Next Article in Journal
Aesculus hippocastanum L.: A Simple Ornamental Plant or a Source of Compelling Molecules for Industry?
Next Article in Special Issue
A New UHPLC Analytical Method for St. John’s Wort (Hypericum perforatum) Extracts
Previous Article in Journal
Traditional Importance, Phytochemistry, Pharmacology, and Toxicological Attributes of the Promising Medicinal Herb Carissa spinarum L.
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

RP-HPLC-Based Bioanalytical Approach for Simultaneous Quantitation of Cinnarizine and Domperidone in Rat Plasma

1
School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
2
Government Pharmacy College, Kangra 176047, Himachal Pradesh, India
3
Bharati Vidyapeeth’s College of Pharmacy, Navi Mumbai 400614, Maharashtra, India
4
Sri Sai College of Pharmacy, Amritsar 143149, Punjab, India
5
Laureate Institute of Pharmacy, Kangra 176031, Himachal Pradesh, India
6
Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
7
Department of Pharmacy Practice, College of Pharmacy, AlMaarefa University, Ad Diriyah 13713, Saudi Arabia
8
Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
9
Department of Pharmaceutical Sciences, School of Applied Science and Technology, University of Kashmir, Srinagar 190006, Jammu and Kashmir, India
*
Author to whom correspondence should be addressed.
Separations 2023, 10(3), 159; https://doi.org/10.3390/separations10030159
Submission received: 13 February 2023 / Revised: 21 February 2023 / Accepted: 23 February 2023 / Published: 25 February 2023

Abstract

:
An accurate, precise and sensitive reverse-phase high-performance liquid chromatography (RP-HPLC) bioanalytical approach was developed for the simultaneous estimation of cinnarizine (CIN) and domperidone (DOM) in rat plasma using irbesartan (IRB) as an internal standard (IS). The proposed RP-HPLC approach was validated as per the latest ICH M10 guidelines. The analytes (CIN and DOM) and IS were extracted from plasma samples using the protein precipitation strategy. Chromatographic separation is accomplished by a C18 SunfireTM (5 µm, 250 mm × 4.6 mm) analytical column, using an isocratic mobile phase consisting of acetonitrile-methanol in 30:70 proportions at a flow rate of 1 mL/min. The detection of all three constituents was recorded at a wavelength of 270 nm with a UV detector. DOM, CIN and IS were eluted at 3.2, 4.5 and 6.1 min, respectively, utilizing a total run time of 10 min. The lower limit of quantification (LLOQ) was 5 ng/mL for CIN and DOM in rat plasma. The proposed RP-HPLC approach was linear in the 5–200 ng/mL range for CIN and DOM. The recovery of the method was greater than 95%, and the relative uncertainty was less than 2%, indicating that the proposed bioanalytical approach was accurate and precise. The limit of detection was established as 1.1 ng/mL for CIN and 1.7 ng/mL for DOM. The created approach was found to be robust and passed all validation criteria; thus, the proposed RP-HPLC approach can be employed successfully for the simultaneous assessment of CIN and DOM in rat plasma.

