Authors:
Senem Şanlı Department of Medical Services and Techniques, Vocational School of Health Services, Usak University, Usak, Turkey

Search for other papers by Senem Şanlı in
Current site
Google Scholar
PubMed
Close
,
Meryem Cansu Şahin Department of Medical Services and Techniques, Vocational School of Health Services, Usak University, Usak, Turkey

Search for other papers by Meryem Cansu Şahin in
Current site
Google Scholar
PubMed
Close
,
Ayşe Özdemir Department of Biochemisty, Faculty of Medicine, Usak University, Usak, Turkey

Search for other papers by Ayşe Özdemir in
Current site
Google Scholar
PubMed
Close
, and
Buket Paşa Department of Medical Services and Techniques, Vocational School of Health Services, Usak University, Usak, Turkey

Search for other papers by Buket Paşa in
Current site
Google Scholar
PubMed
Close
Open access

Abstract

A straightforward, dependable, and quick RP-LC method for the analysis of abiraterone acetate in its dose form and human urine has been devised. With DAD detection, sensitivity was reported to be high. The LOD and LOQ of the procedure were deemed adequate. The suggested approach was exhaustively validated in accordance with ICH requirements, and the findings demonstrated that it was exact, accurate, selective, and sensitive for the analysis of this pharmaceutical. The chromatographic separation was realized using a X-Terra RP-18 (150 × 4.60 mm i.d. × 5 μm) column and a UV detector set at 255 and 267 nm. In addition, pKa values were calculated based on the relationship between the retention factor and the pH of the mobile phase. The influence of the composition of the mobile phase on the ionization constant was investigated by measuring the pKa at various acetonitrile–water combinations ranging from 50 to 70% (v/v).

Abstract

A straightforward, dependable, and quick RP-LC method for the analysis of abiraterone acetate in its dose form and human urine has been devised. With DAD detection, sensitivity was reported to be high. The LOD and LOQ of the procedure were deemed adequate. The suggested approach was exhaustively validated in accordance with ICH requirements, and the findings demonstrated that it was exact, accurate, selective, and sensitive for the analysis of this pharmaceutical. The chromatographic separation was realized using a X-Terra RP-18 (150 × 4.60 mm i.d. × 5 μm) column and a UV detector set at 255 and 267 nm. In addition, pKa values were calculated based on the relationship between the retention factor and the pH of the mobile phase. The influence of the composition of the mobile phase on the ionization constant was investigated by measuring the pKa at various acetonitrile–water combinations ranging from 50 to 70% (v/v).

1 Introduction

Prostate cancer is the second leading cause of cancer-related mortality among men, after lung cancer [1]. Cancer of the prostate is primarily a hormone-dependent tumor. The production of testosterone, which promotes tumor growth, is inhibited by ADT. After chemotherapy, the conventional use of abiraterone acetate for prostate cancer is to treat castration-resistant prostate cancer [2].

Abiraterone acetate is a derivative of steroidal progesterone (Fig. 1). A selective inhibitor of the testosterone-producing enzymes cytochrome P450 17x-hydrolyase/17 and CYP17, CYP complex [3–5]. Abiraterone acetate is classified as an adrenal inhibitor and is used to treat advanced prostate and breast cancer with hormone therapy [6]. Male hormones (testosterones/androgens) promote the development of prostate cancer. Zytiga, a tablet formulation containing Abiraterone acetate and prednisone, has been granted approval by both the US-FDA and EMA for the treatment of metastatic castration-resistant prostate cancer (mCRPC) in males who have previously undergone initial chemotherapy with docetaxel or ketakonazole [7, 8].

Fig. 1.
Fig. 1.

Chemical structure of abiraterone acetate

Citation: Acta Chromatographica 36, 4; 10.1556/1326.2023.01173

Since it aids in the explanation of chemical processes like chemical absorption, distribution, and elimination, the acid-base dissociation constant, or pKa, has long been recognized as an important metric in research on ADME and toxicity. This is because it can explain chemical phenomena such as how chemicals are absorbed, distributed, and eliminated. This notable variable possesses numerous applications within research domains, including but not limited to the advancement of medicinal compounds, solvent separation, acid-base volumetric analysis, and ion conveyance. The toxicity, chromatographic retention behavior, and medicinal qualities of organic acids and bases are influenced by their acid-base characteristics. Pharmaceutical compounds are commonly classified as either weak acids or weak bases, and their absorption primarily occurs through passive diffusion of the non-ionized component. Hence, it holds significant importance to ascertain the ionization state of a drug at a specific pH. The pharmacokinetic behavior of a drug compound within an organism is contingent upon the dissociation constant of said compound. Therefore, accurate determination of the drug dissociation constant of a pharmaceutical compound is crucial. Furthermore, while RP-LC separation methods have employed mobile phases consisting of acetonitrile (ACN) and water, the pKa of abiraterone in binary mixtures of ACN and water remains undetermined. Only two prediction programs values could be found for abiraterone [9, 10].

There have been numerous publications on the determination of this drug, including the use of HPLC [11–16], liquid chromatography–tandem mass spectrometric technique (LC–MS/MS) [17–20], and ultra-performance liquid chromatography (UPLC) [21].

The utilization of RP-LC has been established as a potent method for the determination of dissociation constants. This is attributed to its ability to operate with minimal quantities of compounds, its capacity to analyze impure samples, and its ability to overcome the limitations of poor water solubility, as reported in references [22–24]. This methodology necessitates solely the retention times, without the need for measuring solute or titrant concentrations. Furthermore, the computation process is uncomplicated and unaffected by the level of impurities in the solute.

The present study is centered on the assessment of pKa values of abirateron in various ACN-water mixtures, namely 50, 60, and 70% v/v. The objective of this research is to address the dearth of knowledge pertaining to the acid-base equilibria of such compounds through chromatographic measurements.

Despite the widespread use of abiraterone acetate in the treatment of prostate cancer, there is currently no published methodology for the determination of abiraterone acetate in human urine samples using RP-LC techniques with UV detection, as far as our knowledge extends. Abiraterone was successfully determined in both dosage form and human urine sample using the optimal separation conditions obtained from pKa studies.

2 Experimental section

2.1 Chemicals and reagents

The chemicals were utilized without undergoing additional purification procedures. Abiraterone acetate and everolimus (used as an internal standard) were procured from Sigma-Aldrich (St. Louis MO, USA).

The organic modifier utilized in this study was HPLC grade acetonitrile, which was sourced from Merck in Darmstadt, Germany. The ortho-phosphoric acid utilized in this study, with a minimum purity of 85%, was procured from Riedel (Riedel-de Haen, Seelze, Germany). The chemicals utilized in this study were sodium hydroxide (NaOH), hydrochloric acid (HCl), uracil, and methanol, which were procured from Sigma-Aldrich (St. Louis, MO, USA).

