Int J App Pharm, Vol 16, Issue 1, 2024, 88-93Original Article

AN LC-ESI-MS/MS METHOD DEVELOPMENT AND VALIDATION FOR THE QUANTIFICATION OF INFIGRATINIB IN BIOLOGICAL MATRICES

PHANI KUMAR SUNKARA, SREEDHARA CHAGANTY, K. RAMAKRISHNA*

Department of Chemistry, Institute of Science, GITAM (Deemed to be University), Visakhapatnam-530045, Andhra Pradesh, India
*Corresponding author: K. Ramakrishna; *Email: karipeddirk@gmail.com

Received: 24 Sep 2023, Revised and Accepted: 17 Nov 2023


ABSTRACT

Objective: The study was aimed to develop a precise and simple liquid chromatographic electrospray ionization tandem mass spectrometric (LC--MSMS) technique is essential for the quantification of Infigratinib in biological matrices.

Methods: Chromatographic resolution was attained with PhenominexC18 (50 mm×2.6 mm, 3 µm) stationary column and a mobile solvent composition of 0.1% HCOOH, methyl alcohol and acetonitrile in the proportion of 10:10:80. Chromatograms were resolved by an isocratic separation with a flowing rate of 0.50 ml/min at 40 °C.

Results: Quantitation was executed by monitoring the transitions of m/z. 560.19/189.13 for Infigratinib and 494.5→394.5 for Imatinib internal standard in multiple reaction monitoring. The standard curve regression line was y = 0.0016x+0.0062 and the correction coefficient (r2) was 0.9994. The % CV outcomes for matrix effect at Lower-QC and Higher-QC were 4.95% and 3.61% respectively. The percentage average recoveries for Infigratinib in Higher-QC (900ng/ml), MQC (600ng/ml) and Lower-QC (3ng/ml) were 93.27%, 94.69% and 97.24% respectively. The intra and interday precisions of analytical procedure was estimated by assessing the %CV outcomes and were in between 1.88 to 5.93% for the QC samples.

Conclusion: The developed procedure can be useful for the assessment of Infigratinib in biological matrices in quality control, forensic and bioavailability studies.

Keywords: Infigratinib, Cholangiocarcinoma, LC-MS/MS, Validation, Linearity


INTRODUCTION

Infigratinib, also known by its chemical name 3-(2, 6-dichloro-3,5-dimethoxyphenyl)-1-[6-[4-(4-ethylpiperazin-1-yl) anilino] pyrimidin-4-yl]-1-methylurea, is a cancer drug. The chemical formula is having an empirical formula of C26H31Cl2N7O3. Pan-fibroblasts growth factor receptors (FGFR) kinase inhibition may be achieved with the use of infigratinib. Infigratinib inhibits the FGFR path, which is mutated in malignancies like cholangiocarcinoma. In doing so, it is able to prevent the development of tumors. Cholangiocarcinoma is the most frequent kind of primary cancer that affects the biliary system, and it is also the second most common kind of primary cancers that affects liver. Because it is an ATP-competitive inhibitors of all 4 different FGFR receptors subtype, infitratinib is considered to be a pan-FGFR inhibitor [1-5].

FGFRs are tyrosine kinase receptors that regulate proliferation of cells, migration, differentiation, angiogenesis and survival. After attaching to external signals, such as FGFRs merges to phosphorylate downstream molecules and activate the Ras-MAPK pathway. FGFR signaling pathway disruptions cause uncontrolled expansion and cell proliferation, including malignant cells, in several malignancies. FGFR receptor fusions, amplifications, and mutations, are linked to urothelial, prostate, ovarian, liver and breast, cancers. Recent investigations suggest that up to 45% of intrahepatic cholangiocarcinoma patients have gene rearrangements that produced FGFR2 fusion proteins. Tumors with FGFR mutations may encourage malignant cell growth and survival via constitutive signaling. In cancer cell line with stimulating FGFRs amplification, fusions, or mutations, infigratinib blocks FGFR signalling and suppresses cell propagation. It is reversible, non competitive inhibitors of all 4 subtypes of FGFR. Infigratinib binds best to FGFR1, FGFR2, and FGFR3 among the four subtypes [6, 7]. Literature survey on Infigratinib drug reveals that two quantification approaches for the assessment of Infigratinib in sample plasma were reported. So, there is need of an LC-ESI-MSMS analytical procedure for estimation of Infigratinib in biological matrix.