1. Introduction

Cinnarizine (CIN) is a piperazine derivative with histamine H1-, dopamine D2- and calcium-channel-blocking activity [1,2]. In addition to treating vertigo/Meniere’s disease, nausea, vomiting and motion sickness, it is also effective for vestibular symptoms due to other origins [2,3]. CIN inhibits vascular smooth muscle cell contractions by inhibiting L-type and T-type voltage-gated calcium channels [3]. CIN may alleviate motion sickness vomiting by binding to dopamine D2 receptors, histamine H1 receptors and muscarinic acetylcholine receptors [4]. It works by interfering with signal transmission between the inner ear’s vestibular apparatus and the hypothalamus’ vomiting center [4,5].
Domperidone (DOM) is a specific blocker of dopamine receptors with antiemetic and gastric prokinetic properties, which can be used to relieve motility disorders [6,7]. It is frequently employed as an antiemetic medicine for the temporary relief of nausea and vomiting brought on by Parkinson and anticancer medication. The dopamine-receptor-blocking activity of DOM at the stomach level as well as in the chemoreceptor trigger zone is connected to its antiemetic characteristics. It possesses strong affinity for the chemoreceptor trigger zone’s D2 and D3 dopamine receptors. The peripheral dopamine-receptor-blocking activities of DOM are associated with its gastroprokinetic characteristics [8]. By promoting esophageal and gastric peristalsis and reducing esophageal sphincter pressure, DOM speeds up gastric emptying and shortens small bowel transit time [9]. Both of these drugs are individually available in the market and have the therapeutic potential to treat the conditions of nausea vomiting/motion sickness. There has been a clinical trial performed by Oosterveld wherein he reported that the combination of CIN and DOM is more effective than individual drugs [10]. The combination has found acceptance since then, and many marketed products, such as Stugil tablets by Eris Lifesciences Ltd., Vertigil tablet by Cipla Ltd. and Stetidom tablets by Dr. Morepen Ltd., are available in the market, which have a dose combination of 20 mg of CIN and 15 mg of DOM to produce a synergistic effect. Both drugs (CIN and DOM) belong to biopharmaceutical classification system (BCS) II drugs, which means they have poor solubility and good permeability [1,6]. Considering the synergistic impact of these drugs, their simultaneous quantification in plasma is highly beneficial.
A literature survey reveals that several methods were reported for the simultaneous estimation of DOM and CIN in bulk and dosage forms by the reverse-phase high-performance liquid chromatography (RP-HPLC) method [2,11,12]. Nevertheless, to the best of our knowledge, no RP-HPLC approach for the simultaneous measurement of CIN and DOM in rat plasma has yet to be published. The measurement of CIN and DOM alone in rat or human plasma, however, has been the subject of several pieces of research using HPLC with ultraviolet (UV) and fluorescence detection [13,14]. DOM in human plasma can be detected using an RP-HPLC method that was established in 2000 [13]. The mobile phase was given at a flow rate of 1.4 mL/min and was composed of methanol, water, triethylamine and acetic acid (60:40:0.02:0.3, v/v). A correlation coefficient (R2) value of 0.9998 revealed that the procedure is linear in the range of 1–20 ng/mL. The limit of quantification (LOQ) for DOM was determined to be 1 ng/mL, whereas the limit of detection (LOD) was found to be 0.2 ng/mL [13]. The HPLC method was also created as a bioanalytical approach in a different investigation to estimate the level of CIN in human plasma. The mobile phase was given at a flow rate of 1 mL/min and included 0.01 M ammonium dihydrogen phosphate buffer of pH 4.2 and a 0.038% triethylamine and acetonitrile 25:75 (v/v) mixture. Between a 1 and 100 ng/mL concentration range, the calibration curve was linear. CIN’s LOD was established to be 1.25 ng/mL. The procedure was validated for accuracy, precision, specificity, recovery and stability. For the pharmacokinetic analysis of CIN in human plasma samples, this approach was found to be accurate, precise, straightforward and reliable [14]. CIN and its metabolites were estimated in human plasma using a liquid chromatography tandem mass spectrometry (MS)/MS (LC-MS/MS) approach in 2018 by Mandal et al. CIN and the internal standard (metoprolol) had chromatographic elution times of 2.32 and 2.05 min, respectively, in a very brief run time of 3.50 min [15]. In 2016, Khan et al. created an easy, quick and accurate RP-HPLC/UV technique employing tenofavir as the internal standard for the simultaneous detection of DOM and itopride in pharmaceutical samples and human plasma. Pumped at a flow rate of 1.5 mL/min, the mobile phase was a mixture of water (pH 3.0) and acetonitrile (65:35 v/v), and peaks were observed at a wavelength of 210 nm. This approach was linear in the 20–600 ng/mL range. The LOD and LOQ for DOM were 5 and 10 ng/mL, respectively. DOM fast-dissolving tablets were successfully analyzed in vivo using this newly established technique in healthy human volunteers [16]. Over the last decade, numerous RP-HPLC methods were developed for the simultaneous estimation of CIN and DOM from their market formulations [2,11,12]. However, these methods are only applicable to the quantification of these drugs in nonbiological fluids. Over time, a few RP-HPLC approaches have also been established for the quantification of CIN and DOM in biological fluids, but only for individual quantification, as these approaches are not able to quantify these two drugs simultaneously. To understand the pharmacokinetic profile of this drug combination, their simultaneous quantification in biological fluid, especially in plasma, is a prerequisite. To accomplish this goal, a simultaneous RP-HPLC-based bioanalytical approach was created and validated for the measurement of these medications in rat plasma. In this current research, successful validation is performed according to the ICH guidelines (M10) 2019 in terms of parameters including the lower limit of quantification (LLOQ), linearity, accuracy, precision, matrix specificity and drugs’ stability in plasma [17,18].
The simultaneous assessment of a mixture of CIN and DOM in rat plasma has not been reported. Therefore, for the purpose of determining CIN and DOM in plasma simultaneously, we created and validated an isocratic HPLC assay with UV detection. The technique may be effective for pharmacokinetic profile determination, active ingredient identification in tablet dosage forms and drug monitoring.

2. Materials and Methods

2.1. Materials and Equipment

CIN and DOM were provided as complimentary samples from Ankur Pharmaceuticals Ltd. (Baddi, India). Irbesartan (IRB) was obtained from Helix Pharmaceutical (Nalagarh, India). HPLC-grade acetonitrile and methanol were provided by E-Merck (Mumbai, India). In this work, triple-distilled water that has been purified using a Milli-Q water purification system to make it HPLC-grade water (conductivity < 1.0 µS/cm and resistivity = 18.2 MΩ) was used. For quantitative analysis, an HPLC system (Shimadzu SPD-20A, Tokyo, Japan) with a UV/visible detector and a 20 µL loop (Rheodyne) outfitted with an LC-20AT pump was employed. The cooling centrifuge and vortex mixer were from REMI (New Delhi, India).

2.2. Animals

The current investigation utilized six Wistar male rats. All of the rats ranged in age from 6 to 8 weeks, and they were all between 250 and 300 g in weight. The rats were housed in husk-lined polypropylene cages with a 12:12 light: dark cycle at a 25 °C temperature with a relative humidity of 55%. Water and a typical pellet meal were given to the animals as food. The Pinnacle Biomedical Research Institute’s Institutional Animal Ethics Committee (PBRI/IAEC/29-03/010), Bhopal, India, gave its approval to the study protocol.

2.3. Chromatographic Conditions

A bioanalytical approach was created for the simultaneous measurement of CIN and DOM in rat plasma following the ICH M10 recommendations [17,18]. The separation of CIN and DOM was achieved using a reverse-phase analytical column (C-18 SunfireTM, 5 µm; 250 × 4.6 mm) and was maintained at 30 ± 1 °C. Acetonitrile and methanol were the components of the mobile phase in a ratio of 30:70 v/v, which was pumped at a flow rate of 1 mL/min in an isocratic mode. IRB served as an internal benchmark (IS). A sample volume of 20 μL was injected in triplicate onto the HPLC column. A UV/Vis detector operating at 270 nm was used to find both analytes and the IS.

2.4. Preparation of Blank Plasma

Due to the reduced steps required to extract the drug from the matrix, protein precipitation was the chosen method of separation [19,20]. To separate the plasma proteins, 1 mL of plasma was mixed with 2 mL of a mixture of methanol and acetonitrile (1:1 v/v). The mixture was then agitated for 5 min. The supernatant was taken out and spun in a cooling centrifuge for 15 min at 10,000 rpm. The supernatant was filtered with a syringe filter before being transferred to a volumetric flask with a 100 mL capacity and diluted with the mobile phase to obtain a total of 100 mL of the mobile phase, which was spiked with 1% deproteinized plasma [21].