2.2 Apparatus

An Agilent 1260 series HPLC system equipped with a ternary solvent pump, online degasser, automatic injection system, column heater, and multi-wavelength detector was utilized to perform the LC analysis. Abiraterone acetate and everolimus were subjected to UV detection at wavelengths of 255 and 267 nm, respectively. The experiment was conducted with a flow rate of 1 mL min−1, utilizing an X-Terra RP-18 (150 × 4.60 mm i.d. × 5 μm) column as the stationary phase at a temperature of 25 °C. The pH measurements were conducted using a Mettler Toledo MA 235 pH/ion analyzer in conjunction with a Hanna HI 1332 Ag/AgCl combined glass electrode. The selection of Everolimus as the internal standard was made.

2.3 Chromatographic procedure

In the course of this investigation, the mobile phases that were analyzed consisted of ACN-water blends with volumetric ratios of 50%, 60%, and 70%, respectively, and were supplemented with 10 mM o-phosphoric acid. The pH range of the mobile phase was modulated from 2.5 to 7.0 through the incorporation of sodium hydroxide. The injection volume of 20 μL was maintained at a flow rate of 1.0 mL min−1.

Prior to the initial injection, the column underwent a pre-conditioning process for a minimum of one hour at a low flow rate of 0.5 mL min−1, utilizing the mobile phase at the appropriate pH. Retention time values (tR) for each compound were determined through three distinct injections for every mobile phase composition and pH that was taken into account.

The pH of the mobile phase exerts a significant influence on the chromatographic retention of ionizable compounds. Efficient separations of ionizable compounds by RP-LC often necessitate precise measurement and regulation of the mobile phase pH [25].

Various techniques can be employed for the determination of the pH of the mobile phase [26]. Prior to combining the aqueous buffer with the organic modifier, the prevalent approach involves determining the buffer's pH. A more stringent methodology, as advised by the IUPAC, was employed, wherein the pH of the mobile phase was determined subsequent to the combining of the aqueous buffer and organic modifier. The electrode system utilized for pH measurement in this scenario can be calibrated through the utilization of either aqueous buffers or buffers that have been prepared in the same solvent composition employed as the mobile phase. Acquiring knowledge pertaining to the pH value of reference buffers that are formulated in diverse aqueous-organic solvent mixtures is necessary [27].

The determination of the pKa values of the compounds under investigation was carried out by utilizing the non-linear regression program NLREG [28] to analyze the k-pH data pairs. The program in question is a versatile software application that allows for the specification of the objective function and the parameters to be inferred through the utilization of the integrated program editor.
Um=j=1ns(ki,expki,calc)2

Equation (1) is utilized by NLREG to iteratively refine the parameters, resulting in a minimized sum of square residuals (Um) for the purpose of determining the dissociation constants of the compounds under investigation. The equation for ns involves the variables ki,exp and ki,calc, which respectively represent the experimental and calculated values of the retention factor for a given solution i.

2.4 The process of creating standard solutions and developing a calibration plot

A standard stock solution of the drug was prepared through dissolution in MeOH at a concentration of 100 μg mL−1. The preparation of standard solutions for the analysis of pharmaceutical dosage forms involved the use of abiraterone acetate and everolimus, which were dissolved in the mobile phase. The concentrations of these solutions were adjusted within the range of 1.0–20 μg mL−1. The concentration of drugs in urine was subjected to analysis within the range of 1.0–40 μg mL−1. The construction of calibration curves for RP-LC analysis involved plotting the drug-to-IS peak area ratio against the drug concentration. A stable concentration of IS was sustained at 20.0 μg mL−1. Each solution underwent five injections of 20 µL, and the resulting peak area ratio of each drug with respect to the internal standard was graphed against the corresponding concentration to generate a calibration graph. All solutions were shielded from light and utilized within a 24 h period. The measurement of dead time was conducted through the injection of a uracil solution.

The study assessed the ruggedness and precision of the measurements on separate occasions, distinguishing between within-day (n = 5) and between-day (n = 5) variations. The robustness and precision of the method were evaluated by calculating the relative standard deviations [29–31]. The quantitative depiction of the precision and accuracy of the developed techniques is achieved through the utilization of Bias% (relative error).

2.5 The evaluation of pharmaceutical dosage forms

The pharmaceutical formulation of abiraterone acetate, specifically Zytiga 250 mg, was procured from a nearby pharmacy. A total of 20 tablets were subjected to individual weighing, and subsequently, their average weight was determined and pulverized. A quantity of tablet powder, corresponding to 100 mg of abiraterone acetate, was introduced into a 100 mL volumetric flask. Subsequently, 50 mL of mobile phase was added to the flask, and the mixture was subjected to sonication for a duration of 30 min at a controlled temperature to facilitate the dissolution of the powder. The resulting solution was then brought to volume with the same solvent. Subsequent to the aforementioned procedure, extract precisely 1.0 mL of the specimen solution and transfer it into a volumetric flask with a capacity of 10 mL. Proceed to dilute the solution with the mobile phase until it reaches the mark on the flask, ensuring that it is thoroughly mixed. The clear filtrate was divided into suitable aliquots, and the appropriate internal standard (everolimus) solution was added to generate appropriate solutions. The quantity of the substance was determined using the respective regression formula.

2.6 Analysis of spiked urine samples

The urine specimens were collected from asymptomatic participants in a timely manner prior to the commencement of the experimental procedures. The urine sample was divided into equal volumes and transferred into a flask that had been calibrated for accuracy. A specific volume of urine sample was supplemented with a drug that was dissolved in ultrapure water, resulting in a final concentration of 100 μg mL−1. The quantity of internal standard incorporated into the specimens. The drug concentration was altered within the range of 1–40 μg mL−1 in urine samples obtained from human subjects. The solutions underwent filtration and analysis through RP-LC methodology.

3 Results and discussions

3.1 Determination of dissociation constants (pKa)

Determining the dissociation constants (pKa) of drugs is a fundamental step in pharmaceutical research. Accurate knowledge of pKa values aids in drug design, formulation development, understanding pharmacokinetics, optimizing drug-target interactions, and ensuring therapeutic efficacy and safety. By comprehensively characterizing the ionization behavior of drugs, researchers can make informed decisions that contribute to the successful development of effective and safe medications.

It is customary in contemporary drug development to produce compounds that exhibit high lipophilicity and moderate aqueous solubility. The insolubility of pharmaceuticals in water is frequently considered a disadvantage when evaluating their physicochemical characteristics. The solubility of the drugs under investigation can be enhanced by the addition of an organic co-solvent to the aqueous mobile phase, as reported in previous studies [32, 33]. The present investigation employed LC technology to examine the correlation between the retention of abiraterone and the pH of the mobile phase. Empirical data were employed to determine the pKa values and intrinsic capacity factors. The pKa values were determined by analyzing k/pH data pairs with the aid of the NLREG software [28]. The pKa value of abiraterone is summarized in Table 1.

