were extracted to demonstrate the sensitivity of the proposed
91.28% for both analytes and IS as presented inTable 4.
method.

3.4.6 | Stability
3.4.2 | Calibration curve and quantitation range
Stability of analytes in human plasma was evaluated under different
The calibration plots were found to be linear over the ranges of 0.3–3000
storage conditions at low and high concentrations with three
and 3.0–3000 ng mL−1 for SOF and DAC, respectively, as shown in Table 2.
determinations for each. Samples were exposed to different temperature
Blank and zero samples were assayed in order to verify the absence of
conditions for different time intervals and the results were compared with
interference. The accuracy values for LLOQ were acceptable, ranging from
those obtained from freshly prepared samples. Spiked plasma samples
97.50 to 100.20% for both analytes while precision was acceptable with
were stable after storage at room temperature for 24 h. The extracted
RSD < 0.96%.
samples were analyzed and found to be stable after keeping in the
autosampler at 4°C for 72 h. QC samples were frozen at −80°C and
6 of 1 ABDALLAH ET AL .
0

remained stable for 100 days. Samples were stable after three freeze–thaw 3.4.7 | Dilution integrity
cycles after storing at −80°C for 12 h and thawing unassisted at room Plasma samples were spiked at concentrations 4800 and 9600 ng mL−1 for
temperature. During repeated freezing and thawing, it was observed that
both SOF and DAC. Blank plasma samples were diluted 2‐ and 4‐fold at six
substance loss was negligible, as summarized in Table 5.
determinations per dilution. Precision RSD was between

FIGURE 3 Representative spectra for (a) SOF, (b)


DAC and (c) IS (tadalafil)

TABLE 2
Linearity results for SOF and DAC in human
plasma 2.35 and 4.51%, while accuracy results
ranged from 93.49 to 103.90%
Linear range for the analytes.
Analyte Run (ng mL−1) Linear equation R2
SOF 1 0.3–3000 y = 0.00140x + 0.00277 0.9984 3.4.8 | Incurred plasma samples
2 y = 0.00148x + 0.00317 0.9964 reanalysis
Reanalysis of study samples was done to evaluate
3 y = 0.00153x + 0.00128 0.9998
the accuracy of incurred samples. The percentage
DAC 1 3–3000 y = 0.00170x + 0.00104 0.9984 difference from initial concentration to the final
2 y = 0.00136x + 0.00214 0.9998 concentrations was measured during the repeat
analysis, and ranged from 4.56 to 14.07% for the
3 y = 0.00145x + 0.00113 0.9968
analytes, as shown inTable 6.

TABLE 3 Intra‐ and Inter‐day accuracy and precision results for SOF and (ng mL−1) RE (%) CV (%) RE (%) CV%
DAC
Concentration Intra‐day Inter‐day SOF LLOQ 0.3 −4.93 2.42 0.91 5.42
Analyte QCL 0.9 −2.57 3.57 1.52 3.40
ABDALLAH ET AL . 7 of 1
0

QCM 1200 10.06 1.54 4.20 4.46 4 | APPLICATION TO PHARMACOKINETIC


QCH 2400 7.86 3.85 2.52 8.19
STUDY

DAC LLOQ 3 0.34 3.98 −5.74 15.97 The validated LC‐MS/MS bioanalytical method was successfully applied to
QCL 9 7.25 4.67 6.52 9.02 determine SOF and DAC in human plasma for pharmacokinetic study in

QCM 1200 −6.85 2.16 0.36 4.78 three healthy male volunteers, following oral co‐administration of tablets
containing SOF (400 mg) and DAC (60 mg) in a fasting condition. The mean
QCH 2400 −6.23 4.34 −1.43 8.20
plasma concentration–time profiles are illustrated in (Figure 4). The
n 6 18 maximum plasma concentrations (Cmax) for SOF and DAC were 1837.65 ±
541.27 and 1308.19 ± 6.22 ng mL−1 and were achieved at 0.42 ± 0.12 and
1.63 ± 1.24 h, respectively. The area under the concentration–time curve
(AUC0–t) for SOF and DAC
TABLE 4 Matrix effect, relative recovery and process efficiency for SOF and DAC
Concentration Matrix effect Relative recovery Process efficiency
(ng mL−1)

ME (%) CV (%) RE (%) CV (%) PE (%) CV (%)


Analyte

SOF QCL 0.9 100.98(100.15)a 6.84 (3.96) 83.13 (89.61)b 12.45 (4.56) 84.15 (89.69)c 2.25 (2.65)