2.5. Preparation of Standard Stock Solution

CIN and DOM (each 100 mg) were weighed precisely and transferred to individual 100 mL volumetric flasks containing 20 mL of the mobile phase. The solutions were sonicated for 5 min, and the volume was adjusted to 100 mL with the mobile phase to yield a 1000 μg/mL concentration of CIN (solution A) and DOM (solution A1). To achieve a concentration of 100 μg/mL of each drug, a 10 mL aliquot was withdrawn from each flask into a 100 mL standard volumetric flask, and the volume was adjusted to 100 mL using the mobile phase to obtain solution B and B1. In addition, a 10 mL aliquot from each solution, B and B1, was transferred to 100 mL standard volumetric flasks, and the volume was adjusted to 100 mL using the mobile phase to yield a 10 μg/mL concentration of CIN and DOM (solution C and C1). A total of 10 mL of aliquots from solution C and C1 were transferred to a 100 mL standard volumetric flask, and the volume was adjusted to 100 mL with the mobile phase to achieve a concentration of 1 μg/mL (1000 ng/mL) (solution D and D1). By adding several stock solutions to rat plasma, calibrations and quality control solutions were created. For CIN and DOM in plasma, the calibration curve range was 5–200 ng/mL.

2.6. Preparation of Stock Solution of IS

IRB 10 mg was precisely weighed, dissolved in the mobile phase and transferred to a volumetric flask of 100 mL. The solution was sonicated for 15 min and diluted with the mobile phase to create a solution with a concentration of 100 μg/mL (solution E). To obtain a final solution with a concentration of 10 μg/mL (solution F), an aliquot of 10 mL from solution E was withdrawn in a 100 mL standard volumetric flask, and the volume was adjusted to 100 mL using the mobile phase.

2.7. Method Validation Procedures

2.7.1. LLOQ

Five plasma replicates spiked with CIN and DOM at LLOQ were used to validate the LLOQ. According to US FDA guidelines, LLOQ is estimated as 5% of peak plasma concentration (Cmax). Published studies showed plasma Cmax levels of CIN and DOM up to 100 ng/mL. The coefficient of variation (CV) needs to be under 20%. The lowest calibration standard is known as LLOQ. Furthermore, the LLOQ sample’s analyte signal needs to be at least five times that of a blank sample [22].

2.7.2. Method Specificity

A total of 0.5 mL of blank plasma with a concentration of 1 µg/mL was pipetted out and diluted to 100 mL, obtaining a blank plasma concentration of 5 ng/mL. The method specificity was assessed by injecting 20 µL of this plasma as a blank sample into the HPLC system. Moreover, 5 ng/mL solutions (at the LLOQ level) for CIN and DOM were individually produced from the stock solution in the mobile phase with the spiked matrix. The 50 ng/mL solutions of CIN-DOM (with plasma) were made by diluting 0.5 mL of the 1 µg/mL stock solutions D and D1 to 10 mL. From these solutions, a 1 mL aliquot was pipetted into two 10 mL standard volumetric flasks, 1 mL of the IS from its stock solution (100 µg/mL) was added and the volume of each flask was adjusted to 10 mL to achieve a final concentration of 5 ng/mL for CIN and DOM and 10 µg/mL for the IS. At 270 nm, 20 µL of these solutions were put into the HPLC system and evaluated [23].

2.7.3. Calibration Curves

The seven-point calibration curves for CIN and DOM in the range of 5–200 ng/mL were prepared using aliquots (0.05, 0.1, 0.25, 0.5, 1.0, 1.5 and 2.0 mL) from stock solution (D and D1) having a 1 µg/mL concentration of both drugs. To each of these dilutions, 1 mL of solution F (IRB, 10 µg/mL) and 0.5 mL of rat plasma were added. To obtain concentrations of 5, 10, 25, 50, 100, 150 and 200 ng/mL, the samples were reconstituted to 10 mL using the mobile phase and injected into the HPLC system for measurement at 270 nm. To determine the linearity of the analytical approach, the slope, intercept and R2 of the calibration curves (peak area versus concentration) were calculated [24].

2.7.4. Accuracy Study

A study of accuracy was carried out based on the absolute recovery at each of the four distinct quality control (QC) levels for both samples (CIN and DOM). These levels are labeled as the lower limit of quantification (LLOQ), lower quantified concentration (LQC), medium quantified concentration (MQC) and high quantified concentration (HQC). The values for the LLOQ, LQC, MQC and HQC were 5, 15, 100 and 150 ng/mL, respectively (for CIN and DOM each). The IS was added to all of these solutions, and its concentration was maintained at 10 µg/mL throughout. The accuracy was measured in terms of the percentage of CIN and DOM that was successfully recovered from the solution. The experiment was repeated six times, and the average data were recorded. By using equation 1, we were able to determine the percentage of absolute recovery. The mean, standard deviation (SD) and % CV were calculated at each QC level. Accuracy within 85–115% of the nominal values was considered acceptable [18].
A b s o l u t e   r e c o v e r y % = A c t u a l   c o n c e n t r a t i o n   r e c o v e r e d T h e o r i t i c a l   c o n c e n t r a t i o n × 100

2.7.5. Precision Studies

The intraday, interday and interanalyst precisions were determined at four distinct QC levels, i.e., LLOQ, LQC, MQC and HQC. The IS was added to each of these solutions, and its concentration was maintained at 10 µg/mL. In order to conduct an intraday precision (repeatability) study, the solutions of LLOQ, LQC, MQC and HQC were injected into six replicates on the same day. The same solutions were injected on three distinct days to determine interday precision. To determine the interanalyst precision, these samples were administered in six replicates by three different investigators on the same day. They had their mean, SD and % CV computed [25].