Fig. 1: Infigratinib chemical structure

MATERIALS AND METHODS

Chemicals and reagents

Infigratinib and Imatinib (IS) were gift sample from Solu Clinipharm Pvt. Ltd, Maharastra, India. LC grade CAN (acetonitrile) and methyl alcohol were acquired from J. T. Bakers, Hyderabad. Water utilized for total research work from water purification (Milli-Q) systems. Formic acid of analytical grade was obtained from Merk Pvt. Ltd., Mumbai, India.

Equipment

The Applied Biosystem Sciex-API4000 Tandem mass spectrometer was combined with the Shimadzu LC20ADvp (Shimadzu, Tkyo Japan) high-performance liquid chromatography (HPLC) system that included an auto-sampler. Japan's Shimadzu LC20AD was the device module that served as for the solvent method of delivery. The Analyst program, which was developed by Applied Biosystems and comes in version 01.04.02, was used to combine all of the chromatographic data.

Preparation of quality and calibration standard solutions

A 100µg/ml of Infigratinib and Imatinib (IS) stock solutions executed by solubilizing in exact quantity of mobile phase. Quality and calibration controls were processed with plasma blank samples from these standards stock Infigratinib solution. Eight calibration levels of different concentrations were processed by spike method to the plasma blank with Infigratinib standard solutions to made the concentration levels of 1, 5, 35, 150, 350, 600, 900, and 1200ng/ml. Low-quality (Lower-QC), medium quality (Medium-QC) and higher quality (Higher-QC) levels were executed by spike method to plasma blanks with Infigratinib to made solutions of 3, 6 and 9 ng/ml respectively. From Imatinib stock solution of 350 ng/ml its working solution was processed with mobile phase. Processed solutions were kept at −20 °C till time estimation.

Chromatographic conditions

Chromatographic resolution was attained with PhenominexC18 (50 mm×2.6 mm, 3 µm) stationary column and a mobile solvent composition of 0.1% HCOOH, methyl alcohol and acetonitrile in the proportion of 10:10:80. Chromatograms were resolved by an isocratic separation with a flowing rates of 0.50 ml/min at 40 °C. Auto-sampler and column oven temperature were adjusted to 5 °C and 4 °C correspondingly.

Mass spectroscopic settings

Mass settings were set to source temperatures 450 °C; nebulizer gas 20psi (N2); heater gas 30 psi (N2); curtain gas 25psi (Nitrogen); entrance potentials 15V; CAD gas 8(N2); declustering potential 80V source flow rate 0.5 ml/min with no splitting and 15V collision energy (CE) for both Infigratinib and Imatinib, collision cell exit potentials-15V and dwell time 200 ms for the Infigratinib and collision cells exit potentials-14 V for Imatinib. A turbo-ion-spray interface ran in positive (+) mode of ionization was utilized for the quantitative detection. Parent and product ion detection was employed in MRM (multiple reaction monitoring) manner, with transitions pairs of m/z-560.19/189.13 for Infigratinib and 494.5→394.5 for Imatinib internal standard.

Protocol for sample preparation

Each spiked plasma samples of 50μl mixed with 250μl methyl alcohol having 0.1% of HCOOH to precipitate the proteins present in the mixture. The resultant mixture was subjected for the vortex mixing for 10 min. Then these sample solutions were centrifuge for 20 min at 4.0 °C. Then 150μl of supernatant liquid was relocated to polypropylene tubes, from which an aliquot 5μl of samples were infused into LC-MSMS system. The final concentration of these Imatinib should be 350ng/ml in each sample preparation [14, 15].

Method validation

The accuracy, lower limit of quantification (LLOQ), precision, recovery, linearity, selectivity, and matrix effects of the quantitative determination of infigratinib in plasma were tested in accordance with the Guidelines for bio-analytical technique validation in pharmaceutical development. This was done in order to validate the quantitative determination of infigratinib in plasma [16-18].

RESULTS AND DISCUSSION

Method validation

The chromatographs achieved during the validation process were satisfactory and resultant blank, blank+IS, and LLOQC chromatographs were given in (fig. 2 and fig. 3).

Specificity

In order to obtain a clean separation of infigratinib from endogenous impurities and other impurities found in plasma, a solution with a concentration of 1.0 ng/ml was injected into the chromatographic system under conditions that had been tuned for HPLC. Assessing the plasma interference allowed for an estimation of the method's level of specificity [19, 20]. It is recommended that the peak area of the interference peak be less than 20 percent of the peak area of the LLOQ and less than 5 percent of the mean peak area of the IS. There should not be a deviation of more than twenty percent between the nominal concentration (NC) and the LLOQ concentration.

Fig. 2: Chromatographs of plasma blank (a), plasma blank with Imatinib (b)

Fig. 3: Representative chromatographs of plasma spiked with Infigratinib at LLOQC and Imatinib

Linearity

In order to determine whether or not the approach was linear, a calibration curve was constructed by comparing spiking results to peak area proportions of Infigratinib to IS. Standard values for calibration that range from 1 to 1200 ng/ml. All three calibration curves were linear across the operating range, and a calibration at eight points was employed for quantitative analysis using linear regression (fig. 4) [21, 22]. The standard curve regression equation was found to be y = 0.0016x+0.0062 and with correction coefficient (r2) of 0.9994. The linear curve findings were shown in the table 1.

Table 1: Linearity standard solutions for infigratinib

LS-ID Concentration (ng/ml) Average response IS response Analyte/IS response
LS-1 1 643 402154 0.001599
LS-2 5 3215 402741 0.007983
LS-3 35 26505 402561 0.065841
LS-4 150 96957 402285 0.241016
LS-5 350 237051 402402 0.58909
LS-6 600 387884 402247 0.964293
LS-7 900 561747 402645 1.395142
LS-8 1200 771648 402393 1.917648

CS: Calibration standard.

Fig. 4: Calibration curve of Infigratinib

Intra and interday precisions

Both intra and inter days accuracy and precisions were examined. By randomly injecting QC standards (3, 600, and 900ng/ml) and LLOQ (1.0ng/ml) in five duplicates each day, intraday accuracy and precision were examined. The examination of each quality control standard (3, 600, and 900 ng/ml) and LLOQC standard (1.0ng/ml) once on each of five distinct days was used to estimate the interday accuracy and precision. By examining the % CV data, it was possible to determine the analytical method's intraday and interday accuracy. The obtained results for the QC samples ranged from 1.88 to 5.93%. The outcomes were all within the 15% precision range. The outcomes were compiled in table 2.

Recovery

Recoveries of analytes were executed by equating the area of peak for Infigratinib (extracted samples) with response peaks of control Infigratinib level. The % average recovery for Infigratinib in High-QC (900 ng/ml), MQC (600 ng/ml) and Low-QC(3 ng/ml) were 93.27%, 94.69% and 97.24% respectively (table 3).

Table 2: Infigratinib intra and inter day precision data

QC Infigratinib (µg/ml)
Intra batch Average SD %CV % Bias
LLOQC 0.96 0.02 2.07 -3.44
Low-QC 3.138 0.12 3.82 4.61
Median-QC 586.74 34.82 5.93 -2.21
High-QC 953.60 44.38 4.65 5.96
LLOQ 1.036 0.024 2.31 3.60
Low-QC 2.89 0.129 4.46 -3.67
Median-QC 614.65 20.94 3.41 2.441
High-QC 903.14 39.27 4.35 0.35
LLOQ 0.96 0.018 1.88 -4.44
LLOQ 3.08 0.13 4.22 2.67
Low-QC 579.45 33.89 5.85 -3.425
Median-QC 959.12 42.25 4.41 6.56
Inter batch Average SD %CV % Bias
LLOQ 1.036 0.035 3.37 3.604
Low-QC 2.89 0.098 3.397 -3.66
Median-QC 614.65 25.27 4.117 2.46
High-QC 933.14 42.67 4.577 3.68

Fig. 5: Infigratinib chromatograms for A) Lower-QC B) Medium-QC and C) Higher-QC

Table 3: Analytes recovery

Concentration levels X Y % Recovered % Mean recoveries % RSD
LQC 1929 1875 97.24 95.07 1.73
MQC 385800 365314 94.69
HQC 578700 539753 93.27
IS 402345 395867 98.39

X, recoveries of mean unextracted sample; Y, recoveries of mean extracted sample.

Matrix effect

Following the extraction of six blank plasma samples from six distinct lots in triplicate, the samples were spiked at Low-QC and High-QC levels, and the results were ultimately compared with the same amounts in the alternative infusions. Low-QC samples had a coefficient of variation (CV) of 4.95%, while High-QC samples had a CV of 3.61%. Both of these values fall within the acceptability requirements of 15%. The findings of the matrix factor analysis may be found summarized in table 4.

Table 4: Infigratinib matrix effect

S. No. LQC HQC
Peak area in absence of matrix Peak area in presence of matrix Matrix
factor
Peak area in absence of matrix Peak area in presence of matrix Matrix factor
1. 1892 1788 94.51 578524 592293 102.38
2. 1881 1748 92.94 577952 552927 95.67
3. 1984 2008 101.24 578921 549917 94.99
4. 1991 1957 98.34 578417 545910 94.38
5. 1923 1787 92.96 579014 589552 101.82
6. 1958 2049 104.67 579239 561167 96.88
Mean 97.44 97.69
±SD 4.82 3.52
% CV 4.95 3.61

Stability studies

The requirements for stability were as follows: post-preparative (24 h at 10 degrees Celsius), short-term (19 h at 25 degrees Celsius), long-term (36 d at-70 degrees Celsius), stock solution stability, and freeze/thaw (3 cycles) stability [23]. The findings of the stability tests showed that there was no substantial degradation of infigratinib that happened during the chromatographic method, extraction, or sample storage of infigratinib plasma samples under various storage settings. This was proved by the fact that there was no significant change in the concentration of infigratinib. The statistics about stability were presented in table 5.

Table 5: The stability data of infigratinib in human plasma under different storage conditions

Storage condition LQC MQC HQC
Accuracy (Mean%) Precision
(RSD%)
Accuracy (Mean%) Precision (RSD%) Accuracy (Mean%) Precision (RSD%)
Post preparative (24h at 10 °C) 101.92 4.38 97.69 2.98 92.97 3.28
Short-term (19h at 25 °C) 96.37 5.39 105.97 5.31 93.41 4.58
Long-term (36 d at-70 °C) 95.28 3.28 96.39 4.92 103.25 5.09
Three freeze/thaw (3cycles) 98.17 4.67 95.16 3.67 102.49 2.88
Stock solution (20h at 25 °C) 103.59 5.28 103.77 5.07 96.34 4.77
Stock solution (17day at 2 to 8 °C) 102.36 3.97 96.24 4.08 94.66 5.34

Carry over effect

The carry-overs have been evaluated by calculating the concentration of a blank sample by following the highest calibration standard concentration in six repetitions. The peak response of the blank sample produced after the HQC should not be more than twenty percent higher than the drug response of the LLOQ, nor should it be more than five percent higher than the response of Imatinib. The carrying over of funds was deemed appropriate.

CONCLUSION

A precise and simple liquid chromatographic electrospray ionization tandem mass spectrometric (LC--MS/MS) technique was developed for the quantification of Infigratinib in biological matrices. In the range of 1.0 to 1200 ng/ml, the drug content is directly related to the peak reaction. Chromatographic separation was done using a PhenominexC18 (50 mm×2.6 mm, 3 µm) column and a mobile solvent made of 0.1% HCOOH, methyl alcohol, and acetonitrile in the ratio of 10:10:80. Chromatograms were separated using an isocratic elution at 40 0C and a flow rate of 0.50 ml/min. Compared to other methods for Infigratinib, the total time for research looks good. The regression equation for the linear curve was found to be y = 0.0016x+0.0062, and the correction coefficient (r2) was 0.9994. At the Low-QC and High-QC levels, the % CV results for the matrix effect were 4.95 and 3.61, respectively. Infigratinib was recovered at 93.27%, 94.69%, and 97.24% in High-QC (900 ng/ml), MQC (600 ng/ml), and Low-QC (3 ng/ml), respectively. By looking at the %CV numbers, it was possible to fig. out how accurate the analysis method was during the day and between days. For the QC samples, the numbers found ranged from 1.88 to 5.93%.

FUNDING

Nil

AUTHORS CONTRIBUTIONS

All the authors have contributed equally.

CONFLICT OF INTERESTS

Declared none

REFERENCES

  1. Botrus G, Raman P, Oliver T, Bekaii Saab T. Infigratinib (BGJ398): an investigational agent for the treatment of FGFR-altered intrahepatic cholangiocarcinoma. Expert Opin Investig Drugs. 2021;30(4):309-16. doi: 10.1080/13543784.2021.1864320, PMID 33307867.

  2. Blechacz B. Cholangiocarcinoma: current knowledge and new developments. Gut Liver. 2017;11(1):13-26. doi: 10.5009/gnl15568, PMID 27928095.

  3. Lima NC, Atkinson E, Bunney TD, Katan M, Huang PH. Targeting the Src pathway enhances the efficacy of selective FGFR inhibitors in urothelial cancers with FGFR3 alterations. Int J Mol Sci. 2020;21(9):3214. doi: 10.3390/ijms21093214, PMID 32370101.

  4. Kang C. Correction to: infigratinib: first approval. Drugs Drugs. 2022;82(1):93. doi: 10.1007/s40265-021-01652-5, PMID 34817843.

  5. Roskoski R Jr. Properties of FDA-approved small molecule protein kinase inhibitors: a 2022 update. Pharmacol Res. 2022;175:106037. doi: 10.1016/j.phrs.2021.106037.

  6. Attwa MW, AlRabiah H, Mostafa GAE, Kadi AA. Development of an LC-MS/MS method for quantification of sapitinib in human liver microsomes: in silico and in vitro metabolic stability evaluation. Molecules. 2023;28(5):2322. doi: 10.3390/molecules28052322, PMID 36903565.

  7. Xu Xuegu, Chen C, Liu YN, Meng X, Cai Jp, Xu RA. Establishment and validation of a UPLC-MS/MS bioassay for the quantification of infigratinib in rat plasma. Arab J Chem. 2022;15(7):103893. doi: 10.1016/j.arabjc.2022.103893.

  8. Allard M, Khoudour N, Rousseau B, Joly C, Costentin C, Blanchet B. Simultaneous analysis of regorafenib and sorafenib and three of their metabolites in human plasma using LC-MS/MS. J Pharm Biomed Anal. 2017;142:42-8. doi: 10.1016/j.jpba.2017.04.053, PMID 28494338.

  9. Moola KS, Challa BSR, Bannoth CK. Quantification of tolvaptan in rabbit plasma by LC-MS/MS: application to a pharmacokinetic study. J Pharm Anal. 2015;5(6):371-7. doi: 10.1016/j.jpha.2014.09.001, PMID 29403951.

  10. Dadhaniya T, Chaudhary K, Mehta P. Development of LC-MS/MS method for determination of iloperidone in rabbit plasma: application to a pharmacokinetic study. Int J Pharm Pharm Sci. 2013;7(4):294-7.

  11. Moola KS, Challa BSR, Chandrasekhar KB. Bioanalytical method development and validation of eszopiclone in rabbit plasma by HPLC-MS/MS and its application to pharmacokinetic study. Int J Biol Pharm Res. 2014;5(9):719-27.

  12. Pilla N, Chandanam S, Sreenivas Rao T, Veera Reddy S. Development and validation of liquid chromatography-tandem mass pectrometry for determination of olanzapine in rabbit plasma. DHR Int J Pharm Sci. 2014;5(1):151-9.

  13. Chandrasekar MJN, Chandrasekar AJ, Krishnaraj K, Muralidharan S, Rajan S, Suresh B. Liquid chromatography-mass spectrometry determination of Cetrizine hydrochloride in rabbit plasma. Asian J Chem. 2009;21(8):5821-8.

  14. Kaza M, Karazniewicz Lada M, Kosicka K, Siemiatkowska A, Rudzki PJ. Bioanalytical method validation: new FDA guidance vs. EMA guideline. Better or worse? J Pharm Biomed Anal. 2019;165:381-5. doi: 10.1016/j.jpba.2018.12.030, PMID 30590335.

  15. Smith G. European medicines agency guideline on bioanalytical method validation: what more is there to say? Bioanalysis. 2012;4(8):865-8. doi: 10.4155/bio.12.44, PMID 22533559.

  16. Li X, Shi X, Qin X, Yu L, Zhou Y, Rao C. Interlaboratory method validation of imaged capillary isoelectric focusing methodology for analysis of recombinant human erythropoietin. Anal Methods. 2020;12(30):3836-43. doi: 10.1039/d0ay00823k, PMID 32678383.

  17. Del Mar Ramírez Fernández M, Wille SM, Samyn N. Quantitative method validation for the analysis of 27 antidepressants and metabolites in plasma with ultraperformance liquid chromatography-tandem mass spectrometry. Ther Drug Monit. 2012;34(1):11-24. doi: 10.1097/FTD.0b013e31823bf0fd, PMID 22210095.

  18. Wozniakiewicz M, Wietecha Posłuszny R, Moos A, Wieczorek M, Knihnicki P, Koscielniak P. Development of microextraction by packed sorbent for toxicological analysis of tricyclic antidepressant drugs in human oral fluid. J Chromatogr A. 2014;1337:9-16. doi: 10.1016/j.chroma.2014.02.037, PMID 24636563.

  19. Chambers EE, Woodcock MJ, Wheaton JP, Pekol TM, Diehl DM. Systematic development of an UPLC-MS/MS method for the determination of tricyclic antidepressants in human urine. J Pharm Biomed Anal. 2014;88:660-5. doi: 10.1016/j.jpba.2013.09.001, PMID 24239905.

  20. Patel DS, Sharma N, Patel MC, Patel BN, Shrivastav PS, Sanyal M. Development and validation of a selective and sensitive LC-MS/MS method for determination of cycloserine in human plasma: application to bioequivalence study. J Chromatogr B Analyt Technol Biomed Life Sci. 2011;879(23):2265-73. doi: 10.1016/j.jchromb.2011.06.011, PMID 21727043.

  21. Yadav M, Shrivastav PS. Incurred sample reanalysis (ISR): a decisive tool in bioanalytical research. Bioanalysis. 2011;3(9):1007-24. doi: 10.4155/bio.11.76, PMID 21545349.

  22. Lolla S, Gubbiyappa KS, Cheruku S, Bhikshapathi DVRN. Validation of an LC-MS/MS method for quantitation of fostemsavir in plasma. J Pharmacol Toxicol Methods. 2023;120:107254. doi: 10.1016/j.vascn.2023.107254, PMID 36863666.

  23. Sandhya P, Shankar CH, Bhikshapathi DVRN, Mamatha P. An LC–MS/MS quantification method development and validation for the dabrafenib in biological matrices. Sai Uday Kiran G J Appl Pharm Sci. 2023;13(1):180-6.