Table 1.

The pKa values of abiraterone obtained by chromatographic method in ACN–water media at 25 °C

Prediction programsYasuda–ShedlovskyXACNNLREG
50% (v/v) ACN60% (v/v) ACN70% (v/v) ACN
Abiraterone4.81*

5.50**
4.354.784.13 ± 0.093.90 ± 0.083.72 ± 0.09

* http://www.chemicalize.org. (Accessed 26 April 2023).

** Pan X, Wang H, Li C, et al. MolGpka: A Web Server for Small Molecule pK(a) Prediction Using a Graph-Convolutional Neural Network. J Chem Inf Model 2021; 61(7): 3159–3165. doi:10.1021/acs.jcim.1c00075.

The obtained curves exhibited typical sigmoidal patterns, indicating the correlation between the pH of the mobile phase and the capacity factors of the analyte. Figure 2 illustrates the instances of abiraterone acetate present in the mobile phase, which comprises 70% ACN-water (v/v).

Fig. 2.
Fig. 2.

Capacity factors of the abiraterone are plotted as a function of the mobile phase pH using NLREG program

Citation: Acta Chromatographica 36, 4; 10.1556/1326.2023.01173

The pKa value of a compound in water may be estimated from the known pKa values of the corresponding ACN-water binary combinations using the Yasuda–Shedlovsky (Y–S) equation. Equation (2) displays the results of the Y–S method's application to the determination of the aqueous pKa [34, 35].
pKa+log[H2O]=a1ε+b

The logarithm of the water molar concentration in a solvent combination is denoted by log[H2O], while the dielectric constant of the mixture is denoted by ε. The values of ε for various ACN-water mixtures were calculated by Barbosa et al. [36]. The values of the substance under study were calculated using linear equations created by plotting pKa + log[H2O] data against 1/ε values. If you plot pKa versus 1/ε, you should be able to extrapolate to pure water to get the aqueous pKa of the compounds [33]. The second method is based on the linear connections between the pKa values of the chemicals examined and the mole fraction of ACN (XACN) at 25 °C. The dissociation process is regulated not just by electrostatic interactions but also by particular solute-solvent interactions, which may account for variations in pKa values. Electrostatic interactions are more powerful than solute-solvent interactions, which explains why this is the case [37]. The intercept of linear equations gives the values of the thermodynamic pKa of the substances in question when dissolved in water. Table 1 displays the pKa values calculated using the two different approaches.

As can be seen in Fig. 3, the plots follow a linear pattern with a range of slopes. These straight lines may be clarified and made clearer by factoring in the structural properties of the ACN + water mixtures. Figure 3 displays Y–S graphs for researched chemotherapeutic drugs that comprise basic functional groupings. Both figures show that the slopes of the main functional categories are all negative.

Fig. 3.
Fig. 3.

Yasuda–Shedlovsky and mole fraction plots for the abiraterone

Citation: Acta Chromatographica 36, 4; 10.1556/1326.2023.01173

3.2 Quantitative determination

The present study aimed to determine the optimal chromatographic conditions for RP-LC analysis of drugs. To achieve this, the impact of pH variations in the mobile phase and column temperature was investigated. Multiple pH values ranging from 2.5 to 7.0 were evaluated, and after careful analysis, a pH of 2.5 was determined to be the optimal value based on its superior peak asymmetry and retention values. The pH of the mobile phase has consistently been modified by utilizing a concentration of 10 mM orthophosphoric acid. The temperature of the column was maintained within the range of 25–30 °C. Research has demonstrated that elevating the temperature can mitigate the phenomenon of drug tailing. The selection of 25 °C was based on the advantages of reduced analysis time and enhanced peak morphology. Various mobile phase compositions ranging from 50 to 70% (v/v) were evaluated. The optimal carrier for RP-LC analysis was determined to be a mobile phase consisting of ACN and water in a 70:30 (v/v) ratio, with the addition of 10 mM H3PO4 at pH 2.5 and 4 for pharmaceutical dosage forms and urine, respectively. The flow rate was set at 1.0 mL per minute.

Using the conditions described above, a satisfactory chromatographic peak resolution was obtained in a short analysis time (4 min).

3.3 Analysis of a spiked urine sample

The method was verified in accordance with the ICH recommendations for the validation of analytical methods [3031, 38]. Table 2 presents the validation data pertaining to the linearity range, regression equations, correlation coefficient, LOD, and LOQ values for pharmaceutical dosage forms and urine analysis. The quantitation of the studied compound was performed by utilizing the ratio of the peak area of the compound to the internal standard (IS) against its concentration.

Table 2.

Statistical evaluation of the calibration data of abiraterone acetate for urine and pharmaceutical analysis

UrinePharmaceutical dosage form
Linearity range (µg mL−1)1.0–40 (n = 5)1–20 (n = 5)
Slope0.03970.0392
Intercept0.02110.0363
Correlation coefficient (r)0.9990.999
Detection limit (LOD) (µg mL−1)0.0990.074
Quantitation limit (LOQ) (µg mL−1)0.3000.223

The precision and reproducibility of the method were assessed through the analysis of standard solutions in the mobile phase, with replicates being performed. Table 3 presents a summary of the mean recovery and RSD, which were utilized to characterize the repeatability and reproducibility. Table 3 indicates that there was no statistically significant difference observed for the assay, as determined through within-day and between-day testing. The RSD percentage values ranged from 0.111 to 0.486 for the concentrations under investigation. The findings of this study indicate that the method exhibits a high degree of precision.

Table 3.

Summary of repeatability (intra-day) and reproducibility (inter-day) precision data for abiraterone acetate

ConcentrationIntra-dayInter-day
Mean Recovery* % ± RSD %Mean Recovery* % ± RSD %
Dosage Form1100.410 ± 0.351100.773 ± 0.486
10100.094 ± 0.111100.120 ± 0.312
Urine5100.621 ± 0.337100.643 ± 0.422
2099.989 ± 0.141100.144 ± 0.171

*Each value is obtained from five experiments (n = 5).

The method proposed here was employed for the analysis of drugs present in pharmaceutical dosage forms as well as in human urine specimens. The findings derived from the drug analysis demonstrate that the suggested approach is suitable for the quantification and regular quality assessment of drugs in commercial specimens. The observed disparities between the claimed quantities and the measured quantities were minimal, and the relative standard deviation (RSD%) values were deemed satisfactory. The label of abiraterone acetate indicated a quantity of 250.000 mg, whereas the measured amount was 249.873 mg, accompanied by a relative standard deviation of 0.095 based on a sample size of 5. Following five iterations of experimentation, the average percentage recoveries were determined to be 100.075, exhibiting RSD% values of 0.0595. To assess the suitability of the approach for biological specimens, recovery experiments were conducted for the quantification of drugs in human urine using the calibration curve technique, resulting in the attainment of the designated percentage recoveries. The investigated drug was added to urine samples that had no detectable levels of the substance. Following five iterations of experimentation, the average percentage recoveries for urine samples were determined to be 100.022, with relative standard deviation (RSD%) values of 0.035. Figure 4a and b indicate the customary chromatograms of a pharmaceutical dosage form and a urine sample spiked with the substance, respectively.

Fig. 4.
Fig. 4.

Typical chromatograms Pharmaceutical dosage form a) and for human urine b) spiked with abiraterone acetate (1), everolimus (2) (IS). (Chromatographic condition as: ACN and water in a 70:30 (v/v) ratio, with the addition of 10 mM H3PO4 at pH 2.5 and 4.0 for pharmaceutical dosage forms and urine)

Citation: Acta Chromatographica 36, 4; 10.1556/1326.2023.01173

The analysis of pharmacological dosage forms was conducted at a pH of 2.5, while urine analysis was carried out at a pH of 4.0 due to the presence of an unidentified peak occurring at around 1.5 min. Due to this factor, the retention times exhibit variation. The chromatogram acquired from the urine sample does not exhibit any insignificant peaks. The accuracy and precision of the pharmaceutical formulation and urine were determined through recovery studies utilizing the standard addition method subsequent to their analysis. The results indicate that the proposed method for determining abiraterone acetate in human urine is both reproducible and sensitive. This is evidenced by the high percentage of recovery data and low RSD% values, which suggest that the method is free from interference and easy to perform.

In the literature, there is no method for the determination of abiraterone acetate in human urine samples using RP-LC techniques with UV detection. Sankar et al. [16] determined abiraterone acetate in pharmaceutical dosage form and this method required 7.45 min for analysis time. For this method the limit of detection and the limit of quantitation was found to be 0.451 μg mL−1 and 1.369 μg mL−1 respectively. Reddy et al. [13] was developed RP-HPLC method for the determination assay of abiterone acetate in pharmaceutical with the analysis time of about 5 min. The limits of detection and quantification were 0.23 and 0.70 μg mL. The method described above provides a combination of faster analysis time and improved limits of detection.

4 Conclusions

The RP-LC method that has been put forth allows for the quantification of abiraterone acetate in both human urine samples and dosage forms. This method is characterized by its simplicity, speed, high sensitivity, accuracy, and precision. The X-Terra column facilitated an analysis time of less than 5 min for the compound under study. The report on validation affirms that the proposed technique is suitable as a standard method for detecting the drug in formulations, end products, or exposure investigations.

Furthermore, this manuscript introduces the first investigation pertaining to the determination of pKa values of abiraterone through the employment of RP-LC methodology in ACN-water binary mixtures. The determined pKa values are suitable for estimating the effect of eluent pH on retention.

Conflict of interest statement

The authors have declared that they have no conflicts of interest.

Acknowledgments

The authors are thankful for the financial aid received from the Scientific and Technological Research Council of Turkiye (TUBITAK). We gratefully acknowledge Dr. Jose Luis Beltran from the University of Barcelona for kindly providing the NLREG 4.0 program.

References

  • 1.

    Siegel, R.; Ma, J.; Zou, Z.; Jemal, A. Cancer statistics. Cancer J. Clin. 2014, 64(1), 929. https://doi.org/10.3322/caac.21208.

  • 2.

    Fizazi, K.; Scher, H. I.; Molina, A.; Logothetis, C.; Chi, K. N.; Jones, R.; Staffurth, J.; North, S.; Vogelzang, N.; Saad, F.; Mainwaring, P.; Harland, S.; Goodman, O.; Sternberg, C.; Li, J.; Kheoh, T.; Haqq, C.; Bono, J. Abiraterone acetate for treatment of metastatic castration-resistant prostate cancer: final overall survival analysis of the COU-AA-301 randomised, double-blind, placebo-controlled phase 3 study. Lancet Oncol. 2012, 13(10), 983992. https://doi.org/10.1016/s1470-2045(12)70379-0.

    • Search Google Scholar
    • Export Citation
  • 3.

    Rowlands, M. G.; Barrie, S. E.; Chan, F.; Houghton, J.; Jarman, M.; McCague, R.; Potter, G. A Esters of 3-pyridylacetic acid that combine potent inhibition of 17 alpha-hydroxylase/C17,20-lyase (cytochrome P45017 alpha) with resistance to esterase hydrolysis. J. Med. Chem. 1995, 38(21), 41914197. https://doi.org/10.1021/jm00021a008.

    • Search Google Scholar
    • Export Citation
  • 4.

    Hunt, N. J. Methanesulfonate salts of abiterone-3-esters and recovery of salts of Abiraterone one-3-esters from solution in methyl tetra-butyl ether, 2010, (United States). Pub. No. US 2010/0152437 A1.

    • Search Google Scholar
    • Export Citation
  • 5.

    Sonpavde, G.; Attard, G.; Bellmunt, J.; Mason, M. D.; Malavaud, B.; Tombal, B.; Sternberg, C. N. The role of abiraterone acetate in the management of prostate cancer: a critical analysis of the literature. Eur. Urol. 2011, 60(2), 270278. https://doi.org/10.1016/j.eururo.2011.04.032.

    • Search Google Scholar
    • Export Citation
  • 6.

    Acharya, M.; Bernard, A.; Gonzalez, M.; Jiao, J.; De Vries, R.; Tran, N. Open-label, phase I, pharmacokinetic studies of abiraterone acetate in healthy men. Cancer Chemother. Pharmacol. 2012, 69(6), 15831590. https://doi.org/10.1007/s00280-012-1865-3.

    • Search Google Scholar
    • Export Citation
  • 7.

    O’Donnell, A.; Judson, I.; Dowsett, M.; Raynaud, F.; Dearnaley, D.; Mason, M.; Harland, S.; Robbins, A.; Halbert, G.; Nutley, B.; Jarman, M. Hormonal impact of the 17alpha-hydroxylase/C(17,20)-lyase inhibitor abiraterone acetate (CB7630) in patients with prostate cancer. Br. J. Cancer 2004, 90(12), 23172325. https://doi.org/10.1038/sj.bjc.6601879.

    • Search Google Scholar
    • Export Citation
  • 8.

    Sarker, H.; Miksinski, S. United States food and drug administration, center for drug evaluation and research, chemistry review (S), application number: 202379, 2011. (United States Food and Drug Administration), Pub. No. 202379 Orig 1s000, NAD, 202379.

    • Search Google Scholar
    • Export Citation
  • 9.

    Pan, X.; Wang, H.; Li, C.; Zhang, J. Z. H; Ji, C. MolGpka: a Web server for Small molecule pK(a) prediction using a graph-convolutional neural Network. J. Chem. Inf. Model 2021, 61(7), 31593165. https://doi.org/10.1021/acs.jcim.1c00075.