2.7.6. LOD and LOQ

Equations (2) and (3) were used, respectively, to obtain the LOD and LOQ for CIN and DOM using the “linear regression equation based on the standard deviation of the intercept and the slope” [26].
L O D = 3.3 σ S
L O Q = 10 σ S
Here, σ is the standard deviation of the response and S is the slope of the calibration curve.

2.7.7. System Suitability

For the purpose of determining the system appropriateness characteristics, which include the theoretical plates number, tailing factor, resolution and height equivalent to a theoretical plate (HETP), six replicates were injected with the lowest concentration of drugs in the calibration curve (LLOQ, i.e., 5 ng/mL).

2.7.8. Stability Study

Plasma samples spiked with CIN and DOM were subjected to three freeze–thaw cycles, short-term stability studies and long-term stability studies. For freeze–thaw stability, 3 mL of plasma was collected in a single vial. To achieve a concentration of 1000 µg/mL, 3 mg of CIN and DOM were added to this vial, and the mixture was vortexed for 5 min. This test tube was stored at −20 °C in a deep freezer. After the sample was frozen, the test tube was removed and allowed to defrost at room temperature. A total of 1 mL of plasma was extracted from the thawed samples (cycle 1), and the remaining 2 mL was placed in the deep freezer for the second cycle. The drug-containing plasma (1 mL) was precipitated, and the supernatant was centrifuged. After centrifugation, the clear, translucent supernatant was extracted and diluted to 1000 mL with the mobile phase to reach a 1 µg/mL concentration. In addition, dilutions were produced to obtain concentrations of 15 ng/mL (LQC), 100 ng/mL (MQC) and 150 ng/mL (HQC). Likewise, the remaining frozen plasma sample (2 mL) was removed, thawed at room temperature and 1 mL was extracted (cycle 2), while the remaining 1 mL of thawed plasma was returned to the deep freezer. It was taken out and thawed after it had frozen (cycle 3). As in cycle 1, the method was repeated in cycles 2 and 3 to prepare LQC, MQC and HQC samples. The IS was added to all of these solutions at a concentration of 10 µg/mL. All dilutions were produced in triplicate, injected into HPLC and evaluated. For each concentration, the mean, SD and % CV were computed [18,27].
Similarly, the short-term stability of plasma samples spiked with CIN and DOM at room temperature was assessed. The stability was tested 1 h, 2 h and 3 h before extraction. In the short term, 3 mL of plasma was placed in a vial, 3 mg of CIN and DOM were added (to achieve a concentration of 1000 µg/mL) and the mixture was vortexed for 5 min. Room temperature was maintained for the vial. After each interval, a sample (1 mL) was taken, drugs were removed from plasma and processed for the manufacture of LQC, MQC and HQC samples, and the IS (10 µg/mL) was added. All dilutions were produced in triplicate, injected into HPLC and evaluated. For each concentration, the mean, SD and % CV were computed. For long-term stability, 1 mL of plasma was obtained in three vials, and 1 mg of CIN and DOM were added to each vial (to achieve a concentration of 1000 µg/mL). The mixture was vortexed for 5 min, and all three vials were frozen at −20 °C. After one, two and three weeks, the three vials were removed from the freezer. After each interval, the medicines were removed from plasma and processed for the preparation of LQC, MQC and HQC samples, and the IS (10 µg/mL) was added. All dilutions were produced in triplicate, injected into HPLC and evaluated. For each concentration, the mean, SD and % CV were computed [28].

3. Results and Discussion

3.1. Determination of LLOQ

Based on the results, the LLOQ of CIN and DOM was found at 5 ng/mL with %CV values of 3.78% and 3.61%, respectively. These values of %CV for both drugs met the requirements (i.e., not more than 20%); hence, this concentration (5 ng/mL) is elected as the LLOQ. The chromatogram is shown in Figure 1.

3.2. Chromatograms of the Mixture Containing DOM, CIN and IRB

The chromatogram of DOM, CIN and IRB prepared in the mobile phase is shown in Figure 2. Compounds with structurally similar analogues and similar physiochemical properties and functional groups could be used as ISs according to the literature [29,30]. CIN and DOM belong to BCS class II drugs [1,6]. IRB also belongs to BCS class II drugs, having a similar solubility and permeability profile. Furthermore, the molecular weight of IRB is in the same range as that of analytes. Both the solubility and molecular weight of analytes play a vital role in the separation and retention time of analytes [29]. As a result, we have selected IRB as the IS on the bases of physiochemical properties [29,30]. The retention times for DOM, CIN and IRB were found to be 3.20, 4.57 and 6.1 min, respectively.

3.3. Specificity Studies

Figure 3 displays the chromatogram of blank plasma. It was clear that the plasma matrix had no impact on the quantification of any drug extracted from plasma because there was no peak at the retention times (RTs) of DOM, CIN and IRB. Similar results were obtained when blank rat plasma was spiked with CIN and DOM at LLOQ (5 ng/mL) and blank rat plasma was spiked with IRB at 10 µg/mL, demonstrating the lack of any plasma peak in the retention periods of both drugs (Figure 3). Thus, the method is considered specific and selective.

3.4. Calibration Curves for CIN and DOM

Excellent linearity was evident in the calibration curves for both drugs produced in plasma, with R2 values for CIN and DOM of 0.999 and 0.998, respectively. The linearity range was 5–200 ng/mL for both drugs. The LLOQ for CIN and DOM was found to be 5 ng/mL at the lowest concentration with a CV value of less than 20%.

3.5. Accuracy Studies

The accuracy was determined by calculating the percentage recovery of both drugs in plasma. The % recovery of CIN and DOM in plasma was measured to be higher than 95%, showing that the proposed bioanalytical approach is accurate (Table 1). It is worth noting that the % CV in each case was less than 2%, indicating that the findings were repeatable.

3.6. Precision Studies

CIN and DOM in plasma were studied for intraday, interday and interanalyst precision (Table 2a,b). The % CV of the measured responses of LQC, MQC and HQC samples was less than 2%, suggesting that the procedure was precise.