QCM 1200 99.64 (100.53)a 3.34 (2.60) 86.99 (83.91)b 4.40 (5.40) 86.66 (84.36)c 2.51 (6.46)

QCH 2400 98.11 (98.54)a 2.99 (2.29) 90.75 (88.32)b 5.52 (8.54) 89.01 (87.03)c 4.53 (9.24)

DAC QCL 9 98.21 (100.43)a 3.98 (8.19) 91.28 (85.89)b 6.49 (7.90) 89.48 (86.23)c 4.32 (5.39)

QCM 1200 97.52 (100.53)a 1.34 (2.60) 90.98 (83.91)b 6.90 (5.40) 88.71 (84.36)c 5.52 (6.46)

QCH 2400 98.34 (98.54)a 2.52 (2.29) 88.80 (88.32)b 1.35 (8.54) 87.32 (87.03)c 2.13 (9.24)

n 6 6 6

a
Values of matrix effect for internal standard. b
Values of relative recovery for internal standard.
c
Values of process efficiency for internal standard.

TABLE 5 Stability results for SOF and DAC in plasma at different conditions
Short‐term stability at room Freeze–thaw stability at Long term stability at Processed sample stability at
temperature (24 h) −80 °C (three cycles) −80 °C(100 days) 4 °C(72 h)

Concentration Accuracy CV Stability Accuracy CV Stability Accuracy CV Stability Accuracy CV Stability


Analyte (ng mL−1) (%) (%) (%) (%) (%) (%) (%) (%) (%) (%) (%) (%)
SOF QCL 0.9 100.90 7.81 102.38 101.36 4.82 102.84 101.92 4.55 103.41 96.66 4.72 90.99
QCH 2400 102.77 9.05 96.62 104.98 2.65 98.70 104.58 0.24 98.32 100.59 3.80 93.18

DAC QCL 9 103.05 4.13 95.70 103.57 4.31 96.19 105.02 7.21 97.54 108.85 4.05 104.58
QCH 2400 93.12 2.40 95.63 96.28 5.66 98.88 95.10 3.31 97.66 100.19 2.97 105.03

n 3 3 3 3

TABLE 6 Incurred sample reanalysis


SOF DAC
8 of 1 ABDALLAH ET AL .
0

Incurred Initial concentration Repeat concentration Difference


sample (ng mL−1) (ng mL−1) (%)
reanalysis Initial concentration Repeat concentration Difference
(ng mL−1) (ng mL−1) (%)
833.96 914.50 9.21 1098.02 1040.09 5.42
383.09 441.08 14.07 1312.58 1390.89 5.79
78.02 86.77 10.62 1200.02 1273.66 5.95
5.52 6.01 8.55 251.58 227.08 10.24
0.357 0.339 5.28 79.97 76.40 4.56
9.94 11.28 12.67 73.93 65.13 12.66

FIGURE 4 Mean plasma concentration (± SD) following co‐administration of single oral dose of SOF/DAC tablets contain 400/60 mg SOF/DAC:
(a) SOF; (B) DAC values from different reported studies ranged from 0.5 to 2 h, and it was
observed that there was no disparity between the obtained values in our
study compared with the other reported ones (https://www.accessdata.
TABLE 7 Pharmacokinetic parameters (results ± SD)
fda.gov/drugsatfda_docs/nda/2014/205834Orig1s000ClinPharmR. pdf;
Parameters SOF DAC
German, Mathias, Brainard, & Kearney, 2016; Kirby, Symonds, Kearney, &
Cmax (ng mL−1) 1837.65 ± 541.27 1308.19 ± 6.22
Mathias, 2015; Rezk, Bendas, et al., 2016; Smolders et al., 2016). For DAC,
Tmax (h) 0.42 ± 0.12 1.63 ± 1.24 Cmax, AUC0–∞, T1/2 and Tmax results from different reported studies showed

