Next Article in Journal
Phenolic Composition, Antioxidant and Antibacterial Activities of Extract from Flowers of Rosa damascena from Morocco
Previous Article in Journal
Antioxidant Activity and Phenolic Content of Sonication- and Maceration-Assisted Ethanol and Acetone Extracts of Cymbopogon citratus Leaves
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Determination of Three Alkyl Camphorsulfonates as Potential Genotoxic Impurities Using GC-FID and GC-MS by Analytical QbD

1
College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Incheon 21983, Korea
2
Analytical Research Department, Central Research Institute, Hanmi Fine Chemical, 59 Gyeongje-ro, Siheung-si 15093, Korea
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Submission received: 29 July 2022 / Revised: 12 August 2022 / Accepted: 1 September 2022 / Published: 5 September 2022
(This article belongs to the Section Chromatographic Separations)

Abstract

:
Camphorsulfonic acid salts are commonly used in the manufacturing production of active pharmaceutical ingredients (APIs) and have the potential to form alkyl camphorsulfonates, which can be considered as potential genotoxic impurities (PGIs). Alkyl camphorsulfonates should be controlled using the Threshold of Toxicological Concern (TTC) when detected in APIs due to their genotoxicity. An in silico study utilizing the ICH M7 guideline was performed in order to classify the alkyl camphorsulfonates that can be produced from the reaction of camphorsulfonic acid salts with methanol, ethanol, and isopropyl alcohol, which are commonly used solvents in API manufacturing processes. Two sensitive, reproducible, and accurate analytical methods using GC-FID and GC-MS were developed using the analytical Quality By Design (QbD) approaches for the quantitation of three alkyl camphorsulfonates in APIs satisfying the control limit of PGIs according to the TTC. The detection limits of the GC-FID method were found to be between 1.5 to 1.9 ppm, and the detection limits of the GC-MS method were found to be between 0.055 to 0.102 ppm. The method was validated in terms of accuracy, linearity, precision, detection limit, quantitation limit, specificity and robustness.

Graphical Abstract

1. Introduction

The formation of salts with various pharmaceutically available counter ions has been widely used for the advantages of physicochemical properties including solubility, formulation, purification, stability and other various reasons [1,2]. Approximately 50% of APIs include formed salts that can be used pharmacologically, and thus salt development is an integral part of the process development of APIs [3,4,5]. Various types of cation and anion salts have been used, and sulfonic acid salts, such as methanesulfonic acid, camphorsulfonic acid, toluenesulfonic acid, and benzenesulfonic acid, are the most commonly used anion salts [6]. The use of these sulfonic acid salts could potentially lead to the presence of small amounts of sulfonic acid esters as impurities in the final API. Alkyl sulfonates can mainly be generated by the esterification reaction of sulfonic acid with residual alcohols used in the manufacturing process of APIs such as methanol, ethanol and isopropanol. These alkyl sulfonates are known as alkylating agents and can induce genetic mutations or chromosomal aberrations [7,8,9,10,11,12]. Numerous studies have been conducted on the toxicity and analytical methods for these alkyl sulfonates as potential genotoxic impurities (PGIs).
Alkyl sulfonates should be strictly controlled with tight acceptance criteria if produced or introduced in the production of APIs due to their potentially genotoxic behavior. According to the ICH M7 guideline regarding mutagenic impurities, PGIs should be controlled below the Threshold of Toxicological Concern (TTC) in the absence of appropriate limits based on toxicity data. The limits of PGIs in APIs derived from the TTC can be calculated based on the expected maximum daily dose. The TTC value for PGIs in APIs is set at 1.5 µg/day, corresponding to a 10-5 lifetime risk of cancer [13,14].
There have been numerous studies throughout many years to develop analytical methods for the control of PGIs. Most studies have used chromatographic method such as liquid chromatography (LC), gas chromatography (GC) and ion chromatography (IC) with various detectors such as the evaporative light scattering (ELSD), charged aerosol (CAD), mass spectrometry (MS), flame ionization (FID) and UV detectors depending on the target analyte’s chemical properties. [15,16,17,18,19,20]
Since more than 5% of all APIs are prepared in the presence of sulfonic acid, many studies on sulfonic acid alkyl esters have been conducted [21,22,23,24,25]. However, these studies have only been performed for alkyl methanesulfonate, alkyl benzenesulfonate, alkyl toluenesulfonate and alkyl isethionate. Camphorsulfonic acid (CSA) is the second most used sulfonic acid salt after methanesulfonic acid, but not much research has been done on its possibly formed alkyl sulfonate and alkyl camphorsulfonate. Alkyl camphorsulfonates are potential genotoxic impurities and present structurally alerting properties like other sulfonic acid alkyl esters.
Similarly to other alkyl sulfonates, these three alkyl camphorsulfonates chosen can behave as alkylation reagents and have a possibility of alkylating DNA bases (on N-7 of guanosine and N-3 of adenine). These substances have the possibility of causing genetic mutations and present carcinogenic behaviors at even small exposure levels, and therefore must be accurately quantitated and controlled below the TTC.
In order to control these alkyl camphorsulfonates as PGIs, it is essential to develop a highly sensitive, reproducible and stable analysis method. Analytical Quality by Design (QbD) is a systematic approach to developing an optimal analytical method by applying process QbD concepts to method development, promoting method flexibility, and increasing scientific understanding of critical method parameters (CMPs) [26,27,28,29,30,31,32,33,34]. Many studies in the literature have introduced the concept of analytical QbD to develop analytical methods, but there are few studies that use analytical QbD to develop high-sensitivity GC methods to control trace amount of PGIs.
An in silico study using the quantitative structure activity relationship (Q-SAR) approach was conducted for methyl camphorsulfonate (MCS), ethyl camphorsulfonate (ECS) and isopropyl camphorsulfonate (ICS), which can mostly be produced from the reaction of camphorsulfonic acid and alcohols in API manufacturing processes [35,36]. The term “in silico” refers to the computational models that study pharmacological hypotheses using databases. The use of an in silico study in the prediction of toxicity is very useful in order to speed the rate of discovery while reducing cost and time using computer simulations.
An analytical method using GC with the FID was developed for these alkyl camphorsulfonates as PGIs. The analytical QbD concept was adopted for the method development for PGI analysis that requires high sensitivity and reproducibility to meet regulatory requirements. Since the most important point in developing the PGIs analysis method is to have high sensitivity to detect trace amounts of PGIs, the height of the each PGI peak was selected as the critical method attributes (CMAs). Based on the optimized GC-FID method by analytical QbD, an additional method using the MS was developed to obtain better detection sensitivity. Both methods using GC-FID and GC-MS were validated through analytical method validation according to the ICH Q2 (R2) guideline regarding the validation of analytical procedures in terms of linearity, accuracy, robustness, specificity, precision and system suitability [37]. The potential synthetic route and structures of MCS, ECS and ICS are shown in Figure 1.

2. Materials and Methods

2.1. Reagents, Materials and Standards

The reference standards for (1S)-(+)-10-camphorsulfonic acid methyl ester (MCS, Cas No. 62319-13-5, purity: 99.9%), (1S)-(+)-10-camphorsulfonic acid ethyl ester (ECS, Cas No. 154335-57-6, purity: 94.5%) and (1S)-(+)-10-camphorsulfonic acid isopropyl ester (ICS, Cas No. 247078-58-6, purity: 98.1%) were purchased from Toronto Research Chemicals (Toronto, ON, Canada). All of the reference standards were characterized by NMR, HPLC and MS. Phenyl ether was purchased from Acros Organics (Geel, Belgium). Methanol, ethanol, isopropanol and ethyl acetate were purchased from Merck (Darmstadt, Germany). Acetonitrile was purchased from Fisher Scientific (Pittsburg, PA, USA). (1S)-(+)-10-camphorsulfonic acid was purchased from TCI (CSA, Tokyo, Japan). The Amlodipine camsylate, API (CAS No.: 652969-01-2) bulk drug was obtained from Hanmifine Chemical (Shihung, South Korea).