3.7. LOD and LOQ

In plasma samples, the LOD and LOQ for CIN were determined to be 1.11 and 3.36 ng/mL, respectively. It was determined that the LOD and LOQ for DOM in plasma samples were 1.70 and 5.16 ng/mL, respectively. These results suggested that the approach was sensitive enough to detect both drugs at lower concentrations.

3.8. System Suitability

The tailing factor for both CIN and DOM peaks never exceeded 1 in any of the peaks, which demonstrated good peak regularity (acceptance limits were less than 2). Additionally, the number of theoretical plates for CIN and DOM was more than 50,000 in all runs, whereas the HETP value for CIN was 26.89 and 28.17 for DOM. These parameters’ values ensured excellent column efficiency throughout the development procedure.

3.9. Stability Study of Plasma Samples

For spiking samples of CIN and DOM in plasma, stability studies were performed at three different levels: short term (Table 3a), freeze–thaw cycles (Table 3b) and long term (Table 3c) at LQC, MQC and HQC levels. The results showed that more than 95 % of both drugs were recovered in all instances, with a % CV of less than 2%. The findings of these investigations pointed to the stress and long-term storage stability of both compounds in rat plasma.

4. Conclusions

CIN and DOM can be estimated simultaneously in rat plasma using a straightforward, isocratic RP-HPLC-based bioanalytical approach that has been devised and effectively validated. The approach that was devised is extremely specialized, accurate, precise and repeatable. The technique uses a very small amount of the plasma sample for analysis and requires no complicated sample preparation. According to the ICH’s most recent M10 recommendations, all validation parameters were within allowable ranges. Most importantly, the new technique has set a standard that researchers can utilize for a variety of in vivo studies, including those on pharmacokinetics, drug distribution, plasma protein binding and drug metabolism.

Author Contributions

Conceptualization, M.V., N.D. and P.W.; methodology, M.V., N.D., L.K., A.A. and P.W.; software, S.U.D.W., M.M.G. and F.S.; validation, F.S., S.A. and P.A.; formal analysis, S.U.D.W. and P.A.; investigation, M.V., N.D., L.K., A.A. and P.W.; resources, S.A.; data curation, M.M.G. and P.A.; writing—original draft preparation, M.V.; writing—review and editing, F.S., S.A. and P.A.; visualization, S.A.; supervision, P.W. and N.D.; project administration, P.W.; funding acquisition, S.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Researchers Supporting Project (number RSP2023R146) at King Saud University, Riyadh, Saudi Arabia, and “The APC was funded by RSP”.

Institutional Review Board Statement

The study protocol was approved by the Institutional Animal Ethics Committee of Pinnacle Biomedical Research Institute (PBRI), Bhopal, India (Protocol No. PBRI/IAEC/29-03/010; approval date: 28 March 2022).