    • Search Google Scholar
    • Export Citation
  • 10.

    http://www.chemicalize.org. (2023). (Accessed 26 April 2023).

  • 11.

    Arunkumar, S. G.; Radha, G. V. Analytical method development and validation for theestimation of Abiraterone acetate in pharmaceutical formulation by UV and RP-HPLC. Int. J. Pharm. Bio. Sci. 2016, 7(2), 5761.

    • Search Google Scholar
    • Export Citation
  • 12.

    Latha, P. G.; Yunus, S. B.; Sree, N. B.; Naz, J.; Sagar, M.; Shine, S. A review on analytical methods for estimation of abiraterone acetate. World J. Pharm. Med. Res. 2016, 2, 23.

    • Search Google Scholar
    • Export Citation
  • 13.

    Reddy, B. J. C.; Sarada, N. J. Development and validation of a novel RP-HPLC method for stability-indicating assay of Abiraterone acetate. J. Liquid Chromatogr. & Relat. Tech. 2016, 39(7), 354363. https://doi.org/10.1080/10826076.2016.1163500.

    • Search Google Scholar
    • Export Citation
  • 14.

    Gadhave, R. V.; Tamnar, B. A.; Bansode, S. A.; Choudhari, V. P. Stability indicating RP-HPLC-PDA method for determination of abıraterone acetate and characterization of its base catalyzed degradation product by LC-MS. Int. J. Pharm. Pharm. Sci. 2016, 8(2), 7681.

    • Search Google Scholar
    • Export Citation
  • 15.

    Kavitapu, D.; Maruthapillai, A.; Mahapatra, S.; Selvi, J. A. New stability indicating RP-HPLC method for the determination of Abiraterone acetate, its related substances and degradation products in bulk and dosage form. Mater. Today Proc. 2021, 34, 469478.

    • Search Google Scholar
    • Export Citation
  • 16.

    Sankar, P. R.; Eswarudu, M. M.; Siva, P.; Viswanath, A. Optimized and validated RP-HPLC method for quantification of abıraterone acetate (an antı- prostate cancer drug) in pharmaceutıcal dosage form. High Technol. Lett. 2021, 27(7), 21.

    • Search Google Scholar
    • Export Citation
  • 17.

    Alyamani, M.; Li, Z.; Upadhyay, S. K.; Anderson, D. J.; Auchus, R. J.; Sharifi, N. Development and validation of a novel LC-MS/MS method for simultaneous determination of abiraterone and its seven steroidal metabolites in human serum: innovation in separation of diastereoisomers without use of a chiral column. Steroid Biochem. Mol. Biol. 2017, 172, 231239. https://doi.org/10.1016/j.jsbmb.2016.04.002.

    • Search Google Scholar
    • Export Citation
  • 18.

    Gurav, S.; Punde, R.; Farooqui, J.; Zainuddin, M.; Rajagopal, S.; Mullangi, R. Development and validation of a highly sensitive method for the determination of abiraterone in rat and human plasma by LC-MS/MS-ESI: application to a pharmacokinetic study. Biomed. Chromatogr. 2012, 26(6), 761768. https://doi.org/10.1002/bmc.1726.

    • Search Google Scholar
    • Export Citation
  • 19.

    Khedra, A.; Darwish, I.; Bamanea, F. Analysis of abiraterone stress degradation behavior using liquid chromatography coupled to ultraviolet detection and electrospray ionization mass spectrometry. J. Pharm. Biomed. Anal. 2013, 74, 7782. https://doi.org/10.1016/j.jpba.2012.09.024.

    • Search Google Scholar
    • Export Citation
  • 20.

    van Nuland, M.; Hillebrand, M. J. X.; Rosing, H.; Schellens, J. H. M.; Beijnen, J. H. Development and validation of an LC-MS/MS method for the simultaneous quantification of abiraterone, enzalutamide, and their major metabolites in human plasma. Ther. Drug Monit. 2017, 39(3), 243251. https://doi.org/10.1097/ftd.0000000000000387.

    • Search Google Scholar
    • Export Citation
  • 21.

    Goud, V. M.; Rani, B. S.; Sharma, J. Development and validation for estimation of abiraterone acetate in bulk and pharmaceutical dosage form by UPLC. Res. J. Pharm. Tech. 2019, 12(6), 30293032.

    • Search Google Scholar
    • Export Citation
  • 22.

    Gumustas, M.; Sanli, S.; Sanli, N.; Ozkan, S. A. Development and validation of a liquid chromatographic method for concurrent assay of weakly basic drug verapamil and amphoteric drug trandolapril in pharmaceutical formulations. J. Food Drug Anal. 2012, 20(3), 588596. https://doi.org/10.6227/jfda.2012200304.

    • Search Google Scholar
    • Export Citation
  • 23.

    Sanli, N.; Sanli, S.; Sızır, U.; Gumustas, M.; Ozkan, S. A. Determination of pKa values of cefdinir and cefixime by LC and spectrophotometric methods and their analysis in pharmaceutical dosage forms. Chromatographia 2011, 73(11), 11711176. https://doi.org/10.1007/s10337-011-2013-7.

    • Search Google Scholar
    • Export Citation
  • 24.

    Sanli, S.; Sanli, N.; Gumustas, M.; Karadas, N.; Aboul-Enien, H. Y. Simultaneous estimation of ceftazidime and ceftizoxime in pharmaceutical formulations by HPLC method. Chromatographia 2011, 74(7), 549558. https://doi.org/10.1007/s10337-011-2116-1.

    • Search Google Scholar
    • Export Citation
  • 25.

    Rizzi, A.; Katz, E.; Eksteen, R.; Schoenmakers, P. J. Handbook of HPLC. (Ed.: Marcel Dekker): New York, 1998.

  • 26.

    Sanli, N.; Fonrodona, G.; Barbosa, J.; Ozkan, G.; Beltran, J. L. Modelling retention in liquid chromatography of polyphenolic acids: prediction of solvent composition and pH of the mobile phase. Anal. Chim. Acta 2005, 537(1), 5361. https://doi.org/10.1016/j.aca.2005.01.006.

    • Search Google Scholar
    • Export Citation
  • 27.

    Canals, I., II; Portal, J. A.; Bosch, E.; Roses, M. Retention of ionizable compounds on HPLC. 4. Mobile-phase pH measurement in methanol/water. Anal Chem. 2000, 72(8), 18021809. https://doi.org/10.1021/ac990943i.

    • Search Google Scholar
    • Export Citation
  • 28.

    Sherrod, P. NLREG – nonlinear regression analysis and curve fitting program, 2007. http://www.nlreg.com.

  • 29.

    Erdemgil, F.; Şanli, S.; Şanli, N.; Barbosa, J.; Guiteras, J.; Beltran, J.L. Determination of pKa values of some hydroxylated benzoic acids in methanol–water binary mixtures by LC methodology and potentiometry. Talanta 2007, 72(2), 489496.