2.2. Analytical Condition and Equipments

2.2.1. GC-FID Condition

The analyses and validation of MCS, ECS and ICS were conducted using an Agilent 7890 gas chromatograph equipped with an FID (Agilent Technologies, Santa Clara, CA, USA). Sample injection was performed using an Agilent 7693 auto-sampler (Agilent Technologies, Santa Clara, CA, USA). Mass Hunter Data Analysis Version. B.07.04.2260 was used as the data acquisition software from Agilent Technologies.
The analyses of MCS, ECS, ICS with the FID were performed using an HP-1 capillary column (25 m × 0.32 mm i.d. × 0.52 μm film thickness) from Agilent Technologies. The column oven temperature was set at the following temperature program: initial oven temperature set at 200 °C, held for 8 min, then ramped at 70 °C/min to 260 °C, held for 8 min. The flow rate of the helium carrier gas was set at 2.0 mL/min with the constant flow mode and total flow was set at 25 mL/min. The Split Inlet was used at a split ratio set at 10:1 and split flow was set at 20 mL/min. For injection, a 4 mm straight liner Inlet was used. The temperature of injection heater was set at 280 °C, septum purge flow was set at 3.0 mL/min, the pressure was 13.7 psi and the average velocity was 43.5 cm/s. The detector heater temperature was set at 300 °C, air flow was set at 400 mL/min, H2 fuel flow was set at 30 mL/min and the make-up flow was set at 25 mL/min. Helium was used as the make-up gas. Injection volume was set at 2.0 μL using a 5 μL auto liquid sampler syringe. The syringe needle was rinsed with ethyl acetate between injections. Total run time was 16.9 min with no solvent delay time. Data acquisition rate was 50 Hz and the minimum peak width was set at 0.004 min.

2.2.2. GC-MS Condition

The analyses and validation of MCS, ECS, and ICS were conducted using an Agilent 7890 gas chromatograph equipped with a 5977A Single Quadrupole mass detector (Agilent Technologies, Santa Clara, CA, USA). Sample injection was performed using an Agilent 7693 auto-sampler (Agilent Technologies, Santa Clara, CA, USA). Mass Hunter qualitative analysis version B.07.00 from Agilent Technologies was used for the control of equipment, processing and data acquisition software.
The GC conditions are the same as the GC-FID conditions. The MS was operated using the high efficiency EI as the type of source in Selective Ion Monitoring mode (SIM). The ions with m/z = 81.0, 109.1, 151,1, 170.1 with 50 ms dwell time were selected for analysis. The temperature of source was set at 230 °C, the temperature of the quadrupole was set at 150 °C and the temperature of transfer line was set at 280 °C. The gain factor was set at 1.0. Injection volume was set at 2.0 μL using a 5 μL auto liquid sampler syringe. The syringe needle was rinsed with ethyl acetate between injections. Total run time was 16.9 min with a solvent delay time of 0.8 min.

2.3. Preparation of Standard Solution and Sample Solution

To enhance the precision of measurements, phenyl ether was used as internal standard.

2.3.1. Internal Standard Solution Preparation

A quantity of 0.1 g of phenyl ether was weighed into a 100 mL volumetric flask, then sufficiently mixed, dissolved and diluted to the volume mark with ethyl acetate. Then, 5.0 mL of this solution was added to a 100 mL volumetric flask, then sufficiently mixed and diluted to the volume mark with ethyl acetate.

2.3.2. Standard Stock Solution Preparation (STD-2)

Totals of 0.1 g of the MCS reference standard, 0.1 g of the ECS reference standard and 0.1 g of the ICS reference standard were weighed to a 100 mL volumetric flask, then sufficiently mixed, dissolved and diluted to the volume mark with ethyl acetate (STD-1). 6.0 mL of STD-1 solution was added to a 100 mL volumetric flask, then sufficiently mixed and diluted to the volume mark with ethyl acetate (STD-2).

2.3.3. Standard Solution Preparation

Quantities of 5.0 mL of the STD-2 solution and 5.0 mL of the internal standard solution were added to a 50 mL volumetric flask, then sufficiently mixed and diluted to the volume mark with ethyl acetate. This solution was sonicated for 1 min and filtered through a 0.45 µm membrane filter. The concentration of the standard solution is 150 ppm for each of the target analytes of MCS, ECS, and ICS.

2.3.4. Sample Solution Separation

Approximately 2.0 g of the test specimen and 5.0 mL of the internal standard solution were weighed and added to a 50 mL volumetric flask and then sufficiently mixed, dissolved and diluted to the volume mark with ethyl acetate. This solution was sonicated for 1 min and filtered through a 0.45 µm membrane filter (concentration: 4 mg/mL).

2.4. In Silico Study

According to the ICH guidelines, the toxicity assessments of bacterial mutation assay can be computationally assessed using two correlative rules-based and statistics-based Q-SAR methodologies. The rules-based software used was Derek Nexus (version 6.2.0, built on 11 March 2022) from Lhasa Limited (Leeds, UK), and the statistics-based software used was Sarah Nexus (v.3.2.0 built on 11 March 2022) purchased from Lhasa Limited [38]. Both softwares have been validated according to the guidelines set by the Organization for Economic Cooperation and Development (OECD). For additional analysis of the results, the free software VEGA (v.1.1.5, Vegahub, Milan, Italy), country was used [39]. The in silico study was conducted for the chemical structures of MCS, ECS, ICS as well as CSA.

2.5. Method Screening

Fusion QbD Professional 9.8.1.135 (S-matrix, Eureka, CA, USA) was used to create the Design of Experiment (DoE) for the method screening and optimization study [40]. The major parameters which can affect the analysis results were screened. The screening of various types of columns was performed using the one-factor-at-a-time (OFAT) method. Then the column oven temperature, inlet temperature, flow rate of carrier gas, detector temperature and injection volume were screened to understand the interactions of each analytical parameters using the DoE and analytical QbD concepts. The DoE was conducted in two-level-resolution V type. Three center points were included in the model.

2.6. Method Optimization

The developed analytical methods were optimized using the DoE for the selected CMPs using the results of the screening study. The column oven temperature, flow rate of carrier gas and injector temperature were determined as the CMPs. The DoE was created using a response surface-central composite including three center points. These results were used to determine the method operable design region (MODR) and the proven acceptable range (PAR), which allows for flexibility in the analytical method.

2.7. Method Validation

The ICH Q2 guideline was used for the validation of the developed methods. The method validation consisted of the parameters of specificity, detection limit, quantitation limit, accuracy, precision, linearity, and robustness.

2.7.1. Determination of Specificity

Specificity was evaluated by determining the resolution between the MCS, ECS and ICS peaks from the chromatograms. The standard solution prepared was sampled and analyzed in 6 replicates. Using the chromatogram, the resolutions between MCS, ECS and ICS peaks were calculated automatically using the computerized data system for peak integration.

2.7.2. Determination of the Detection Limit and Quantitation Limit

The detection limit (DL) and quantitation limit (QL) were determined by calculating the signal-to-noise ratio obtained from the comparison of each signal (height) of analytes and noise in the given time range closest to the analyte. The signal-to-noise ratios at approximately 3:1 and 10:1 were considered acceptable for estimating the DL and QL, respectively.

2.7.3. Determination of Accuracy

Accuracy was evaluated using the recovery study of spiked amounts of MCS, ECS and ICS. Each of the analyzed accuracy solutions were prepared in triplicates at three levels of the specifications with known amounts of MCS, ECS, and ICS spiked to a single batch of the matrix sample. The recoveries of the accuracy solutions of MCS, ECS, and ICS were then calculated.

2.7.4. Determination of Precision

Precision was evaluated to prove the analytical method’s ability to obtain close data results between a series of measurements from several injections of the standard solution over a short time interval. Precision was evaluated based on the repeatability precision of the results.

2.7.5. Determination of Linearity

Linearity was evaluated by assessing the linear proportionality of the concentration of the analytes to the signals obtained over the specific range from the reporting level of genotoxic impurities (the QL concentration) to 120% of the specifications. The results were evaluated using the slope, y-intercept, and correlation coefficient of the linear regression obtained from the linearity solutions at five concentration levels within the specified range.

2.7.6. Determination of Robustness

The robustness of the proposed analytical methods was evaluated by determining the influences on the analysis results when varying method parameters such as the column oven temperature and the flow rate of the carrier gas.

3. Results

3.1. In Silico Study

Alkyl sulfonates such as alkyl methanesulfonates, alkyl toluenesulfonates and alkyl benzenesulfonates are chemicals widely known to be genotoxic. However, since the genotoxicity of alkyl camphorsulfonates are not well known, an in silico study was performed according to the ICH M7 guideline for their classification. Derek Nexus, an expert knowledge-based software, and Sarah Nexus, a statistical-based software, were used along with the VEGA Q-SAR model to correspondingly evaluate the genotoxicity of the target analytes.
CSA was found to be negative in both Derek and Sarah prediction results. MCS, ECS and ICS were all found to be positive and have structural alerts and were classified as class 3. The VEGA Q-SAR model also showed similar results. The genotoxicity predictions for the alkyl camphorsulfonates are shown in Table 1. The detailed prediction results are summarized in Table S1 of the Supporting Document.

3.2. Method Screening

The type of column used is a very important factor due to the various effects the column has on the CMAs such as resolution, peak shape, reproducibility and sensitivity. Capillary columns were chosen due to their separation ability and convenience of use, and various types of capillary columns were screened using the one-factor-at-a-time (OFAT) method. The WAX column (100% polyethylene glycol), FFAP column (acid modified polyethylene glycol), CAM (base deactivated polyethylene glycol) were excluded from the column screening process due to their low analysis temperature range. DB-624 (6% cyanopropylphenyl/94% dimethylpolysiloxane) and HP-1 (100% dimethylpolysiloxane) were both tested.
Ethyl acetate was used as the diluent in order obtain sufficient resolution between the solvent peak and the target analytes. All of the target analytes have high boiling points and, therefore, a relatively faster eluting solvent was chosen rather than a slowly eluting solvent. The reasons for choosing ethyl acetate were due to the fact that all three analytes chosen for this study presented sufficient solubility in this solvent, the solvent being a GC-MS compatible, and its faster elution time before the target analytes.
The peak height of each analyte and the resolution between adjacent peaks were selected as the critical method attributes (CMAs) since the most critical analytical target profiles (ATPs) in the analysis of PGIs is detection sensitivity and specificity due to their low specification limits. A risk assessment was performed in order to determine the critical method parameters (CMPs) expected to have the greatest impacts on the CMAs. Through the risk assessment, the flow rate of carrier gas, inlet temperature, detector temperature, column oven temperature and sample injection volume were chosen as the CMPs.
The Fusion QbD software was used to create an experimental plan using a two-level-resolution V type including three center points. A total of 19 experiments were performed. The Design of Experiments (DoE) is tabulated in Table 2 and the experimental results of each CMA are tabulated in Table 3. The Analysis of Variance (ANOVA) statistical test was performed in order to determine the statistical significance of each response, shown in Table 4. The p-values of the responses for each of the PGIs were found to be below 0.05 and, thus, were proven to have statistical significance.
The model term ranking Pareto charts represent the effects and interactions of each of the parameters based on the CMAs established by the model term effect and the cumulative percentage. The model term ranking Pareto charts are shown in Figure 2. The Pareto charts show that the column oven temperature and/or the flow rate had the greatest effect on the CMAs, such as the peak height of each of the target analytes, the resolution between each of the target analytes and the retention time of ICS. The selected responses were shown to not be affected or partially affected by the detector temperature and injection volume. As a result, the column oven temperature, flow rate of the carrier gas and inlet temperature were selected as the CMPs for the method optimization study.

3.3. Method Optimization by Analytical QbD

The analytical method evaluated through the screening process was optimized using the analytical QbD and DoE approaches. Optimization designs are typically used to define the experiment variable level settings used to predict the best overall performance, and the experiment variable ranges used to predict the acceptable overall mean performance and acceptable robustness. Similarly to the screening process, the CMAs were chosen to be the peak height of each of the target analytes, resolution between each of the target analytes, and the retention time of ICS. The CMPs were chosen based on the screening results to be the column oven temperature, flow rate of the carrier gas and inlet temperature, which were shown to have the greatest effects on the CMAs.
The experiments were designed using a response surface-central composite full type including three center points by a DoE using the Fusion QbD software. The Design of Experiments (DoE) is tabulated in Table 5 and the experimental results of each CMA are tabulated in Table 6. The models created were statistically validated using general regression statistics and the ANOVA statistical test. As shown in Table 7, the p-values for each of the responses are less than 0.0001, which is substantially smaller than the acceptance criteria of 0.05.
The response surface methodology (RSM) analysis was reviewed using 3D response surface plots to identify underlying interactions between selected parameters (Figure 3). The inlet temperature, selected as one of the CMPs, was shown to have no significant effect on the response and tendency of the CMA in all of the results. The 3D response surface plots at 260 °C and 300 °C are summarized in Figures S1 and S2 of the Supporting Document, respectively.
The flow rate was shown to have little effect on (a) the peak height of MCS and (b) the peak height of ECS, and the respective peak heights showed a decreasing linear trend with a decrease in column oven temperature. The response surface plots of the peak height of ICS (c) showed a curved relationship between the flow rate and column oven temperature. Both CMPs demonstrated a more varied interaction with the change in peak height. The DoE was shown to have a maximum point when the column oven temperature and flow rate is low. The 3D response surface plot for the peak height of ICS (c) was shown to have a saddle shape with the minimum point of the peak height being when the column oven temperature was low and the flow rate was high, or when the column oven temperature was high and the flow rate was low. The resolution between MCS and ECS (d) was shown to have little effect on the flow rate and tended to increase linearly with a decrease in column oven temperature. The resolution of ECS and ICS (e) showed an increase as the oven temperature decreased and had a maximum point when the flow rate was around 2.0 mL/min and decreased at both ends. The retention time of ICS (f) showed a tendency to decrease with an increase in column oven temperature rather than the effect of flow rate. Overall, the flow rate was shown to affect the established CMA only to a certain extent, but the column oven temperature was shown to have a significant effect. The method operable design region (MODR) and proven acceptable ranges (PARs) are shown in Figure 4.

3.4. Design Space

The optimal region was set by superimposing all responses of interest. The acceptance criteria for each of the responses were set to satisfy the purpose of the ATP. The acceptance criteria of all peak heights were set to 2 or greater, in order to ensure sufficient sensitivity in order to detect trace amounts of each of the PGIs. To ensure that each peak is sufficiently separated, and inter-peak interference is minimized, the acceptance criteria for the degree of separation were set at 7 or greater for MCS and ECS and 3 or greater for ECS and ICS. The retention time of the last eluted peak, ICS, was set to 7 min or less in order to shorten the analysis time. The shaded contour areas in Figure 4 indicate the unacceptable area and the unshaded areas indicate acceptable areas. The unshaded area depicts the parameter values that can be represented as a design space with analytical method flexibility. The proven acceptable range (PAR) was defined up to the set center point of the parameter range within a predefined MODR based on a linear combination of parameters. The resolution between ECS and ICS and the peak height of ICS had the most influence on establishing the MODR and PAR.

3.5. Apply MS

Based on developed GC-FID analytical method using analytical QbD, a GC-MS analytical method was developed and compared by introducing the mass-selective detector (MS) instead of the flame ionization detector (FID) under the same analytical conditions. The specifications of the PGIs of interest were set using the concept of TTC and sufficient sensitivity was obtained depending on the target API’s maximum daily dose. Generally, the MS operated under SIM mode presents a significantly higher sensitivity as opposed to the FID, and therefore, the MS was chosen as the detector to be used alongside the GC.
A total of four ions were selected through a standard solution analysis using GC-MS operated under scan mode for analyzing all esters and internal standards simultaneously. The selected ions had m/z values of 81.0, 109.1, 151,1, and 170.1. The fragments at m/z values of 81.0, 109.1 and 151.1 were identified as MCS, ECS and ICS. These m/z values were considered the most suitable for the detection of MCS, ECS and ICS. The fragment at the m/z value of 170.1 was identified as the internal standard (diphenyl ether) and the fragment at the m/z value of 170.1 was not found in any of the signals of MCS, ECS and ICS. The same split ratio was used for both the MS and FID methods in order to set the method parameters similarly for both methods. Sufficient sensitivity was obtained for the MS method with a larger split ratio. However, unlike typical impurities, genotoxic impurities require the most optimal sensitivity due to their low acceptance criteria and therefore, the split ratio was not altered. There were also no carry-over problems observed even when using a small split ratio with the MS. As shown in Figure 5 and Figure 6, the results of the GC-FID analytical method (Figure 5) were almost the same as the results obtained from the GC-MS analytical method (Figure 6). As a result of the change in detector from the FID to MS, the detection limits (DL) for each of the target analytes were found to be between 1.5 to 1.9 ppm initially and improved to be between 0.055 to 0.102 ppm. The quantitation limits (QL) for each of the target analytes were found to be between 4.9 to 6.4 ppm initially and improved to be between 0.185 to 0.340 ppm. Thus, the DL and QL showed approximately 18 to 27 times improved limits with a change in the detector.
Both the GC-FID and GC-MS analytical methods demonstrate sufficient detection limits capable of controlling the specifications for the PGIs of amlodipine (daily dose = 10 mg) set at less than 150 ppm, calculated using the TTC and ICH M7 guideline. For APIs, including amlodipine, with PGI specifications set at greater than 10 ppm or with a maximum daily dose set at 150 mg or lower, using the FID instead of the MS is a much more cost-effective analytical method with sufficient sensitivity.

3.6. Applicability of the Method to Real Sample

In order to verify that the developed analytical method for the target PGIs is applicable to APIs, the API amlodipine camsylate (Cas No. 652969-01-2) that uses CSA as a salt, was selected as a matrix API. Amlodipine camsylate was chosen as the API of interest because alcohols including methanol and ethanol are used in the manufacturing process and thus, has potential to have residual alkyl camphorsulfonates in the final product. The matrix API amlodipine camsylate was provided by Hanmi Fine Chemical located in South Korea.
Amlodipine camsylate was also chosen as the matrix API as there was no residual alkyl camphorsulfonates detected in the sample when analyzed using the two analytical methods using GC-FID and GC-MS developed by QbD, and thus, the experiment was carried out by spiking the matrix sample with the specified limits of each of the three alkyl camphorsulfonates calculated based on the TTC. Each of the experiments were carried out three times, and the recovery and repeatability (precision) were determined for each of the results. Recovery and repeatability are two representative parameters in the ICH Q2 guideline regarding the validation of analytical procedures that identifies whether the sample API has an effect on the analytical method. Recovery was found to be between 99.9 to 103.2% when using the FID, and between 89.4 to 99.8% when using the MS. Precision (%RSD) was found to be between 0.64 to 2.11% when using the FID, and 0.83 to 2.47% when using the MS. No issues or interferences were shown when applying the analytical method developed to the API amlodipine camsylate, with accuracy and precision being confirmed for all of the alkyl camphorsulfonates. Therefore, applicability of the method to real samples was confirmed for the GC-FID and GC-MS analytical methods. All results are presented in Table 8.

3.7. Analytical Method Validation and Robustness Test

The analytical methods developed using GC-FID and GC-MS were validated for the purpose of analyzing the contents of PGIs according to the ICH guideline Q2 Validation of analytical procedures. The analytical methods were evaluated based on the parameters of the detection limit, quantitation limit, linearity, precision, accuracy and specificity. A robustness test was conducted to determine the ability of the analytical method to remain unaffected by small, deliberate variations in method parameters. The matrix sample amlodipine camsylate was used for the validation study. The analytical method validation and robustness test results are summarized in Table 9 and Table 10.

3.7.1. Limits of Detection and Quantification

The detection limit refers to the minimum amount of the target analyte that the analytical method is able to detect. The quantitation limit refers to the minimum amount of the target analyte that the analytical method is able to precisely and accurately quantitate. The quantitation limit is a validation parameter for the quantitative testing of samples containing trace amounts of analyte, or in particular for the determination of impurities and decomposition products. For the determination of the detection limit and quantitation limit, the signal-to-noise ratio of a reference solution containing a low concentration of 30 ppm of each of the target analytes was analyzed. The signal-to-noise ratio of 3:1 was considered for the determination of the detection limit and 10:1 was considered for the determination of the quantitation limit. When obtained using GC-FID, the detection limits were found to be 1.5 ppm, 1.5 ppm and 1.9 ppm, and the quantification limits were found to be 4.9 ppm, 5.1 ppm, 6.4 ppm for MCS, ECS and ICS, respectively. When obtained using GC-MS, the detection limits were found to be 0.055 ppm, 0.069 ppm, 0.102 ppm, and the quantification limits were found to be 0.185 ppm, 0.232 ppm, 0.340 ppm for MCS, ECS and ICS, respectively. As expected, the analytical method for the MS operated under SIM mode had much better sensitivity than the analytical method for the FID. The baseline noise was determined to be significantly better when operated using SIM mode. In addition, injection precision was confirmed through six repeated injections at a low concentration near the quantification limit. All results satisfied the acceptance criteria with the relative standard deviation (%RSD) being below 10.0%. Both of the analytical methods were confirmed to have suitable detection sensitivity for each purpose.

3.7.2. Linearity and Range

Linearity refers to the ability of the analytical method to detect a linear response of the reference standard with respect to its relative concentration within a specific range. Linearity was evaluated at five concentration levels at the QL concentration to 180 ppm (120% of the specified limit). The range was chosen to be from the range of 30 ppm to 150 ppm for MCS, ECS, and ICS.
Linearity was evaluated as coefficient of determination (R2) and all of the results using both FID and MS were not less than 0.99. The overall linearity was better established in the method using FID than MS.

3.7.3. Precision and Accuracy

Precision refers to the degree of dispersion between a series of measurements of a uniform sample analyzed using the same analytical method. Accuracy refers to the ability of the analytical method to obtain results close to a standard or known true value. Precision was evaluated by determining the %RSD of the peak area ratios between MCS, ECS, ICS and the internal standard obtained from six replicate injection of the standard solution. Precision was evaluated at the concentration levels of 10 ppm, 30 ppm and 150 ppm. All results satisfied the acceptance criteria, with %RSD values below 10.0 %.
Accuracy was evaluated by determining the recovery of the sample solutions spiked with known concentrations of MCS, ECS, and ICS. The spiked sample solutions were prepared in triplicate at three levels using the drug substance, amlodipine camsylate as the matrix sample. The amount of each target analyte was quantitated using the developed analytical method and the recoveries for each of the solutions were calculated.
Accuracy was evaluated at 10 ppm, 120 ppm, 150 ppm and 180 ppm. All results satisfied the acceptance criteria, with the recovery being within 100 ± 15% for all target analytes at each concentration level.

3.7.4. Specificity

Specificity refers to the ability of the method to quantitate the amount of each target analyte without any interferences when deliberately spiked with impurities or degradants of the sample. The resolution between each of the target analytes for the standard solution were determined to evaluate the specificity of the analytical method. The resolution between MCS and ECS was determined to be 9.51 and 4.53 between ECS and ICS obtained using the GC-FID analytical method. The resolution between MCS and ECS was determined to be 10.21 and 4.78 between ECS and ICS obtained using the GC-MS analytical method. All resolution values between adjacent peaks were not less than 3.0, meaning that all peaks were well separated from one another.

3.7.5. Robustness

Robustness of the analytical method refers to the ability of the analytical method to remain unchanged when the conditions are intentionally changed. Robustness is a validation parameter indicating how reliable the test method is during normal use. From the established analytical method, the flow rate of the carrier gas was changed from 1.8 mL/min to 2.2 mL/min, and the column oven temperature was changed from 195 °C to 205 °C. All other analytical parameters except for the flow rate and column oven temperature were fixed at their original set values in order to verify the effect of flow rate and column oven temperature on the analysis results. To determine the effects of the changes in the results, precision of altered analytical method was evaluated using the %RSD and standard deviation of the peak area ratios between MCS, ECS, ICS and the internal standard obtained from six replicate injections of the standard solution. The resolution between adjacent peaks and tailing factor were also evaluated for each peak. For the analytical method using GC-FID, the obtained precision values satisfied the acceptance criteria with %RSD being below 10.0%. The resolution for robustness conditions 1 to 5 were found to be between 7.90 and 10.03 between the peaks of MCS and ECS, and between 3.80 and 4.68 between the peaks of ECS and ICS. All results were not less than 3.0. The tailing factor was found to be between 0.85 and 1.04. For the analytical method using GC-MS, the obtained precision values also satisfied the acceptance criteria with %RSD being below 10.0%. The resolution for robustness conditions 1 to 5 were found to be between 8.85 and 10.56 between the peaks of MCS and ECS, and between 4.15 and 4.93 between the peaks of ECS and ICS. All results were not less than 3.0. The tailing factor was found to be between 0.90 and 1.02. The analytical results for both the GC-FID and GC-MS were determined to be robust even with changes in flow rate and column oven temperature.

4. Conclusions

The three alkyl camphorsulfonates were identified as PGIs through an in silico study according to the ICH M7 guideline. An analytical method for three alkyl camphorsulfonates using GC was developed by analytical QbD demonstrating high sensitivity, reproducibility and a short time of analysis. The concept of analytical QbD was effective to develop the analytical method within a design space to satisfy the needs of high sensitivity and specificity as CMAs when analyzing PGIs. The developed PGI analysis methods have been validated according to the ICH Q2 guideline. The GC-FID method showed better results in linearity and recovery, and the GC-MS method showed better results in detection sensitivity. Both the GC-FID and GC-MS methods can be used to effectively carry out routine analysis in the quality control of PGIs in the pharmaceutical industry, and the developed analytical methods can be applied to APIs containing camphorsulfonic acid salt to control PGIs. The limitations to this study may be the fact that only helium was used as the carrier gas. There may be additional testing required in order to check the effects of carrier gases other than helium on the analytical method’s capabilities. Moreover, additional studies for the development of an analytical method for other alkyl camphorsulfonate PGIs other than methyl, ethyl, and isopropyl camphorsulfonate is needed.

Supplementary Materials

The following supporting information can be downloaded at: https://0-www-mdpi-com.brum.beds.ac.uk/article/10.3390/separations9090246/s1, Table S1: In silico prediction results; Figure S1: 3D response surface plots for each response as a function of flow rate and column oven temperature at 260 °C of inlet temperature; Figure S2: 3D response surface plots for each response as a function of flow rate and column oven temperature at 300 °C of inlet temperature.

Author Contributions

Conceptualization, K.L.; methodology, K.L. and W.Y.; software, K.L.; validation, K.L. and W.Y.; formal analysis, K.L. and W.Y.; writing—original draft preparation, K.L.; writing—review and editing, W.Y.; visualization, K.L.; supervision, J.H.J.; project administration, J.H.J. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Serajuddin, A.T.M. Salt Formation to Improve Drug Solubility. Adv. Drug Deliv. Rev. 2007, 59, 603–616. [Google Scholar] [CrossRef] [PubMed]
  2. Elder, D.P.; Holm, R.; de Diego, H.L. Use of Pharmaceutical Salts and Co crystals to Address the Issue of Poor Solubility. Int. J. Pharm. 2013, 453, 88–100. [Google Scholar] [CrossRef] [PubMed]
  3. Berge, S.M.; Bighley, L.D.; Monkhouse, D.C. Pharmaceutical Salts. J. Pharm. Sci. 1977, 66, 417–435. [Google Scholar] [CrossRef]
  4. Elder, D.P.; Delaney, E.; Teasdale, A.; Eyley, S.; Reif, V.D.; Jacq, K.; Facchine, K.L.; Oestrich, R.S.; Sandra, P.; David, F. The Utility of Sulfonate Salts in Drug Development. J. Pharm. Sci. 2010, 99, 2948–2961. [Google Scholar] [CrossRef] [PubMed]
  5. Gupta, D.; Bhatia, D.; Dave, V.; Sutariya, V.; Gupta, S.V. Salts of Therapeutic Agents: Chemical, Physicochemical, and Biological Considerations. Molecules 2018, 23, 1719. [Google Scholar] [CrossRef] [PubMed]
  6. Teasdale, A.; Delaney, E.J.; Eyley, S.C.; Jacq, K.; Taylor-Worth, K.; Lipczynski, A.; Hoffmann, W.; Reif, V.; Elder, D.P.; Facchine, K.L.; et al. A Detailed Study of Sulfonate Ester Formation and Solvolysis Reaction Rates and Application toward Establishing Sulfonate Ester Control in Pharmaceutical Manufacturing Processes. Org. Process Res. Dev. 2010, 14, 999–1007. [Google Scholar] [CrossRef]
  7. Snodin, D.; Teasdale, A. Mutagenic Alkyl-Sulfonate Impurities in Sulfonic Acid Salts: Reviewing the Evidence and Challenging Regulatory Perceptions. Org. Process Res. Dev. 2015, 19, 1465–1485. [Google Scholar] [CrossRef]
  8. Glowienke, S.; Frieauff, W.; Allmendinger, T.; Martus, H.J.; Suter, W.; Mueller, L. Structure-Activity Considerations and in Vitro Approaches to Assess the Genotoxicity of 19 Methane-, Benzene- and Toluenesulfonic Acid Esters. Mutat. Res.-Genet. Toxicol. Environ. Mutagenesis 2005, 581, 23–34. [Google Scholar] [CrossRef]
  9. Sobol, Z.; Engel, M.E.; Rubitski, E.; Ku, W.W.; Aubrecht, J.; Schiestl, R.H. Genotoxicity Profiles of Common Alkyl Halides and Esters with Alkylating Activity. Mutat. Res.-Genet. Toxicol. Environ. Mutagenesis 2007, 633, 80–94. [Google Scholar] [CrossRef]
  10. Couch, D.B.; Forbes, N.L.; Hsie, A.W. Comparative Mutagenicity of Alkylsulfate and Alkanesulfonate Derivatives in Chinese Hamster Ovary Cells. Mutat. Res.-Fundam. Mol. Mech. Mutagenesis 1978, 57, 217–224. [Google Scholar] [CrossRef]
  11. Ashby, J.; Basketter, D.A.; Paton, D.; Kimber, I. Structure Activity Relationships in Skin Sensitization Using the Murine Local Lymph Node Assay. Toxicology 1995, 103, 177–194. [Google Scholar] [CrossRef]
  12. Roberts, D.W.; Basketter, D.A. Quantitative Structure-Activity Relationships: Sulfonate Esters in the Local Lymph Node Assay. Contact Dermat. 2000, 42, 154–161. [Google Scholar] [CrossRef] [PubMed]
  13. Kroes, R.; Kozianowski, G. Threshold of Toxicological Concern (TTC) in Food Safety Assessment. Toxicol. Lett. 2002, 127, 43–46. [Google Scholar] [CrossRef]
  14. International Council for Harmonisation Guideline M7 (R1) on Assessment and Control of DNA Reactive (Mutagenic) Impurities in Pharmaceuticals to Limit Potential Carcinogenic Risk Step 4 Version 2017. Available online: https://database.ich.org/sites/default/files/M7_R1_Guideline.pdf (accessed on 3 May 2022).
  15. Lee, C.R.; Hubert, M.; Nguyen Van Dau, C.; Peter, D.; Krstulovic, A.M. Determination of N,N-Dimethylaminoethyl Chloride and the Dimethylaziridinium Ion at Sub-Ppm Levels in Diltiazem Hydrochloride by LC-MS with Electrospray Ionisation. Analyst 2000, 125, 1255–1259. [Google Scholar] [CrossRef]
  16. Li, S.; Dong, L.; Tang, K.; Lan, Z.; Liu, R.; Wang, Y.; Wang, R.; Lin, H. Simultaneous and Trace Level Quantification of Two Potential Genotoxic Impurities in Valsartan Drug Substance Using UPLC-MS/MS. J. Pharm. Biomed. Anal. 2022, 212, 114630. [Google Scholar] [CrossRef]
  17. Liu, D.Q.; Sun, M.; Kord, A.S. Recent Advances in Trace Analysis of Pharmaceutical Genotoxic Impurities. J. Pharm. Biomed. Anal. 2010, 51, 999–1014. [Google Scholar] [CrossRef]
  18. al Azzam, K.M.; Aboul-Enein, H.Y. Recent Advances in Analysis of Hazardous Genotoxic Impurities in Pharmaceuticals by HPLC, GC, and CE. J. Liq. Chromatogr. Relat. Technol. 2016, 39, 1–7. [Google Scholar] [CrossRef]
  19. Reddy, A.V.B.; Jaafar, J.; Umar, K.; Majid, Z.A.; bin Aris, A.; Talib, J.; Madhavi, G. Identification, Control Strategies, and Analytical Approaches for the Determination of Potential Genotoxic Impurities in Pharmaceuticals: A Comprehensive Review. J. Sep. Sci. 2015, 38, 764–779. [Google Scholar] [CrossRef]
  20. Elder, D.P.; Snodin, D.J. Drug Substances Presented as Sulfonic Acid Salts: Overview of Utility, Safety and Regulation. J. Pharm. Pharmacol. 2009, 61, 269–278. [Google Scholar] [CrossRef]
  21. Li, W. Trace Analysis of Residual Methyl Methanesulfonate, Ethyl Methanesulfonate and Isopropyl Methanesulfonate in Pharmaceuticals by Capillary Gas Chromatography with Flame Ionization Detection. J. Chromatogr. A 2004, 1046, 297–301. [Google Scholar] [CrossRef]
  22. Raman, N.V.V.S.S.; Reddy, K.R.; Prasad, A.V.S.S.; Ramakrishna, K. Development and Validation of a GC-MS Method for the Determination of Methyl and Ethyl Camphorsulfonates in Esomeprazole Magnesium. Chromatographia 2008, 68, 675–678. [Google Scholar] [CrossRef]
  23. Jacq, K.; Delaney, E.; Teasdale, A.; Eyley, S.; Taylor-Worth, K.; Lipczynski, A.; Reif, V.D.; Elder, D.P.; Facchine, K.L.; Golec, S.; et al. Development and Validation of an Automated Static Headspace Gas Chromatography-Mass Spectrometry (SHS-GC-MS) Method for Monitoring the Formation of Ethyl Methane Sulfonate from Ethanol and Methane Sulfonic Acid. J. Pharm. Biomed. Anal. 2008, 48, 1339–1344. [Google Scholar] [CrossRef] [PubMed]
  24. Liu, Z.; Fan, H.; Zhou, Y.; Qian, X.; Tu, J.; Chen, B.; Duan, G. Development and Validation of a Sensitive Method for Alkyl Sulfonate Genotoxic Impurities Determination in Drug Substances Using Gas Chromatography Coupled to Triple Quadrupole Mass Spectrometry. J. Pharm. Biomed. Anal. 2019, 168, 23–29. [Google Scholar] [CrossRef] [PubMed]
  25. Szekely, G.; de Sousa, M.C.A.; Gil, M.; Ferreira, F.C.; Heggie, W. Genotoxic Impurities in Pharmaceutical Manufacturing: Sources, Regulations, and Mitigation. Chem. Rev. 2015, 115, 8182–8229. [Google Scholar] [CrossRef]
  26. Dinh, N.P.; Shamshir, A.; Hulaj, G.; Jonsson, T. Validated Modernized Assay for Foscarnet in Pharmaceutical Formulations Using Suppressed Ion Chromatography Developed through a Quality by Design Approach. Separations 2021, 8, 209. [Google Scholar] [CrossRef]
  27. Rozet, E.; Lebrun, P.; Hubert, P.; Debrus, B.; Boulanger, B. Design Spaces for Analytical Methods. TrAC-Trends Anal. Chem. 2013, 42, 157–167. [Google Scholar] [CrossRef]
  28. Patel, M.N.; Kothari, C.S. Review on Implementation of Multivariate Approach for Forced Degradation Study and Impurity Profiling with Regulatory Considerations. Chromatographia 2018, 81, 105–125. [Google Scholar] [CrossRef]
  29. Parr, M.K.; Schmidt, A.H. Life Cycle Management of Analytical Methods. J. Pharm. Biomed. Anal. 2018, 147, 506–517. [Google Scholar] [CrossRef]
  30. Tome, T.; Casar, Z. Development of a Unified Reversed-Phase HPLC Method for Efficient Determination of EP and USP Process-Related Impurities in Celecoxib Using Analytical Quality by Design Principles. Molecules 2020, 25, 809. [Google Scholar] [CrossRef]
  31. Orlandini, S.; Pasquini, B.; del Bubba, M.; Pinzauti, S.; Furlanetto, S. Quality by Design in the Chiral Separation Strategy for the Determination of Enantiomeric Impurities: Development of a Capillary Electrophoresis Method Based on Dual Cyclodextrin Systems for the Analysis of Levosulpiride. J. Chromatogr. A 2015, 1380, 177–185. [Google Scholar] [CrossRef]
  32. Frederick, G.; Vogt, A.S.K. Development of Quality-By-Design Analytical Methods. J. Pharm. Sci. 2012, 101, 2271–2280. [Google Scholar] [CrossRef]
  33. Basso, J.; Mendes, M.; Cova, T.F.G.G.; Sousa, J.J.; Pais, A.A.C.C.; Vitorino, C. Analytical Quality by Design (AQbD) as a Multiaddressable Platform for Co-Encapsulating Drug Assays. Anal. Methods 2018, 10, 5659–5671. [Google Scholar] [CrossRef]
  34. International Council for Harmonisation Guideline on Q14 Analytical Procedure Development Draft Version 2022. Available online: https://database.ich.org/sites/default/files/ICH_Q14_Document_Step2_Guideline_2022_0324.pdf (accessed on 3 May 2022).
  35. Elder, D.P.; White, A.; Harvey, J.; Teasdale, A.; Williams, R.; Covey-Crump, E. Mutagenic Impurities: Precompetitive/Competitive Collaborative and Data Sharing Initiatives. Org. Process Res. Dev. 2015, 19, 1476–1494. [Google Scholar] [CrossRef]
  36. Amberg, A.; Beilke, L.; Bercu, J.; Bower, D.; Brigo, A.; Cross, K.P.; Custer, L.; Dobo, K.; Dowdy, E.; Ford, K.A.; et al. Principles and Procedures for Implementation of ICH M7 Recommended (Q)SAR Analyses. Regul. Toxicol. Pharmacol. 2016, 77, 13–24. [Google Scholar] [CrossRef] [Green Version]
  37. International Council for Harmonisation Guideline on Q2 (R2) Validation of Analytical Procedures: Draft Version 24 March 2022. Available online: ICH_Q2-R2_Document_Step2_Guideline_2022_0324.pdf (accessed on 10 August 2022).
  38. Lhasa Website. Available online: https://www.lhasalimited.org (accessed on 20 July 2022).
  39. Vega Hub Website. Available online: https://www.vegahub.eu (accessed on 20 May 2022).
  40. S-Matrix Website. Available online: https://www.smatrix.com (accessed on 20 July 2022).
Figure 1. Potential synthetic route and structures of MCS, ECS and ICS.
Figure 1. Potential synthetic route and structures of MCS, ECS and ICS.
Separations 09 00246 g001
Figure 2. Model term ranking Pareto Chart. (a) Peak height of MCS; (b) Peak Height of ECS; (c) Peak height of ICS; (d) Resolution of between MCS and ECS; (e) Resolution of between ECS and ICS; (f) retention time of ICS. A: Effect of flow rate of carrier gas; B: Effect of column oven temperature; C: Effect of inlet temperature; D: Effect of detector temperature, E: Effect of Injection volume; Blue bar: Positive effect; Grey bar: Negative effect.
Figure 2. Model term ranking Pareto Chart. (a) Peak height of MCS; (b) Peak Height of ECS; (c) Peak height of ICS; (d) Resolution of between MCS and ECS; (e) Resolution of between ECS and ICS; (f) retention time of ICS. A: Effect of flow rate of carrier gas; B: Effect of column oven temperature; C: Effect of inlet temperature; D: Effect of detector temperature, E: Effect of Injection volume; Blue bar: Positive effect; Grey bar: Negative effect.
Separations 09 00246 g002
Figure 3. 3D response surface plots for each response as a function of flow rate and column oven temperature at 280 °C of inlet temperature. (a) peak height of MCS; (b) peak height of ECS; (c) peak height of ICS; (d) resolution between MCS and ECS; (e) resolution between ECS and ICS; (f) retention time of ICS.
Figure 3. 3D response surface plots for each response as a function of flow rate and column oven temperature at 280 °C of inlet temperature. (a) peak height of MCS; (b) peak height of ECS; (c) peak height of ICS; (d) resolution between MCS and ECS; (e) resolution between ECS and ICS; (f) retention time of ICS.
Separations 09 00246 g003
Figure 4. Method operable design region (MODR) and proven acceptable ranges (PARs).
Figure 4. Method operable design region (MODR) and proven acceptable ranges (PARs).
Separations 09 00246 g004
Figure 5. Typical GC-FID chromatogram of standard solution. (1) internal standard (Diphenyl ether), (2) MCS, (3) ECS and (4) ICS.
Figure 5. Typical GC-FID chromatogram of standard solution. (1) internal standard (Diphenyl ether), (2) MCS, (3) ECS and (4) ICS.
Separations 09 00246 g005
Figure 6. Typical GC-MS chromatogram of standard solution. (1) internal standard (Diphenyl ether), (2) MCS, (3) ECS and (4) ICS.
Figure 6. Typical GC-MS chromatogram of standard solution. (1) internal standard (Diphenyl ether), (2) MCS, (3) ECS and (4) ICS.
Separations 09 00246 g006
Table 1. The genotoxicity predictions for alkyl camphorsulfonates.
Table 1. The genotoxicity predictions for alkyl camphorsulfonates.
NameDerek
Prediction
Sarah
Prediction
Vega
Prediction
Overall
In Silico *
ICH M7
Class *
CSANegativeNegativeNegativeNegativeClass 5
MCSPositiveNegativeNegativePositiveClass 3
ECSPositiveEquivocalPositivePositiveClass 3
ICSPositivePositivePositivePositiveClass 3
* Overall in silico result and ICH M7 class were automatically calculated by Lhasa Nexus program.
Table 2. Design of Experiment for screening.
Table 2. Design of Experiment for screening.
RunFlow Rate (mL/min)Oven Temp (°C)Inlet Temp (°C)Detector Temp (°C)Inject Vol (µL)
11.51703403302.2
21.52303403301.8
31.51701803301.8
42.51703402702.2
52.51701803302.2
62.51701802701.8
72.52301803301.8
82.52303402701.8
92.02002803002.0
102.02002803002.0
112.52303403302.2
121.52303402702.2
132.52301802702.2
141.52301802701.8
151.52301803302.2
162.02002803002.0
171.51703402701.8
181.51701802702.2
192.51703403301.8
Table 3. Response corresponding DoE.
Table 3. Response corresponding DoE.
RunR1R2R3R4R5
12.061.750.4614.346.74
24.154.260.594.281.98
31.571.321.1414.086.61
42.752.250.8313.466.19
52.722.192.0012.925.95
62.321.891.6813.316.21
75.285.393.573.931.97
85.435.671.374.051.94
94.203.933.098.083.94
104.233.963.148.153.97
116.396.601.643.911.87
124.484.670.654.031.84
135.895.903.853.731.87
143.603.722.014.052.02
154.454.602.474.042.02
164.274.003.128.284.03
171.681.400.3814.616.85
181.921.651.4214.046.65
192.281.840.7213.566.20
R1 = peak height of MCS, R2 = peak height of ECS, R3 = peak height of ICS, R4 = resolution between MCS and ECS, R5 = resolution between ECS and ICS.
Table 4. Statistical significance for each response.
Table 4. Statistical significance for each response.
R2Adj. R2Sum of SquaresDegree of FreedomMean SquareF-Ratiop-Value
MCSHeight0.99950.998738.8914103.88911334<0.0001
ECSHeight0.99980.999551.1069105.11073658<0.0001
Resolution0.99990.9998385.4958755.070815098<0.0001
ICSHeight0.99990.999714.8934121.24113349<0.0001
Resolution0.99970.999581.2602613.54345475.8411<0.0001
Table 5. Design of Experiment for Optimization.
Table 5. Design of Experiment for Optimization.
RunFlow Rate (mL/min)Oven Temp (°C)Inlet Temp (°C)
12220280
22200300
32.2220300
41.8180300
51.8200280
62.2180260
72200260
82180280
91.8220300
102200280
112200280
122200280
132.2180300
141.8220260
152.2200280
162.2220260
171.8180260
Table 6. Response Corresponding DoE.
Table 6. Response Corresponding DoE.
RunR1R2R3R4R5R6
13.224.112.684.912.432.95
22.743.352.238.213.964.60
33.534.582.675.162.532.75
41.751.983.1912.324.928.46
52.463.042.068.374.044.96
61.992.241.6811.815.507.58
72.693.312.438.474.094.61
81.842.122.0412.096.368.10
93.094.032.075.462.653.17
102.603.212.158.274.004.60
112.753.342.258.604.114.60
122.693.282.298.364.104.60
131.982.251.4612.015.547.57
142.903.742.185.222.583.17
152.713.242.388.053.864.03
163.734.703.235.552.722.76
171.741.973.9412.234.848.46
R1 = peak height of MCS, R2 = peak height of ECS, R3 = peak height of ICS, R4 = resolution between MCS and ECS, R5 = resolution between ECS and ICS, R6 = retention time of ICS.
Table 7. Statistical significance for each response.
Table 7. Statistical significance for each response.
R2Adj. R2Sum of SquaresDegree of FreedomMean SquareF-Ratiop-Value
R10.99540.99085.702580.7128215<0.0001
R20.99340.986811.972781.4966150<0.0001
R30.99230.98245.723890.636100<0.0001
R40.99430.9935117.0834258.54171,224<0.0001
R50.99710.994823.400273.3429434<0.0001
R61.00001.00009076.3381,134.54672,190<0.0001
R1 = peak height of MCS, R2 = peak height of ECS, R3 = peak height of ICS, R4 = resolution between MCS and ECS, R5 = resolution between ECS and ICS, R6 = retention time of ICS.
Table 8. Applicability of the method on the API Amlodipine camsylate.
Table 8. Applicability of the method on the API Amlodipine camsylate.
Present Amount in API (P)Theoretical Spiked Amount (T)Actual Detection
Amount
(A)
Recovery(%)
((A − P) /T × 100)
Precision
(%RSD)
Effect of API
FIDMCS0 ppm120 ppm120.5 ppm100.4%
122.1 ppm101.8%0.76%None
122.1 ppm101.8%
0 ppm150 ppm152.4 ppm101.6%2.11%None
152.5 ppm101.7%
146.9 ppm97.9%
0 ppm180 ppm179.4 ppm99.7%
179.7 ppm99.8%0.73%None
177.3 ppm98.5%
ECS 123.5 ppm102.9%
0 ppm120 ppm118.6 ppm98.8%2.06%None
120.4 ppm100.3%
0 ppm150 ppm150.5 ppm100.4%2.10%None
152.6 ppm101.7%
146.4 ppm97.6%
177.9 ppm98.9%
0 ppm180 ppm179.9 ppm99.9%1.17%None
175.7 ppm97.6%
ICS 122.6 ppm102.2%
0 ppm120 ppm121.6 ppm101.4%1.11%None
120.0 ppm100.0%
0 ppm150 ppm155.7 ppm103.8%0.64%None
153.7 ppm102.5%
154.8 ppm103.2%
186.9 ppm103.8%
0 ppm180 ppm18.3 ppm101.8%0.96%None
185.3 ppm102.9%
MS 114.5 ppm95.4%
0 ppm120 ppm110.1 ppm 91.8%4.06%None
105.5 ppm87.9%
MCS0 ppm150 ppm135.8 ppm90.5%0.83%None
134.8 ppm89.9%
133.6 ppm89.1%
168.2 ppm93.5%
0 ppm180 ppm167.0 ppm92.8%0.35%None
167.6 ppm93.1%
117.0 ppm97.5%
0 ppm120 ppm111.0 ppm92.5%4.94%None
106.0 ppm88.3%
ECS0 ppm150 ppm135.4 ppm90.3%0.93%None
133.9 ppm89.3%
132.9 ppm88.6%
167.2 ppm92.9%
0 ppm180 ppm166.0 ppm92.2%0.36%None
166.3 ppm92.4%
139.9 ppm116.6%
0 ppm120 ppm132.6 ppm110.5%5.79%None
124.6 ppm103.8%
ICS0 ppm150 ppm153.3 ppm102.2%2.47%None
149.7 ppm99.8%
145.9 ppm97.3%
178.2 ppm99.0%
0 ppm180 ppm174.8 ppm97.1%1.35%None
173.6 ppm96.5%
Table 9. Validation results summary.
Table 9. Validation results summary.
Validation ParametersFIDMS
MCSECSICSMCSECSICS
DL QLDL1.5 ppm1.5 ppm1.9 ppm0.055 ppm0.069 ppm0.102 ppm
QL4.9 ppm5.1 ppm6.4 ppm0.185 ppm0.232 ppm0.340 ppm
LinearityR20.999530.999880.999830.995320.994120.99370
Slope0.005010.005440.004940.005340.005470.00391
Y-intercept0.012000.001920.018380.079470.090330.06289
AccuracyLow conc. (10 ppm)95.3%102.6%96.4%94.5%94.9%93.9%
Mid conc. (120 ppm)101.3%100.7%101.2%91.7%92.8%110.3%
Mid conc. (150 ppm)100.4%99.9%103.2%89.8%89.4%99.8%
High conc. (180 ppm)99.3%98.8%102.9%93.1%92.5%97.5%
Precision
Acceptance
criteria
(%RSD ≤ 10)
Low conc. (10 ppm)1.14%2.50%5.58%3.16%4.92%1.73%
Low conc. (30 ppm)7.79%6.97%6.64%3.14%3.36%2.49%
Mid conc. (150 ppm)2.27%3.22%8.83%2.16%2.21%2.47%
SpecificityResolution 9.514.53 10.214.78
Table 10. Robustness of the analytical methods.
Table 10. Robustness of the analytical methods.
ConditionParameterFIDMS
MCSECSICSMCSECSICS
1Precision (%RSD)2.27%3.22%8.83%2.16%2.21%2.47%
Precision (SDV)0.015580.023040.055390.016520.017060.01348
Resolution 9.514.53 10.214.78
Tailing factor0.900.910.990.971.000.95
2Precision (%RSD)2.07%1.20%2.73%0.97%1.16%1.40%
Precision (SDV)0.014100.008550.016040.007330.008930.00804
Resolution 9.074.43 9.974.66
Tailing factor0.880.901.000.960.970.97
3Precision (%RSD)1.84%1.63%9.06%3.03%3.49%3.83%
Precision (SDV)0.012490.011530.058020.021950.025700.02168
Resolution 9.124.33 9.594.49
Tailing factor0.870.941.020.970.980.99
4Precision (%RSD)2.27%1.28%1.29%2.16%2.70%2.38%
Precision (SDV)0.015260.009090.008270.016350.020800.01496
Resolution 10.034.68 10.564.93
Tailing factor0.900.931.041.021.000.94
5Precision (%RSD)1.24%0.96%1.68%2.91%3.49%3.86%
Precision (SDV)0.008330.006770.011200.021840.027070.02405
Resolution 7.903.80 8.854.15
Tailing factor0.850.900.970.900.970.95
Condition 1: flow rate 2.0 mL/min, oven temp 200 °C (Normal condition); Condition 2: flow rate 1.8 mL/min, oven temp 200 °C; Condition 3: flow rate 2.2 mL/min, oven temp 200 °C; Condition 4: flow rate 2.0 mL/min, oven temp 195 °C, Condition 5: flow rate 2.0 mL/min, oven temp 205 °C.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Lee, K.; Yoo, W.; Jeong, J.H. Determination of Three Alkyl Camphorsulfonates as Potential Genotoxic Impurities Using GC-FID and GC-MS by Analytical QbD. Separations 2022, 9, 246. https://0-doi-org.brum.beds.ac.uk/10.3390/separations9090246

AMA Style

Lee K, Yoo W, Jeong JH. Determination of Three Alkyl Camphorsulfonates as Potential Genotoxic Impurities Using GC-FID and GC-MS by Analytical QbD. Separations. 2022; 9(9):246. https://0-doi-org.brum.beds.ac.uk/10.3390/separations9090246

Chicago/Turabian Style

Lee, Kyoungmin, Wokchul Yoo, and Jin Hyun Jeong. 2022. "Determination of Three Alkyl Camphorsulfonates as Potential Genotoxic Impurities Using GC-FID and GC-MS by Analytical QbD" Separations 9, no. 9: 246. https://0-doi-org.brum.beds.ac.uk/10.3390/separations9090246

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