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors are thankful to the Researchers Supporting Project (number RSP2023R146) at King Saud University, Riyadh, Saudi Arabia, for supporting this research. The authors are also thankful to AlMaarefa University for their generous support for this article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Shakeel, F.; Kazi, M.; Alanazi, F.K.; Alam, P. Solubility of cinnarizine in (Transcutol + water) mixtures: Determination, Hansen solubility parameters, correlation, and thermodynamics. Molecules 2021, 26, E7052. [Google Scholar] [CrossRef]
  2. Kumari, S.A.; Naga, S.M. Validated RP-HPLC method for simultaneous estimation of cinnarizine and domperidone in bulk and pharmaceutical dosage form. J. Pharm. Sci. Innov. 2013, 2, 46–50. [Google Scholar]
  3. Martí-Massó, J.F.; Poza, J.J. Cinnarizine-induced parkinsonism: Ten years later. Mov. Disord. 1998, 13, 453–456. [Google Scholar] [CrossRef]
  4. Emanuel, M.B.; Will, J.A. Cinnarizine in the treatment of peripheral vascular disease: Mechanisms related to its clinical action. Proc. R. Soc. Med. 1977, 70, 7–12. [Google Scholar] [CrossRef] [Green Version]
  5. Kirtane, M.V.; Bhandari, A.; Narang, P.; Santani, R. Cinnarizine: A contemporary review. Indian J. Otolaryngol. Head Neck Surg. 2019, 71, 1060–1668. [Google Scholar] [CrossRef]
  6. Shazly, G.A.; Alshehri, S.; Ibrahim, M.A.; Tawfeek, H.M.; Razik, J.A.; Hassan, Y.A.; Shakeel, F. Development of domperidone solid lipid nanoparticles: In vitro and in vivo characterization. AAPS PharmSciTech 2018, 19, 1712–1719. [Google Scholar] [CrossRef] [PubMed]
  7. Champion, M.C.; Hartnett, M.; Yen, M. Domperidone, a new dopamine antagonist. Can. Med. Assoc. J. 1986, 135, 457–461. [Google Scholar]
  8. Barone, J.A. Domperidone: Mechanism of action and clinical use. Hosp. Pharm. 1998, 33, 191–197. [Google Scholar]
  9. Orihata, M.; Sarna, S.K. Contractile mechanisms of action of gastroprokinetic agents: Cisapride, metoclopramide, and domperidone. Am. J. Physiol-Gastrointest. Liver Physiol. 1994, 266, G665–G676. [Google Scholar] [CrossRef]
  10. Oosterveld, W.J. The combined effect of cinnarizine and domperidone on vestibular susceptibility. Aviat. Space Environ. Med. 1987, 58, 218–223. [Google Scholar] [PubMed]
  11. Kalyankar, T.M.; Kulkarni, P.D.; Panchakshari, P.P.; Narute, A.S. Simultaneous RP-HPLC estimation of cinnarizine and domperidone in tablet. Res. J. Pharm. Technol. 2014, 7, 650–654. [Google Scholar]
  12. Argekar, A.P.; Shah, S.J. Simultaneous determination of cinnarizine and domepiridone maleate from tablet dosage form by reverse phase ion pair high performance liquid chromatography. J. Pharm. Biomed. Anal. 1999, 19, 813–817. [Google Scholar] [CrossRef] [PubMed]
  13. Kobylińska, M.; Kobylińska, K. High-performance liquid chromatographic analysis for the determination of domperidone in human plasma. J. Chromatogr. B 2000, 744, 207–212. [Google Scholar] [CrossRef] [PubMed]
  14. Nowacka-Krukowska, H.; Rakowska, M.; Neubart, K.; Kobylińska, M. High-performance liquid chromatographic assay for cinnarizine in human plasma. Acta Pol. Pharm. 2007, 64, 407–411. [Google Scholar] [PubMed]
  15. Mandal, P.; Dan, S.; Bose, A. LC-MS/MS method development and validation of an antihistaminic, calcium channel blocker, di-phenyl-methyl-piperazine group containing cinnarizine in human plasma with an application to BA/BE studies in Indian volunteer. Pharm. Pharmacol. Int. J. 2018, 6, 475–482. [Google Scholar]
  16. Khan, A.; Iqbal, Z.; Khadra, I.; Ahmad, L.; Khan, A.; Khan, M.I.; Ullah, Z. Simultaneous determination of domperidone and Itopride in pharmaceuticals and human plasma using RP-HPLC/UV detection: Method development, validation and application of the method in in-vivo evaluation of fast dispersible tablets. J. Pharm. Biomed. Anal. 2016, 121, 6–12. [Google Scholar] [CrossRef] [PubMed]
  17. Harahap, Y.; Azizah, N.; Andalusia, R. Simultaneous analytical method development of 6-mercaptopurine and 6-methylmercaptopurine in plasma by high performance liquid chromatography-photodiode array. J. Young Pharm. 2017, 9, S29–S34. [Google Scholar] [CrossRef] [Green Version]
  18. Khursheed, R.; Wadhwa, S.; Kumar, B.; Gulati, M.; Gupta, S.; Chaitanya, M.; Kumar, D.; Jha, N.K.; Gupta, G.; Prasher, P.; et al. Development and validation of RP-HPLC based bioanalytical method for simultaneous estimation of curcumin and quercetin in rat’s plasma. S. Afr. J. Bot. 2022, 149, 870–877. [Google Scholar] [CrossRef]
  19. Elzayat, E.M.; Shakeel, F.; Alshehri, S.; Ibrahim, M.A.; Altamimi, M.A.; Kazi, M.; Alanazi, F.K.; Haq, N. UHPLC assisted simultaneous separation of apigenin and prednisolone and its application in the pharmacokinetics of apigenin. J. Chromatogr. B 2019, 1117, 58–65. [Google Scholar] [CrossRef]
  20. Alam, P.; Iqbal, M.; Foudah, A.I.; Alqarni, M.H.; Shakeel, F. Quantitative determination of canagliflozin in human plasma samples using a validated HPTLC method and its application to a pharmacokinetic study in rats. Biomed. Chromatogr. 2020, 34, E4929. [Google Scholar] [CrossRef]
  21. Mannemala, S.S.; Nagarajan, J.S.K. Development and validation of a HPLC-PDA bioanalytical method for the simultaneous estimation of aliskiren and amlodipine in human plasma. Biomed. Chromatogr. 2015, 29, 346–352. [Google Scholar] [CrossRef] [PubMed]