    • Search Google Scholar
    • Export Citation
  • 30.

    Ermer, J.; Miller, J. H. Method Validation in Pharmaceutical Analysis in A Guide to Best Practice; VCH, Wiley Pub: Germany, 2005.

  • 31.

    Swartz, M. E.; Krull, I. S. Analytical Method Development and Validation; Marcel Dekker: New York, 1997.

  • 32.

    Barbosa, J.; Fonrodona, G.; Marqués, I.; Sanz-Nebot, V.; Toro, I. Solvent effects on protonation equilibria of peptides and quinolones by factor analysis applied to the correlation between dissociation constants and solvatochromic parameters in acetonitrile-water mixtures. Anal. Chim. Acta 1997a, 351(1–3), 397405.

    • Search Google Scholar
    • Export Citation
  • 33.

    Sanli, S.; Sanli, N. Determination of pKa values for some tyrosine kinase inhibitors using the reversed-phase liquid chromatography. J. Solution Chem. 2023, 52(6), 658670. https://doi.org/10.1007/s10953-023-01264-5.

    • Search Google Scholar
    • Export Citation
  • 34.

    Yasuda, M. Dissociation constants of some carboxylic acids in mixed aqueous solvents. J. Bull. Chem. Soc. Jpn. 1959, 32(5), 429432.

  • 35.

    Shedlovsky, T. Electrolytes; Peasce, B., Ed. Pergamon Press: New York, 1962.

  • 36.

    Barbosa, J.; Toro, I.; Sanz-Nebot, V. Acid-base behaviour of tripeptides in solvents used in liquid chromatography. Correlation between pK values and solvatochromic parameters of acetonitrile-water mixtures. Anal. Chim. Acta 1997b, 347(3), 295304. https://doi.org/10.1016/S0003-2670(97)00163-3.

    • Search Google Scholar
    • Export Citation
  • 37.

    Sanli, N.; Sanli, S.; Özkan, G.; Denizli, A. Determination of pKa values of some sulfonamides by LC and LC-PDA methods in acetonitrile-water binary mixtures. J. Braz. Chem. Soc. 2010, 21, 19521960.

    • Search Google Scholar
    • Export Citation
  • 38.

    CPMP/ICH/281/95 Note for Guidance on Validation of Analytical Procedure: Methodology, ICH Topic Q2B Validation of Analytical Procedure: Methodology, 1996. Step 4.

    • Search Google Scholar
    • Export Citation
  • 1.

    Siegel, R.; Ma, J.; Zou, Z.; Jemal, A. Cancer statistics. Cancer J. Clin. 2014, 64(1), 929. https://doi.org/10.3322/caac.21208.

  • 2.

    Fizazi, K.; Scher, H. I.; Molina, A.; Logothetis, C.; Chi, K. N.; Jones, R.; Staffurth, J.; North, S.; Vogelzang, N.; Saad, F.; Mainwaring, P.; Harland, S.; Goodman, O.; Sternberg, C.; Li, J.; Kheoh, T.; Haqq, C.; Bono, J. Abiraterone acetate for treatment of metastatic castration-resistant prostate cancer: final overall survival analysis of the COU-AA-301 randomised, double-blind, placebo-controlled phase 3 study. Lancet Oncol. 2012, 13(10), 983992. https://doi.org/10.1016/s1470-2045(12)70379-0.

    • Search Google Scholar
    • Export Citation
  • 3.

    Rowlands, M. G.; Barrie, S. E.; Chan, F.; Houghton, J.; Jarman, M.; McCague, R.; Potter, G. A Esters of 3-pyridylacetic acid that combine potent inhibition of 17 alpha-hydroxylase/C17,20-lyase (cytochrome P45017 alpha) with resistance to esterase hydrolysis. J. Med. Chem. 1995, 38(21), 41914197. https://doi.org/10.1021/jm00021a008.

    • Search Google Scholar
    • Export Citation
  • 4.

    Hunt, N. J. Methanesulfonate salts of abiterone-3-esters and recovery of salts of Abiraterone one-3-esters from solution in methyl tetra-butyl ether, 2010, (United States). Pub. No. US 2010/0152437 A1.

    • Search Google Scholar
    • Export Citation
  • 5.

    Sonpavde, G.; Attard, G.; Bellmunt, J.; Mason, M. D.; Malavaud, B.; Tombal, B.; Sternberg, C. N. The role of abiraterone acetate in the management of prostate cancer: a critical analysis of the literature. Eur. Urol. 2011, 60(2), 270278. https://doi.org/10.1016/j.eururo.2011.04.032.

    • Search Google Scholar
    • Export Citation
  • 6.

    Acharya, M.; Bernard, A.; Gonzalez, M.; Jiao, J.; De Vries, R.; Tran, N. Open-label, phase I, pharmacokinetic studies of abiraterone acetate in healthy men. Cancer Chemother. Pharmacol. 2012, 69(6), 15831590. https://doi.org/10.1007/s00280-012-1865-3.

    • Search Google Scholar
    • Export Citation
  • 7.

    O’Donnell, A.; Judson, I.; Dowsett, M.; Raynaud, F.; Dearnaley, D.; Mason, M.; Harland, S.; Robbins, A.; Halbert, G.; Nutley, B.; Jarman, M. Hormonal impact of the 17alpha-hydroxylase/C(17,20)-lyase inhibitor abiraterone acetate (CB7630) in patients with prostate cancer. Br. J. Cancer 2004, 90(12), 23172325. https://doi.org/10.1038/sj.bjc.6601879.

    • Search Google Scholar
    • Export Citation
  • 8.

    Sarker, H.; Miksinski, S. United States food and drug administration, center for drug evaluation and research, chemistry review (S), application number: 202379, 2011. (United States Food and Drug Administration), Pub. No. 202379 Orig 1s000, NAD, 202379.

    • Search Google Scholar
    • Export Citation
  • 9.

    Pan, X.; Wang, H.; Li, C.; Zhang, J. Z. H; Ji, C. MolGpka: a Web server for Small molecule pK(a) prediction using a graph-convolutional neural Network. J. Chem. Inf. Model 2021, 61(7), 31593165. https://doi.org/10.1021/acs.jcim.1c00075.

    • Search Google Scholar
    • Export Citation
  • 10.

    http://www.chemicalize.org. (2023). (Accessed 26 April 2023).

  • 11.

    Arunkumar, S. G.; Radha, G. V. Analytical method development and validation for theestimation of Abiraterone acetate in pharmaceutical formulation by UV and RP-HPLC. Int. J. Pharm. Bio. Sci. 2016, 7(2), 5761.

    • Search Google Scholar
    • Export Citation
  • 12.

    Latha, P. G.; Yunus, S. B.; Sree, N. B.; Naz, J.; Sagar, M.; Shine, S. A review on analytical methods for estimation of abiraterone acetate. World J. Pharm. Med. Res. 2016, 2, 23.

    • Search Google Scholar
    • Export Citation
  • 13.

    Reddy, B. J. C.; Sarada, N. J. Development and validation of a novel RP-HPLC method for stability-indicating assay of Abiraterone acetate. J. Liquid Chromatogr. & Relat. Tech. 2016, 39(7), 354363. https://doi.org/10.1080/10826076.2016.1163500.

    • Search Google Scholar
    • Export Citation
  • 14.

    Gadhave, R. V.; Tamnar, B. A.; Bansode, S. A.; Choudhari, V. P. Stability indicating RP-HPLC-PDA method for determination of abıraterone acetate and characterization of its base catalyzed degradation product by LC-MS. Int. J. Pharm. Pharm. Sci. 2016, 8(2), 7681.

    • Search Google Scholar
    • Export Citation
  • 15.

    Kavitapu, D.; Maruthapillai, A.; Mahapatra, S.; Selvi, J. A. New stability indicating RP-HPLC method for the determination of Abiraterone acetate, its related substances and degradation products in bulk and dosage form. Mater. Today Proc. 2021, 34, 469478.

    • Search Google Scholar
    • Export Citation
  • 16.

    Sankar, P. R.; Eswarudu, M. M.; Siva, P.; Viswanath, A. Optimized and validated RP-HPLC method for quantification of abıraterone acetate (an antı- prostate cancer drug) in pharmaceutıcal dosage form. High Technol. Lett. 2021, 27(7), 21.

    • Search Google Scholar
    • Export Citation
  • 17.

    Alyamani, M.; Li, Z.; Upadhyay, S. K.; Anderson, D. J.; Auchus, R. J.; Sharifi, N. Development and validation of a novel LC-MS/MS method for simultaneous determination of abiraterone and its seven steroidal metabolites in human serum: innovation in separation of diastereoisomers without use of a chiral column. Steroid Biochem. Mol. Biol. 2017, 172, 231239. https://doi.org/10.1016/j.jsbmb.2016.04.002.

    • Search Google Scholar
    • Export Citation
  • 18.

    Gurav, S.; Punde, R.; Farooqui, J.; Zainuddin, M.; Rajagopal, S.; Mullangi, R. Development and validation of a highly sensitive method for the determination of abiraterone in rat and human plasma by LC-MS/MS-ESI: application to a pharmacokinetic study. Biomed. Chromatogr. 2012, 26(6), 761768. https://doi.org/10.1002/bmc.1726.

    • Search Google Scholar
    • Export Citation
  • 19.

    Khedra, A.; Darwish, I.; Bamanea, F. Analysis of abiraterone stress degradation behavior using liquid chromatography coupled to ultraviolet detection and electrospray ionization mass spectrometry. J. Pharm. Biomed. Anal. 2013, 74, 7782. https://doi.org/10.1016/j.jpba.2012.09.024.

    • Search Google Scholar
    • Export Citation
  • 20.

    van Nuland, M.; Hillebrand, M. J. X.; Rosing, H.; Schellens, J. H. M.; Beijnen, J. H. Development and validation of an LC-MS/MS method for the simultaneous quantification of abiraterone, enzalutamide, and their major metabolites in human plasma. Ther. Drug Monit. 2017, 39(3), 243251. https://doi.org/10.1097/ftd.0000000000000387.

    • Search Google Scholar
    • Export Citation
  • 21.

    Goud, V. M.; Rani, B. S.; Sharma, J. Development and validation for estimation of abiraterone acetate in bulk and pharmaceutical dosage form by UPLC. Res. J. Pharm. Tech. 2019, 12(6), 30293032.

    • Search Google Scholar
    • Export Citation
  • 22.

    Gumustas, M.; Sanli, S.; Sanli, N.; Ozkan, S. A. Development and validation of a liquid chromatographic method for concurrent assay of weakly basic drug verapamil and amphoteric drug trandolapril in pharmaceutical formulations. J. Food Drug Anal. 2012, 20(3), 588596. https://doi.org/10.6227/jfda.2012200304.

    • Search Google Scholar
    • Export Citation
  • 23.

    Sanli, N.; Sanli, S.; Sızır, U.; Gumustas, M.; Ozkan, S. A. Determination of pKa values of cefdinir and cefixime by LC and spectrophotometric methods and their analysis in pharmaceutical dosage forms. Chromatographia 2011, 73(11), 11711176. https://doi.org/10.1007/s10337-011-2013-7.

    • Search Google Scholar
    • Export Citation
  • 24.

    Sanli, S.; Sanli, N.; Gumustas, M.; Karadas, N.; Aboul-Enien, H. Y. Simultaneous estimation of ceftazidime and ceftizoxime in pharmaceutical formulations by HPLC method. Chromatographia 2011, 74(7), 549558. https://doi.org/10.1007/s10337-011-2116-1.

    • Search Google Scholar
    • Export Citation
  • 25.

    Rizzi, A.; Katz, E.; Eksteen, R.; Schoenmakers, P. J. Handbook of HPLC. (Ed.: Marcel Dekker): New York, 1998.

  • 26.

    Sanli, N.; Fonrodona, G.; Barbosa, J.; Ozkan, G.; Beltran, J. L. Modelling retention in liquid chromatography of polyphenolic acids: prediction of solvent composition and pH of the mobile phase. Anal. Chim. Acta 2005, 537(1), 5361. https://doi.org/10.1016/j.aca.2005.01.006.

    • Search Google Scholar
    • Export Citation
  • 27.

    Canals, I., II; Portal, J. A.; Bosch, E.; Roses, M. Retention of ionizable compounds on HPLC. 4. Mobile-phase pH measurement in methanol/water. Anal Chem. 2000, 72(8), 18021809. https://doi.org/10.1021/ac990943i.

    • Search Google Scholar
    • Export Citation
  • 28.

    Sherrod, P. NLREG – nonlinear regression analysis and curve fitting program, 2007. http://www.nlreg.com.

  • 29.

    Erdemgil, F.; Şanli, S.; Şanli, N.; Barbosa, J.; Guiteras, J.; Beltran, J.L. Determination of pKa values of some hydroxylated benzoic acids in methanol–water binary mixtures by LC methodology and potentiometry. Talanta 2007, 72(2), 489496.

    • Search Google Scholar
    • Export Citation
  • 30.

    Ermer, J.; Miller, J. H. Method Validation in Pharmaceutical Analysis in A Guide to Best Practice; VCH, Wiley Pub: Germany, 2005.

  • 31.

    Swartz, M. E.; Krull, I. S. Analytical Method Development and Validation; Marcel Dekker: New York, 1997.

  • 32.

    Barbosa, J.; Fonrodona, G.; Marqués, I.; Sanz-Nebot, V.; Toro, I. Solvent effects on protonation equilibria of peptides and quinolones by factor analysis applied to the correlation between dissociation constants and solvatochromic parameters in acetonitrile-water mixtures. Anal. Chim. Acta 1997a, 351(1–3), 397405.

    • Search Google Scholar
    • Export Citation
  • 33.

    Sanli, S.; Sanli, N. Determination of pKa values for some tyrosine kinase inhibitors using the reversed-phase liquid chromatography. J. Solution Chem. 2023, 52(6), 658670. https://doi.org/10.1007/s10953-023-01264-5.

    • Search Google Scholar
    • Export Citation
  • 34.

    Yasuda, M. Dissociation constants of some carboxylic acids in mixed aqueous solvents. J. Bull. Chem. Soc. Jpn. 1959, 32(5), 429432.

  • 35.

    Shedlovsky, T. Electrolytes; Peasce, B., Ed. Pergamon Press: New York, 1962.

  • 36.

    Barbosa, J.; Toro, I.; Sanz-Nebot, V. Acid-base behaviour of tripeptides in solvents used in liquid chromatography. Correlation between pK values and solvatochromic parameters of acetonitrile-water mixtures. Anal. Chim. Acta 1997b, 347(3), 295304. https://doi.org/10.1016/S0003-2670(97)00163-3.

    • Search Google Scholar
    • Export Citation
  • 37.

    Sanli, N.; Sanli, S.; Özkan, G.; Denizli, A. Determination of pKa values of some sulfonamides by LC and LC-PDA methods in acetonitrile-water binary mixtures. J. Braz. Chem. Soc. 2010, 21, 19521960.

    • Search Google Scholar
    • Export Citation
  • 38.

    CPMP/ICH/281/95 Note for Guidance on Validation of Analytical Procedure: Methodology, ICH Topic Q2B Validation of Analytical Procedure: Methodology, 1996. Step 4.

    • Search Google Scholar
    • Export Citation
  • Collapse
  • Expand

Senior editors

Editor(s)-in-Chief: Sajewicz, Mieczyslaw, University of Silesia, Katowice, Poland

Editors(s)

  • Danica Agbaba, University of Belgrade, Belgrade, Serbia (1953-2024)
  • Łukasz Komsta, Medical University of Lublin, Lublin, Poland
  • Ivana Stanimirova-Daszykowska, University of Silesia, Katowice, Poland
  • Monika Waksmundzka-Hajnos, Medical University of Lublin, Lublin, Poland

Editorial Board

  • Ravi Bhushan, The Indian Institute of Technology, Roorkee, India
  • Jacek Bojarski, Jagiellonian University, Kraków, Poland
  • Bezhan Chankvetadze, State University of Tbilisi, Tbilisi, Georgia
  • Michał Daszykowski, University of Silesia, Katowice, Poland
  • Tadeusz H. Dzido, Medical University of Lublin, Lublin, Poland
  • Attila Felinger, University of Pécs, Pécs, Hungary
  • Kazimierz Glowniak, Medical University of Lublin, Lublin, Poland
  • Bronisław Glód, Siedlce University of Natural Sciences and Humanities, Siedlce, Poland
  • Anna Gumieniczek, Medical University of Lublin, Lublin, Poland
  • Urszula Hubicka, Jagiellonian University, Kraków, Poland
  • Krzysztof Kaczmarski, Rzeszow University of Technology, Rzeszów, Poland
  • Huba Kalász, Semmelweis University, Budapest, Hungary
  • Katarina Karljiković Rajić, University of Belgrade, Belgrade, Serbia
  • Imre Klebovich, Semmelweis University, Budapest, Hungary
  • Angelika Koch, Private Pharmacy, Hamburg, Germany
  • Piotr Kus, Univerity of Silesia, Katowice, Poland
  • Debby Mangelings, Free University of Brussels, Brussels, Belgium
  • Emil Mincsovics, Corvinus University of Budapest, Budapest, Hungary
  • Ágnes M. Móricz, Centre for Agricultural Research, Budapest, Hungary
  • Gertrud Morlock, Giessen University, Giessen, Germany
  • Anna Petruczynik, Medical University of Lublin, Lublin, Poland
  • Robert Skibiński, Medical University of Lublin, Lublin, Poland
  • Bernd Spangenberg, Offenburg University of Applied Sciences, Germany
  • Tomasz Tuzimski, Medical University of Lublin, Lublin, Poland
  • Adam Voelkel, Poznań University of Technology, Poznań, Poland
  • Beata Walczak, University of Silesia, Katowice, Poland
  • Wiesław Wasiak, Adam Mickiewicz University, Poznań, Poland
  • Igor G. Zenkevich, St. Petersburg State University, St. Petersburg, Russian Federation

 

SAJEWICZ, MIECZYSLAW
E-mail:mieczyslaw.sajewicz@us.edu.pl

Indexing and Abstracting Services:

  • Science Citation Index
  • Sci Search
  • Research Alert
  • Chemistry Citation Index and Current Content/Physical
  • Chemical and Earth Sciences
  • SCOPUS
  • GoogleScholar
  • Index Copernicus
  • CABI
  • CABELLS Journalytics

2023  
Web of Science  
Journal Impact Factor 1.7
Rank by Impact Factor Q3 (Chemistry, Analytical)
Journal Citation Indicator 0.43
Scopus  
CiteScore 4.0
CiteScore rank Q2 (General Chemistry)
SNIP 0.706
Scimago  
SJR index 0.344
SJR Q rank Q3

Acta Chromatographica
Publication Model Online only
Gold Open Access
Submission Fee none
Article Processing Charge 400 EUR/article
Effective from  1st Feb 2025:
700 EUR/article
Regional discounts on country of the funding agency World Bank Lower-middle-income economies: 50%
World Bank Low-income economies: 100%
Further Discounts Editorial Board / Advisory Board members: 50%
Corresponding authors, affiliated to an EISZ member institution subscribing to the journal package of Akadémiai Kiadó: 100%
Subscription Information Gold Open Access
Purchase per Title  

Acta Chromatographica
Language English
Size A4
Year of
Foundation
1988
Volumes
per Year
1
Issues
per Year
4
Founder Institute of Chemistry, University of Silesia
Founder's
Address
PL-40-007 Katowice, Poland, Bankowa 12
Publisher Akadémiai Kiadó
Publisher's
Address
H-1117 Budapest, Hungary 1516 Budapest, PO Box 245.
Responsible
Publisher
Chief Executive Officer, Akadémiai Kiadó
ISSN 2083-5736 (Online)

Monthly Content Usage

Abstract Views Full Text Views PDF Downloads
Jul 2024 0 71 13
Aug 2024 0 164 28
Sep 2024 0 165 21
Oct 2024 0 313 19
Nov 2024 0 314 40
Dec 2024 0 297 31
Jan 2025 0 93 26