T1/2 (h) 1.56 ± 1.21 11.16 ± 1.71 values in the ranges 928–1396 ng mL−1, 9486–14,572 ng h mL−1, 11.9–15 h
and 0.75–2 h, respectively. The results were in close agreement with those
AUC0–t (ng h mL−1) 1322.14 ± 437.61 15,668.80 ± 1308.29
reported in our study
AUC0–∞(ng h mL−1) 1322.69 ± 436.90 15,825.55 ± 1394.77
(https://www.accessdata.fda.gov/drugsatfda_docs/nda/
Ke (h−1) 0.64 ± 0.50 0.06 ± 0.01 2015/206843Orig1s000ClinPharmR.pdf; Bifano et al., 2011; Kawakami et
CL [mg/(ng/mL h)] 5.33 ± 1.76 0.063 ± 0.006 al., 2016; Smolders et al., 2016). It was noted in the present study that there
Vz [mg h/(ng/mL h)] 807.95 ± 803.77 58.68 ± 0.09 is no apparent effect of SOF on DAC exposure, while DAC modestly
increases SOF exposure, with the results are in close agreement with the
Vss [mg h/(ng/mL h)] 246.84 ± 78.06 53.61 ± 3.82
earlier reported studies (Eley et al., 2013) as DAC and SOF are required with
Cmax, Maximum plasma concentration; Tmax, time to peak concentration; T1/2,
elimination half‐life; AUC, area under the concentration–time curve; Ke, co‐administration.
elimination constant; CL, clearance; Vz, Apparent volume of distribution during
terminal phase; Vss, Apparent volume of distribution at steady state.
5 | CONCLUSION
was 1322.14 ± 437.61 and 15,668.80 ± 1308.29 ng h mL−1, respectively. The
AUC0–∞ for SOF and DAC was 1322.69 ± 436.90 and 15,825.55 ± 1394.77 ng The present LC‐MS/MS method was fully validated according to US Food
h mL−1, respectively. The elimination half‐life (T1/2) for SOF and DAC was and Drug Administration guidelines and successfully applied to support
1.56 ± 1.21 and 11.16 ± 1.71, respectively, as presented in Table 7. pharmacokinetic and clinical studies. The new bio‐analytical method for
The presented values were compared with earlier reported studies for simultaneous determination of SOF and DAC is considered a reliable, robust
SOF and DAC separately. For SOF, the pharmacokinetic results reported and patient‐friendly and is expected to be used widely. The application of
from different studies with respect to Cmax, AUC0–∞ and T1/ 2 were in the incurred sample reanalysis in the validation protocol helped to ensure the
ranges 622–928.7 ng mL−1, 629–1202.47 ng h mL−1 and 0.4–1.24 h, validity of results and supports the use of the proposed method as a routine
respectively. Regarding Cmax values, it has been observed that there was a assay for bioequivalence studies, which will be very useful in therapeutic
significant increase in Cmax compared with that found in the literature when drug monitoring.
administered alone. AUC0–∞ increased slightly upon co‐administration with
DAC and the half‐life was slightly longer. Time to peak concentration (Tmax) ORCID
ABDALLAH ET AL . 9 of 1
0

Ahmed M. Abdel‐Megied http://orcid.org/0000-0003-0180-0918 hepatitis C virus NS5B polymerase inhibitor sofosbuvir. Clinical
Pharmacokinetics, 54(7), 677–690.
REFERENCES Matuszewski, B. K., Constanzer, M. L., & Chavez‐Eng, C. M. (2003). Strategies for
Abdallah, O. M., Abdel‐Megied, A. M., & Gouda, A. S. (2017). Development a the assessment of matrix effect in quantitative bioanalytical methods based
validated highly sensitive LC‐MS/MS method for simultaneous quantification of on HPLC‐MS/MS. Analytical Chemistry, 75(13), 3019–3030.
Ledipasvir, sofosbuvir and its major metabolite GS‐331007 in human plasma: Nannetti, G., Messa, L., Celegato, M., Pagni, S., Basso, M., Parisi, S. G., … Loregian,
Application to a human pharmacokinetic study. Journal of Pharmaceutical and A. (2017). Development and validation of a simple and robust HPLC method
Biomedical Analysis, 143, 305–310. Ariaudo, A., Favata, F., De Nicolo, A., Simiele, with UV detection for quantification of the hepatitis C virus inhibitor
M., Paglietti, L., Boglione, L., … D'Avolio, A. (2016). A UHPLC‐MS/MS method for daclatasvir in human plasma. Journal of Pharmaceutical and Biomedical
the quantification of direct antiviral agents simeprevir, daclatasvir, ledipasvir, Analysis, 134, 275–281.
sofosbuvir/GS331007, dasabuvir, ombitasvir and paritaprevir, together with
Nebsen, M., & Elzanfaly, E. (2016). Stability‐indicating method and LC‐MSMS
ritonavir, in human plasma. Journal of Pharmaceutical and Biomedical Analysis,
characterization of forced degradation products of sofosbuvir. Journal of
125, 369–375.
Chromatographic Science, 54(9), 1–10.
Azab, S. M., & Fekry, A. M. (2017). Electrochemical design of a new nanosensor
Palumbo, E. (2011). Pegylated interferon and ribavirin treatment for hepatitis C
based on cobalt nanoparticles, chitosan and MWCNT for the determination
virus infection. Therapeutic Advances in Chronic Disease, 2(1), 39–45.
of daclatasvir: A hepatitis C antiviral drug. RSC Advances, 7, 1118–1126.
Product leaflet (2013). https://www.accessdata.fda.gov/drugsatfda_docs/
Baker, M. M., El‐Kafrawy, D. S., Mahrous, M. S., & Belal, T. S. (2017). Validated label/2015/20467 1s002lbl.pdf (accessed 1 December 2017).
stability‐indicating HPLC‐DAD method for determination of the recently
Product monograph (2016). Daklinza (daclatasvir) prescribing information.
approved hepatitis C antiviral agent daclatasvir. Annales Pharmaceutiques
Qu, L., Wang, W., Zeng, D., Lub, Y., & Yin, Z. (2015). Quantitative performance of
Françaises, 75(3), 176–184.
online SPE‐LC coupled to Q‐Exactive for the analysis of sofosbuvir in human
Belema, M., Nguyen, V. N., Bachand, C., Deon, D. H., Goodrich, J. T., James, C. A.,
plasma. Journal of RSC Advances, 5, 98269–98277.
… Hamann, L. G. (2014). Hepatitis C virus NS5A replication complex
Rezk, M. R., Basalious, E. B., & Amin, M. E. (2016). Novel and sensitive UPLC‐
inhibitors: the discovery of daclatasvir. Journal of Medicinal Chemistry, 57(5),
MS/MS method for quantification of sofosbuvir in human plasma:
2013–2032. https://doi.org/10.1021/jm401836p.
Application to a bioequivalence study. Biomedical Chromatography, 30(9),
Bifano, M., Sevinsky, H., Persson, A., Chung, E., Wind‐Rotolo, M., Hwang, C., ...
1354–1362.
Bertz, R. (2011). Single‐dose pharmacokinetics of Daclatasvir (DCV; BMS‐
Rezk, M. R., Basalious, E. B., & Karim, I. A. (2015). Development of a sensitive
790052) in subjects with hepatic impairment compared with healthy
UPLC‐ESI‐MS/MS method for quantification of sofosbuvir and its metabolite,
subjects. Paper presented at the 62th Annual meeting of the American
GS‐331007, in human plasma: Application to a bioequivalence study. Journal
Association for the Study of Liver Diseases, San Francisco, CA.
of Pharmaceutical and Biomedical Analysis, 114, 97–104.
Bristol‐Myers‐Squibb (2014). Daklinza (daclatasvir) summary of product
https://doi.org/10.1016/j.jpba.2015.05.006.
characteristics. European Union.
Rezk, M. R., Bendas, E. R., Basalious, E. B., & Karim, I. A. (2016). Development and
Eley, T., You, X., Huang, S.‐P., Symonds, W., Sherman, D., Mathias, A., ... Bertz, R. validation of sensitive and rapid UPLC‐MS/MS method for quantitative
(2013). Evaluation of drug interaction potential between daclatasvir and determination of daclatasvir in human plasma: Application to a
sofosbuvir. Presented at 8th Interational workshop on clinical pharmacology bioequivalence study. Journal of Pharmaceutical and Biomedical Analysis,
of heapatits therapy, Cambridge. Retrieved from http://regist2.virology‐ 128, 61–66.
education.com/2013/8hepcam/docs/20_Bertz. pdf.
Shi, X., Zhu, D., Lou, J., Zhu, B., Hu, A. R., & Gan, D. (2015). Evaluation of a rapid
German, P., Mathias, A., Brainard, D., & Kearney, B. P. (2016). Clinical method for the simultaneous quantification of ribavirin, sofosbuvir and its
pharmacokinetics and pharmacodynamics of ledipasvir/sofosbuvir, a fixed‐ metabolite in rat plasma by UPLC‐MS/MS. Journal of Chromatography B:
dose combination tablet for the treatment of hepatitis C. Clinical Analytical Technology in Biomedicine and Life Sciences, 1002, 353–357.
Pharmacokinetics, 55(11), 1337–1351.
Smith, M. A., Regal, R. E., & Mohammad, R. A. (2016). Daclatasvir: A NS5A
Hassib, S. T., Taha, E. A., Elkady, E. F., & Barakat, G. H. (2017). Reversed‐phase replication complex inhibitor for hepatitis C infection. Annals of
liquid chromatographic method for determination of daclatasvir Pharmacotherapy, 50(1), 39–46.
dihydrochloride and study of its degradation behavior. Chromatographia,
Smolders, E. J., de Kanter, C. T., van Hoek, B., Arends, J. E., Drenth, J. P., & Burger,
80(7), 1101–1107.
D. M. (2016). Pharmacokinetics, efficacy, and safety of hepatitis C virus drugs
Jiang, H., Kandoussi, H., Zeng, J., Wang, J., Demers, R., Eley, T., … Arnold, M. E. in patients with liver and/or renal impairment. Drug Safety, 39(7), 589–611.
(2015). Multiplexed LC‐MS/MS method for the simultaneous quantitation of
Sofia, M. J., Bao, D., Chang, W., Du, J., Nagarathnam, D., Rachakonda, S., …
three novel hepatitis C antivirals, daclatasvir, asunaprevir, and beclabuvir in
Furman, P. A. (2010). Discovery of a beta‐d‐2′‐deoxy‐2′‐alpha‐fluoro2′‐beta‐
human plasma. Journal of Pharmaceutical and Biomedical Analysis, 107,
C‐methyluridine nucleotide prodrug (PSI‐7977) for the treatment of hepatitis
409–418.
C virus. Journal of Medicinal Chemistry, 53(19), 7202–7218.
Kandeel, A., Genedy, M., El‐Refai, S., Funk, A. L., Fontanet, A., & Talaat, M. (2017). Souri, E., Jalalizadeh, H., & Shafiee, A. (2007). Optimization of an HPLC method
The prevalence of hepatitis C virus infection in Egypt 2015: Implications for for determination of gabapentin in dosage forms through derivatization with
future policy on prevention and treatment. Liver International, 37(1), 45–53. 1‐fluoro‐2,4‐dinitrobenzene. Chemical & Pharmaceutical Bulletin (Tokyo),
Kawakami, Y., Imamura, M., Ikeda, H., Suzuki, M., Arataki, K., Moriishi, M., … 55(10), 1427–1430.
Chayama, K. (2016). Pharmacokinetics, efficacy and safety of daclatasvir plus Srinivasu, G., Kumar, K. N., & Thirupathi, C. (2016). Development and validation
asunaprevir in dialysis patients with chronic hepatitis C: Pilot study. Journal of the chiral HPLC method for daclatasvir in gradient elution mode on
of Viral Hepatology, 23(11), 850–856. amylose‐based immobilized chiral stationary phase. Chromatographia,
Kirby, B. J., Symonds, W. T., Kearney, B. P., & Mathias, A. A. (2015). 79(21), 1457–1467.
Pharmacokinetic, pharmacodynamic, and drug–interaction profile of the
1 of 1 ABDALLAH ET AL .
0 0

Thein, H., Yi, Q., Dore, G., & Krahn, M. (2008). Estimation of stage‐specific fibrosis
progression rates in chronic hepatitis C virus infection: A meta‐analysis and
meta‐regression. Hepatology, 48(2), 418–431.
US Food and Drug Administration. Guidance for Industry (2001). Bioanalytical
Method Validation. US Department of Health and Human Services, Food and
Drug Administration, Center for Drug Evaluation and Research and Center
for Veterinary Medicine.
WHO Prequalification Team – Medicines (2015). Guidance Document. Notes on
the Design of Bioequivalence Study. Retrieved from http://
apps.who.int/prequal/info_applicants /BE/2015/ Sofosbuvir_.
Zimmer, D. (2014). New US FDA draft guidance on bioanalytical method
validation versus current FDA and EMA guidelines: Chromatographic
methods and ISR. Bioanalysis, 6, 13–19.
How to cite this article: Abdallah OM, Abdel‐Megied AM, Gouda AS.
Development and validation of LC‐MS/MS method for
simultaneous determination of sofosbuvir and daclatasvir in human
Plasma: Application to pharmacokinetic study. Biomedical
Chromatography. 2018;e4186. https://doi.org/10.1002/

bmc.4186

You might also like