  22. Bhinge, S.D.; Malipatil, S.M.; Sonawane, L.V. Bioanalytical method development and validation for simultaneous estimation of cefixime and dicloxacillin by RP-HPLC in human plasma. Acta Chim. Slov. 2014, 61, 580–586. [Google Scholar] [PubMed]
  23. Police, A.; Gurav, S.; Dhiman, V.; Zainuddin, M.; Bhamidipati, R.; Rajagopal, S.; Mullangi, R. Development and validation of an RP-HPLC method for the quantitation of odanacatib in rat and human plasma and its application to a pharmacokinetic study. Biomed. Chromatogr. 2015, 29, 1664–1669. [Google Scholar] [CrossRef]
  24. Eswarudu, M.M.; Rao, A.L.; Vijay, K. Bioanalytical method development and validation for simultaneous determination of chlorthalidone and cilnidipine drugs in human plasma by RP-HPLC. Int. J. Res. Pharm. Chem. 2019, 9, 33–44. [Google Scholar] [CrossRef]
  25. Talele, G.S.; Porwal, P.K. Development of validated bioanalytical HPLC-UV method for simultaneous estimation of amlodipine and atorvastatin in rat plasma. Indian J. Pharm. Sci. 2015, 77, 742–750. [Google Scholar]
  26. Dange, Y.; Bhinge, S.; Salunkhe, V. Optimization and validation of RP-HPLC method for simultaneous estimation of palbociclib and letrozole. Toxicol. Mech. Methods. 2018, 28, 187–194. [Google Scholar] [CrossRef]
  27. Maneesh, M.U.; Ahmed, S.S.; Pasha, T.Y.; Ramesh, B.; Majumder, M. A simple bioanalytical method for simultaneous estimation of amlodipine and celecoxib in rat plasma by high performance liquid chromatography. J. Chromatogr. Sci. 2021, 59, 627–633. [Google Scholar] [CrossRef] [PubMed]
  28. Kumar, R.; Kumar, R.; Khursheed, R.; Awasthi, A.; Khurana, N.; Singh, S.K.; Khurana, S.; Sharma, N.; Gunjal, P.; Kaur, J. Development and validation of RP-HPLC method for estimation of fisetin in rat plasma. S. Afr. J. Bot. 2021, 140, 284–289. [Google Scholar] [CrossRef]
  29. Yang, S.; Mu, L.; Feng, R.; Kong, X. Selection of internal standards for quantitative matrix-assisted laser desorption/ionization mass spectrometric analysis based on correlation coefficients. ACS Omega 2019, 4, 8249–8254. [Google Scholar] [CrossRef] [Green Version]
  30. Drotleff, B.; Lammerhofer, M. Guidelines for selection of internal standard-based normalization strategies in untargeted lipidomic profiling by LC-HR-MS/MS. Anal. Chem. 2019, 91, 9836–9843. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Reverse-phase high-performance liquid chromatography (RP-HPLC) chromatogram of DOM and CIN in plasma at LLOQ concentration with IRB as internal standard.
Figure 1. Reverse-phase high-performance liquid chromatography (RP-HPLC) chromatogram of DOM and CIN in plasma at LLOQ concentration with IRB as internal standard.
Separations 10 00159 g001
Figure 2. RP-HPLC chromatogram of DOM, CIN and IRB in the mobile phase.
Figure 2. RP-HPLC chromatogram of DOM, CIN and IRB in the mobile phase.
Separations 10 00159 g002
Figure 3. RP-HPLC chromatographs of (a) blank rat plasma, (b) blank rat plasma spiked with IS and (c) blank rat plasma spiked with CIN and DOM at LLOQ (5 ng/mL) and IS.
Figure 3. RP-HPLC chromatographs of (a) blank rat plasma, (b) blank rat plasma spiked with IS and (c) blank rat plasma spiked with CIN and DOM at LLOQ (5 ng/mL) and IS.
Separations 10 00159 g003
Table 1. Results of accuracy studies for CIN and DOM.
Table 1. Results of accuracy studies for CIN and DOM.
LevelsActual Concentration of Drug (ng/mL)Amount of Drug Recovered in Plasma Sample (ng/mL) N = 5)Recovery in Plasma (%)Mean Recovery (%)
CIN
LLOQ5Mean = 4.95; SD = 0.04; CV(%) = 0.8499.00
LQC15Mean = 14.88; SD = 0.14; CV(%) = 0.9499.20
MQC100Mean = 99.28; SD = 0.65; CV(%) = 0.6599.2899.18
HQC150Mean = 148.89; SD = 01.28; CV(%) = 0.8699.26
DOM
LLOQ5Mean = 4.96; SD = 0.04; CV(%) = 0.8199.20
LQC15Mean = 14.86; SD = 0.19; CV(%) = 1.2799.06
MQC100Mean = 99.32; SD = 0.71; CV(%) = 0.7199.3299.28
HQC150Mean = 149.32; SD = 0.96; CV(%) = 0.6499.54
Table 2. (a) Results of precision studies for CIN. (b) Results of precision studies for DOM.
Table 2. (a) Results of precision studies for CIN. (b) Results of precision studies for DOM.
(a)
ParametersLevelConc.Analytical Responses (Area)MeanSDCV
(ng/mL)(N = 6)
RepeatabilityLLOQ511,78511,78111,80511,79811,87811,78011,804.5037.360.32
LQC1512,05512,04312,06512,06412,03312,09612,059.3321.810.18
MQC10013,22913,15613,20513,23213,20513,20113,204.6727.280.21
HQC15013,81913,86213,84713,92113,79313,87013,852.0044.230.32
Intermediate precision (Interday)
DAY 1LLOQ511,78011,78911,78611,80411,87511,78711,803.5035.930.30
LQC1512,06512,11712,04112,11712,09812,03812,079.3336.250.30
MQC10013,23213,22813,20513,19013,19713,16813,203.3324.080.18
HQC15013,82113,79413,80913,87613,86513,81013,829.1733.330.24
DAY 2LLOQ511,81811,79711,88611,81411,76911,78011,810.6641.480.35
LQC1512,01612,03412,11512,05012,17812,11912,085.3362.110.51
MQC10013,13013,19813,24513,10313,34213,25513,212.1787.910.67
HQC15013,79913,62113,83513,94213,85113,79013,806.33105.730.77
DAY 3LLOQ511,75811,79911,78811,77011,86511,87511,809.1649.300.42
LQC1512,08712,10212,08912,15812,09612,13412,11128.650.24
MQC10013,20013,17813,29713,18813,31613,19713,229.3360.560.46
HQC15013,80213,74513,91513,87013,85613,78913,829.5061.950.45
Intermediate precision (Interanalyst)
Analyst 1LLOQ511,77111,81411,79011,85411,84511,78911,810.5033.280.28
LQC1512,01012,17812,05612,13412,08812,00512,078.5068.810.57
MQC10013,15513,20013,11813,10913,36213,15813,183.6793.210.71
HQC15013,78713,72713,73913,85113,89713,87513,812.6771.960.52
Analyst 2LLOQ511,84511,80211,79911,76511,80511,84311,809.8330.160.26
LQC1512,04712,09912,14312,00512,05612,07512,070.8347.200.39
MQC10013,18913,05613,25413,16813,30113,24513,202.1786.020.65
HQC15013,71713,79113,85513,87013,88813,80213,820.5063.440.46
Analyst 3LLOQ511,79211,88211,78711,80311,80511,76611,805.8339.860.34
LQC1511,97512,18611,96612,14512,00512,15812,072.50100.850.84
MQC10013,09813,13913,18913,11913,25413,32013,186.5086.060.65
HQC15013,76913,93613,87613,79113,88113,79713,841.6765.520.47
(b)
ParametersLevelCon.Analytical Responses (area), InjectionsMeanSDCV
(ng/mL)(N = 6)
RepeatabilityLLOQ55212519952345287516052505223.6743.840.84
LQC155778582857895765568657705769.3346.620.81
MQC1008069802681238105805879458054.3363.670.79
HQC1509445955895149615951095979539.8362.930.66
Intermediate precision
DAY 1LLOQ55160523052095188527451905208.5039.670.76
LQC155765571356895834581357895767.1756.730.98
MQC1008112807781348078805680978092.3327.950.35
HQC1509456955596349542957896179563.6763.460.66
DAY 2LLOQ55213518952435217527851585216.3341.650.80
LQC155815586057855743578658145800.5039.220.68
MQC1008133800881328115814880718101.1752.800.65
HQC1509603945697439612957595489589.5093.810.98
DAY 3LLOQ55250518951485245521551985207.5038.060.73
LQC155798571657285808574557895764.0039.190.68
MQC1008078816181348098817382668151.6766.680.82
HQC1509645946695899607946996149565.0077.660.81
Intermediate precision (Interanalyst)
Analyst 1LLOQ55213519852845255529652025241.3342.950.82
LQC155780569857115823578557485757.5047.640.83
MQC1008104815679788132820481648123.0078.480.97
HQC1509587957496079486955496419574.8352.700.55
Analyst 2LLOQ55232527751785207522751665214.5040.270.77
LQC155796584557955808587959145839.5049.000.84
MQC1008015804381167998810380398052.3347.390.59
HQC1509474962495659516959796759575.1773.030.76
Analyst 3LLOQ55287527852665150520852455239.0051.910.99
LQC155769583057905824571658645798.8352.310.90
MQC1008176809881138066813781138117.1737.080.46
HQC1509465961495869523968395709573.5075.000.78
Table 3. (a) Short-term stability for plasma samples of CIN and DOM. (b) Freeze–thaw stability for plasma samples of CIN and DOM. (c) Long-term stability for plasma samples of CIN and DOM.
Table 3. (a) Short-term stability for plasma samples of CIN and DOM. (b) Freeze–thaw stability for plasma samples of CIN and DOM. (c) Long-term stability for plasma samples of CIN and DOM.
(a)
LevelMean AreaS.D.CVAmount of Drug Recovered in Plasma Sample (ng/mL)Recovery (%)
CIN
1 h
1511,924.0046.130.3914.9699.74
10013,141.0069.160.53100.85100.85
15013,831.3338.850.28149.5699.71
2 h
1511,923.6737.820.3214.9499.58
10013,126.6774.040.5699.8499.84
15013,834.0052.030.38149.7599.84
3 h
1511,923.0046.160.3914.8999.27
10013,125.0071.840.5599.7299.72
15013,820.6737.100.27148.8199.21
DOM
1 h
155514.0031.220.5715.01100.08
1008019.0063.690.7999.9099.90
1509494.0038.040.40149.8899.92
2 h
155512.3345.370.8214.9699.70
1008016.6762.000.7799.8299.82
1509478.6757.840.61149.3699.57
3 h
155508.3338.210.6914.8298.80
1008015.0062.550.7899.7699.76
1509458.0044.510.47148.6699.11
(b)
LevelMean AreaS.D.CVAmount of Drug Recovered in Plasma Sample (ng/mL)Recovery (%)
CIN
Cycle 1
1511,923.6780.900.6814.9499.58
10013,128.0042.510.3299.9399.93
15013,835.3357.500.42149.8599.90
Cycle 2
1511,922.6776.070.6414.8799.11
10013,121.6749.170.3799.4899.48
15013,826.6771.600.52149.2499.49
Cycle 3
1511,922.0085.110.7114.8298.80
10013,126.6749.540.3899.8499.84
15013,803.0076.730.56147.5798.38
DOM
Cycle 1
155511.6727.790.5014.9399.55
1008015.3340.500.5199.7799.77
1509489.6759.000.62149.7399.82
Cycle 2
155510.0031.320.5714.8899.18
1008009.6755.050.6999.5899.58
1509473.0067.980.72149.1799.45
Cycle 3
155507.3339.530.7214.7998.57
1007998.3344.000.5599.2099.20
1509466.3342.900.45148.9499.30
(c)
LevelMean AreaS.D.CVAmount of Drug Recovered in Plasma Sample (ng/mL)Recovery (%)
CIN
Week 1
1511,924.0081.610.6814.9699.74
10013,125.6742.500.3299.7699.76
15013,834.6733.650.24149.8099.87
Week 2
1511,922.3387.020.7314.8498.96
10013,113.6759.620.4598.9298.92
15013,821.3370.000.51148.8699.24
Week 3
1511,921.6797.520.8214.8098.64
10013,115.3349.520.3899.0499.04
15013,817.3377.240.56148.5899.05
DOM
Week 1
155511.0044.930.8214.9199.40
1008020.3346.370.5899.9499.94
1509488.3340.070.42149.6999.79
Week 2
155510.3344.000.8014.8999.25
1008007.6753.110.6699.5199.51
1509474.3366.160.70149.2199.48
Week 3
155508.6744.960.8214.8398.87
1008008.6737.500.4799.5599.55
1509451.0054.370.58148.4298.95
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Vij, M.; Dand, N.; Kumar, L.; Ankalgi, A.; Wadhwa, P.; Alshehri, S.; Shakeel, F.; Ghoneim, M.M.; Alam, P.; Wani, S.U.D. RP-HPLC-Based Bioanalytical Approach for Simultaneous Quantitation of Cinnarizine and Domperidone in Rat Plasma. Separations 2023, 10, 159. https://doi.org/10.3390/separations10030159

AMA Style

Vij M, Dand N, Kumar L, Ankalgi A, Wadhwa P, Alshehri S, Shakeel F, Ghoneim MM, Alam P, Wani SUD. RP-HPLC-Based Bioanalytical Approach for Simultaneous Quantitation of Cinnarizine and Domperidone in Rat Plasma. Separations. 2023; 10(3):159. https://doi.org/10.3390/separations10030159

Chicago/Turabian Style

Vij, Mohit, Neha Dand, Lalit Kumar, Amardeep Ankalgi, Pankaj Wadhwa, Sultan Alshehri, Faiyaz Shakeel, Mohammed M. Ghoneim, Prawez Alam, and Shahid Ud Din Wani. 2023. "RP-HPLC-Based Bioanalytical Approach for Simultaneous Quantitation of Cinnarizine and Domperidone in Rat Plasma" Separations 10, no. 3: 159. https://doi.org/10.3390/separations10030159

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop