An Overview of Analytical and Bioanalytical Techniques for the determination of Tyrosine Kinase Inhibitors
M. Gowthami, Rajesh R*
Department of Pharmaceutical Analysis, Acharya & BM Reddy College of Pharmacy,
Soladevanahalli, Hessarghatta Main Road, Bengaluru, Karnataka, India.
*Corresponding Author E-mail: rajeshr@acharya.ac.in
ABSTRACT:
Tyrosine KIs have become a targeted drug therapy for different malignancies. Over the past ten years, kinase inhibitors, including monoclonal antibodies and small-molecule TKIs targeted at kinases, have become a significant class of chemotherapeutic agents. A number of studies documenting the design, usage and validation of bioanalytical methods for TCKIs have been published as a result of the rising need for bioanalytical approaches to both qualitatively and quantitatively study such compounds. Many biomatrices, including blood, cerebrospinal fluid, urine, tissue, and even liver microsomes, can be used to quantify TCKIs. The majority of papers explain the technological framework of analytical methods that can do this. In recent times, there has also been an increase in interest in the discovery of intermediates and biotransformation mechanisms for novel TCKIs. We give a summary of TCKI bioanalytical techniques.
KEYWORDS: Anticancer drugs, Bioanalytical techniques, Tyrosine Kinase Inhibitors, HPLC (High-Performance/ Pressure Liquid Chromatography), Liquid Chromatography-Mass Spectrometry (LC-MS).
INTRODUCTION:
One of the most important medical discoveries in recent decades seems to have been chemotherapy for cancer. Unfortunately, because of the narrow therapeutic index of the drugs used in this treatment, the outcomes are frequently only temporary and unreliable. In contrast, more recent tailored medicines focus on chemicals and signalling pathways that are unique to cancer, causing fewer non-specific side effects. The fact that tyrosine kinases are involved in the modification of growth factor signalling makes them a particularly crucial target1. An enzyme called protein kinase phosphorylates other proteins. The end-phosphate unit of ATP is directly joined to threonine, serine, or tyrosine amino acids during kinase phosphorylation2,3. It is widely accepted that kinase dysregulation increases oncogenic potential and dysfunction can be brought on by point mutations, overexpression, fusion, translocation, and dysregulation of upstream pathways, among many other methods4,5,6,7. TCKIs signalling pathways are intricate, including a growing number of biochemical processes and molecular regulators in intricate signalling systems.
TCKI has an effect on a few intermediaries, including phosphoinositide 3-kinase, mitogen-activated protein kinases (MAPK) and calcium signalling. RTKs primarily activate the Pi3K/AKT/mTOR, phospholipase C (PLC) or CaMK-PKC pathways, RAS/MAPK, and Janus kinase (JAK) which are downstream effectors of various mechanisms during cancer pathogenesis (Figure.1)8. In addition to afatinib, zanubrutinib, osimertinib, neratinib, ibrutinib, acalabrutinib and dacomitinib, which have all received FDA approval, there are other TCKIs that are under clinical investigation. A number of articles describing the design, validation, and application of bioanalytical methods for tyrosine KIs have been published as a result of the growing need for bioanalytical techniques to qualitatively and quantitatively study such substances9, 10,11.
For example, the signalling network of the epidermal growth factor receptor includes 211 biological reactions and 322 signalling compounds12. The EGFR family encompasses four types of growth factor receptors, such as EGFR (ErbB1- EGFR/HER1R, ErbB2 (HER-2 neu in rodents), ErbB3 (HER-3) and ErbB4 (HER-4). Many human tumours have mutations that result in continuous activation, amplification, and overexpression of EGFR proteins, including tumours of the mammary glands, ovaries, kidneys, and lung. These alterations are a factor in tumour metastasis. EGFR overexpression or excess activation is common in Non-small cell lung cancer (NSCLC), which is the foremost reason for cancer-associated deaths in the Western world13,14,15,16 EGFR mutations are found in about 10% of all patients, but this rate can reach 40% in Asians, non-smokers, and patients with adenocarcinoma histology. The most common EGFR mutations are deletions at exon 19 and point mutations at exon 21 (L858R). The existence of these alterations suggests susceptibility to EGFR TKIs like gefitinib, erlotinib, and afatinib. Gefitinib and Erlotinib (first generation), second generation (Afatinib, dacomitinib, neratinib), and third generation (AZD9291 (osimertinib), CO-1686 (rociletinib), HM61713, EGF816X, ASP8273) TKIs) TKIs are now a prominent therapeutic option widely chosen in daily routine practice because of the considerable survival benefits they have produced17,18.
As numerous attempts are made to investigate more Tyrosine KI candidates, For the development of this innovative class of medications, bioanalysis is increasingly important, particularly for the pharmacokinetic and pharmacodynamic evaluation of those chemicals. Because of this particular cause, therapeutic drug monitoring (TDM) of TKIs and TKIs is frequently utilised for clinical outcomes, highlighting the significance of TCKIs bioanalysis in a regular clinical environment19.
In light of the quick rise and development of TCKIs, it is crucial to have this review describe the mechanism of TCKIs, current bioanalytical assays of tyrosine KIs, and their metabolites in order to offer a glimpse into the most recent bioanalytical patterns in this oncologic research area and to provide preliminary data that could be helpful to developing bioanalytical methods for approaching potential molecules in the TCKIs group.
Figure 1: Mechanism of Tyrosine kinase pathway8
Analytical and Bioanalytical Techniques:
Pharmacokinetic knowledge is necessary for the discovery and characterization of novel therapeutics and drug candidates, including TCKIs. To determine a drug's pharmacokinetic properties, reliable and precise bioanalytical procedures to measure the amount of the drug and its by-products in human biological fluids are needed. In many cases, preliminary research on small rodents (mice and rats) is the first step before moving on to clinical treatments using human samples. Samples generated from blood (i.e., serum and plasma) are used for the majority of bioanalytical procedures because they provide the fundamental information required for study and the assessment of treatment outcomes as well as the efficacy of drugs20. Plasma is the biological resource most frequently used in the clinical context for drug assessment for TCKIs. The development of quick, accurate, and highly sensitive bio-analytical tests is required to accomplish this goal. Only a few HPTLC, High Performance Liquid Chromatography –UV detector, and LC-MS/MS procedures have been published.
The sample techniques which are typically used in bioanalysis include:
MS DETECTION:
The fraction of a molecule that has become an ion or has subsequently produced smaller fragments is detected by a mass spectrometer (MS). In TCKI Bioanalytical assays, tandem mass spectrometry (MS/MS) is frequently utilised. When compared to other techniques, MS has some advantages, such as its outstanding specificity and responsiveness Also, it has a connection to both gas-phase (GC-MS) separation and liquid-phase (LC-MS) and. Liquid chromatography in conjunction with mass spectrometry is currently the primary method used to analyse small-molecule tyrosinase inhibitors (LC-MS)21,22,23,24 .
As these advances in technology, it has lately been used to analyse metabolites and other chemicals in biological fluids including urine and plasma. The phrase "MS/MS" refers to the mass spectrometry investigation, which involves subjecting mass-selected ions to a second spectrometric measurement. Thus, for MS/MS, at least two phases of m/z analysis are required. There are various MS/MS varieties that are offered commercially. Although only these MS/MS instruments have been used in TCKI's bioanalysis to date only quadrupole TOF (Q-TOF, triple quadrupole (QqQ), and quadrupole-orbitrap (Q-Orbitrap) will be taken into consideration in this study 25,26,27.
In comparison to light spectroscopic detection techniques, bioanalytical studies using MS detectors require a rather expensive detector, and such equipment isn't always accessible in every diagnostic laboratory or hospital. So, instead of using MS/MS in such a case, a different detector like a UV-Vis/diode array detector (DAD) might prove more appropriate. The selectivity of DAD is reported to be at least one or two orders of magnitude less sensitive than low-resolution MS 28, 29. Higher sample volumes with greater analyte quantities (ca. 100–500 µL) may be an alternative to solve this issue30. According to Croitoru et al., sensitivity below 1 ng/ml can be attained by Diode Array Detector in ibrutinib quantification with a minimum of 1 ml of human plasma samples prepared by the liquid-liquid Extraction technique. Consequently, the necessity of 1 ml of human plasma sample will be a difficulty in preclinical research using small rodents (rats). This is likely the cause of the reason that plasma is used for Tyrosine KIs bioanalysis using an ultraviolet-diode array detector (Table1) 31.
For example, Wen Jiang et al employed a method and then compared it with other published techniques. They effectively built and optimized a method employing UPLC-MS/MS for the detection of 9 tyrosine KIs in plasma. They employed a Hypersil GOLD VANQUISH C18 column with a solvent system of ACN and 0.1% formic acid in water. The following procedure for gradient elution was executed: The mobile phase B was initially set to 85 percent for 0.5 minutes, after which the proportion was gradually reduced to 5% at 0.5-2 minutes and kept for 4 minutes, the mobile phase B was then quickly restored to 85%, and in the final minute, equilibrium was maintained.
They also stated that the analysis took 8 minutes to complete and that the flow rate of mobile phase was 0.3 ml/min with a 5 L injection volume. The drugs like cabozantinib, apatinib, Lenvatinib, sorafenib, gefitinib, regorafenib, and anlotinib all showed good linearity over the 0.1–10 ng/ml range, and tivantinib and galunisertib showed linearity over the entire 1-100 ng/ml range. Linear correlation coefficients for all standard curves are less than 0.9966. Finally, the study found that the limits of quantitation at 0.003 to 0.11 ng/ml and detection (LOD) at 0.01-0.37 ng/ml. This method's selectivity, accuracy, and matrix effect (ME) range of 90.48–107.77% were all assessed as adequate 32.
Table 1: Detailed information on bioanalysis of TKIs are listed above 31
|
Drugs |
Another drug/ metabolite |
Medium |
Column |
Mobile-phase |
Elution |
Range (ng/ mL) |
Detector |
|
Naquotinib33 |
Not any |
Rat Plasma |
Ethylene Bridged Hybrid-C18 |
A:5mM Ammonium formate buffer B: ACN-0.1% F |
Gradient |
5 – 491 |
QqQ |
|
Neratinib 34 |
No |
Human liver microsomes |
Eclipse Plus C18 |
Acetonitrile-10 mM and NH4COOH (45:55 v/v) |
Isocratic mode |
5 – 500 |
QqQ |
|
Olmutinib 35 |
No |
Rat plasma |
Eclipse Plus RRHD C18 |
A: 0.1% Formic acid in water B: Acetonitrile |
Gradient mode |
1 – 500 |
QqQ |
|
Pelitinib 36 |
No |
Human plasma |
Ethylene Bridged Hybrid-C18 |
A: 0.1% Formic acid B: ACN C: water, D: CH3OH |
Gradient mode |
1 – 200 |
QqQ |
|
Spebrutinib 37 |
No |
Human plasma |
Eclipse C18 |
10mM Ammonium formate and ACN (60:40 v/v) |
Isocratic mode |
5 – 500 |
QqQ |
Table 2: Determination of Tyrosine KIs by using HPLC Method
|
Drugs |
Material |
Column |
Mobile phase |
Detection (UV)nm |
Ref |
|
Dasatinib |
Dosage form |
C18 |
CH3OH and ACN mixed in the ratio of 50:50 v/v |
323 |
|
|
Lapatinib |
Tablet dosage form |
ODS C18 RP |
ACN and H2O (50:50 v/v) |
232 |
|
|
Sorafinib |
Tablet |
Phenomen ex C18 |
ACN and water in the ratio of 82. 5: 17.5 v/v. |
265 |
|
|
Imatinib |
Pharmaceutical dosage form |
Phenomenex (4.6mm X150 mm) 5µ |
Orthophosphori-c (pH 2.5): CH3OH (50:50) |
263 |
|
|
Nintedanib |
Drug |
Silica gel-60 F-254 |
CHCl3:CH3OH in the ratio 7:3 v/v. |
386 |
|
|
Sunitinib |
Human plasma |
Cyanopropyl |
Ammonium acetate buffer: ACN(55:45, v/v) |
431 |
Table 3: Validation parameters by using HPLC
|
Parameters |
Gefitinib |
Icotinib |
Erlotinib |
Crizotinib |
Osimertinib |
Afatinib |
|
Linearity range (ng per spot) |
100–1000 |
50.0–4000.0 |
50.0–4000. |
10.0–1000.0 |
10.0–1000.0 |
5.0–400.0 |
|
Precision (%R.S.D.) |
6.31 |
1.81 |
1.87 |
7.27 |
7.27 |
5.76 |
|
Inter-day (n = 6) |
10.59 |
-0.40 |
-5.64 |
7.71 |
11.38 |
8.92 |
|
intra-day (n = 6) |
13.04 |
−1.68 |
−3.25 |
7.71 |
12.35 |
10.33 |
HPLC:
In order to measure tyrosine KIs, such as imatinib, nilotinib, and dasatinib in body fluid, analytical techniques based on HPLC-UV were published.38,39
Yanping Liu et al., quantitatively analysed 6 TKIs (gefitinib, osimertinib, icotinib, erlotinib, afatinib and crizotinib) by using HPLC.40-46
HPTLC:
A recent study developed and validated a HPTLC technique for detection of imatinib mesylate that is straightforward, accurate, and resilience. The protein kinase inhibitor efficiently suppresses the Abelson tyrosine kinase. Protein Tyrosine kinase has a ability to transfers phosphate molecule from Adenosine Tri Phosphate to tyrosine residues in cytoplasmic protein substrates. PTKs have a vital function in modulating signal transduction pathways47. Because of its advantages, HPTLC is now becoming a standard analytical method48,49.
In contrast to HPLC, HPTLC enables the simultaneous testing of multiple samples using a minimal amount of mobile phase, which shortens analysis times and lowers per-analysis costs. In the literature, various methods for analysing imatinib mesylate are available, such as High Performance Liquid Chromatography and LC-MS-MS. However, there is no existing HPTLC approach to assessing imatinib mesylate in bulk and dosage form50, 51,52.
Furthermore, none of them is a method for indicating stability. They sampled the spotted with a Camag microliter syringe on precoated Plate 60F-254 using a Camag Linomat IV, according to previously published article. A continuous application speed of 100nl/s was used, with an 8 mm space between two bands. The mobile system was mixture of chloroform and methanol (6:4 v/v). The chamber was optimised, and the mobile phase system was run for 30 minutes at room temperature. The chromatogram run length was around 70 mm, with absorbance mode set to 276nm (Table.4)53.
Table 4: Overall validation parameters of imatinib mesylate
|
Parameter |
Data |
|
Linearity range (ng/spot) |
between 100–1000 |
|
Correlation coefficient |
0.9966 ± 0.0013 |
|
LOD (ng/spot) |
10 |
|
LOQ (ng per spot) |
30 |
|
Precision (%R.S. D) |
|
|
Repeatability of application (n = 6) |
0.35 |
|
Repeatability of measurement (n = 6) |
0.16 |
|
Interday (n = 6) |
0.74 |
|
Intraday (n = 6) |
0.48 |
|
Robustness |
Robust |
|
Specificity |
Specific |
DISCUSSION AND CONCLUSION:
In general, bioanalysis may contain as few manual processes as possible and simple, high-throughput experiments. The clinical relevance of bioanalysis of multianalytes in oncology is frequently noted because many cancer patients take numerous drugs. Gradient elution is frequently utilised in these situations, with more than 10 analytes, to get a trustworthy separation in the least amount of run time54.
The bioanalysis of TCKIs is significant for the study of drugs and cancer. The tests created in recent years offer a better understanding of PK/PD characteristics, metabolism, drug-drug interactions, and drug-food interactions. These assays can increase efficacy and decrease the toxicity of tyrosine KIs through TDM in conjunction with research on the therapeutic target level and clinical exposure effect. The researchers have an interest in learning more about this group of anti-cancer drugs. This article's information cuts down on the time and amount invested in developing analytical methods for analysing tyrosine kinase inhibitors from the very beginning. This knowledge will be useful for any future studies.
CONFLICT OF INTEREST:
The authors have no conflicts of interest regarding this investigation.
ACKNOWLEDGEMENT:
The authors are thankful to the principal and management of Acharya and BM Reddy College of Pharmacy, Bengaluru for providing facilities including plagiarism, interest, books etc.
FUNDING:
This work did not receive any specific grant from funding agencies in the public, commercial, or not -for-profit sectors.
REFERENCES:
1. Eskens FA. Angiogenesis inhibitors in clinical development; where are we now and where are we going?. British Journal of Cancer. 2004; 90(1): 1-7. https://doi.org/10.1038/sj.bjc.6601401
2. Kannaiyan R. Mahadevan D. A comprehensive review of protein kinase inhibitors for cancer therapy. Expert Review of Anticancer Therapy. 2018; 18(12): 1249-70. https://doi.org/10.1080/14737140.2018.1527688
3. Bhullar KS. Lagarón NO. McGowan EM. Parmar I. Jha A. Hubbard BP. Rupasinghe HV. Kinase-targeted cancer therapies: progress, challenges and future directions. Molecular Cancer. 2018; 17: 1-20. https://doi.org/10.1186/s12943-018-0804-2
4. Du Z. Lovly CM. Mechanisms of receptor tyrosine kinase activation in cancer. Molecular Cancer. 2018; 17: 1-3. https://doi.org/10.1186/s12943-018-0782-4
5. Zhao Z. Bourne PE. Progress with covalent small-molecule kinase inhibitors. Drug Discovery Today. 2018; 23(3): 727-35. https://doi.org/10.1016/j.drudis.2018.01.035
6. Maurer G. Tarkowski B. Baccarini M. Raf kinases in cancer–roles and therapeutic opportunities. Oncogene. 2011; 30(32): 3477-88. https://doi.org/10.1038/onc.2011.160
7. Gudimetla K. Prabhakar O. Pal A. Review on Pathophysiological and Pharmacotherapeutic approach on Chronic Myeloid Leukemia. Research Journal of Pharmacy and Technology. 2020; 13(6): 2971-6. http://dx.doi.org/10.5958/0974-360X.2020.00526.0
8. Oda K.. Matsuoka Y. Funahashi A. Kitano H. A comprehensive pathway map of epidermal growth factor receptor signaling. Molecular Systems Biology. 2005; 1(1): 2005-0010. https://doi.org/10.1038/msb4100014
9. Syed YY. Zanubrutinib: first approval. Drugs. 2020; 80(1): 91-7. https://doi.org/10.1007/s40265-019-01252-4 Markham A. Dhillon S. Acalabrutinib: First Global Approval. Drugs, 2018; 78: 139-45. https://doi.org/10.1007/s40265-017-0852-8
10. Lemmon MA. Schlessinger J. Cell signaling by receptor tyrosine kinases. Cell. 2010; 141(7): 1117-34. https://doi.org/10.1016/j.cell.2010.06.011
11. Arteaga CL. The epidermal growth factor receptor: from mutant oncogene in nonhuman cancers to therapeutic target in human neoplasia. Journal of clinical oncology: Official Journal of the American Society of Clinical Oncology. 2001; 19(18): 32S-40S.
12. Bergsland EK. Vascular endothelial growth factor as a therapeutic target in cancer. American journal of health-system pharmacy. 2004; 61(5): S4-11. https://doi.org/10.1093/ajhp/61.suppl_5.S4
13. Gudimetla K. Prabhakar O. Pal A. Review on Pathophysiological and Pharmacotherapeutic approach on Chronic Myeloid Leukemia. Research Journal of Pharmacy and Technology. 2020; 13(6): 2971-6. https://doi.org/10.5958/0974-360X.2020.00526.0
14. Wikstrand CJ. Bigner DD. Prognostic applications of the epidermal growth factor receptor and its ligand, transforming growth factor-α. JNCI: Journal of the National Cancer Institute. 1998; 90(11): 799-813. https://doi.org/10.1093/jnci/90.11.799
15. Shigematsu H. Lin L. Takahashi T. Nomura M. Suzuki M. Wistuba II. Fong KM. Lee H. Toyooka S. Shimizu N. Fujisawa T. Clinical and biological features associated with epidermal growth factor receptor gene mutations in lung cancers. Journal of the National Cancer Institute. 2005; 97(5): 339:46. https://doi.org/10.1093/jnci/dji055
16. Maione P. Sacco PC. Sgambato A. Casaluce F. Rossi A. Gridelli C. Overcoming resistance to targeted therapies in NSCLC: current approaches and clinical application. Therapeutic Advances in Medical Oncology. 2015; 7(5): 263-73. https://doi.org/10.1177/1758834015595048
17. Robert J. Flanagan. Eva C. Hans H. Maurer. Robin W. Therapeutic Drug Monitoring, in Fundamentals of Analytical Toxicology. 2020; 479-504. https://doi.org/10.1002/9781119122357.ch20
18. Kulkarni P. Karanam A. Gurjar M. Dhoble S. Naik AB. Vidhun BH. Gota V. Effect of various anticoagulants on the bioanalysis of drugs in rat blood: implication for pharmacokinetic studies of anticancer drugs. Springerplus. 2016; 5: 1-8. https://doi.org/10.1186/s40064-016-3770-4
19. Raju CP. Babu GR. Sowjanya M. Ramayyappa M. Evaluation of Cancer Bio-markers through Hyphenated Analytical Techniques. Asian Journal of Pharmaceutical Analysis. 2021; 11(3): 235-42. http://dx.doi.org/10.52711/2231-5675.2021.00041
20. Khan ZG. Bari SB. Gujarathi SN. Gujarathi SB. Patil PB. Azilsartan: A Review of Analytical Methods for estimation in Pharmaceutical Formulation. Asian Journal of Pharmaceutical Analysis. 2018; 8(4): 227-32. http://dx.doi.org/10.5958/2231-5675.2018.00041.8
21. Vyas AK. Mishra SB. Patel AB. Patel NK. Shah SR. Sheth DB. A brief review on liquid chromatography-mass spectrometry/ LC MS and its application. Asian Journal of Pharmaceutical Analysis. 2022; 12(3): 203-10. http://dx.doi.org/10.52711/2231-5675.2022.00034
22. Saha D. Design and Potential of Mass Spectrometry. Asian Journal of Pharmaceutical Analysis. 2011; 1(1): 1-2.
23. Liu W. Li S. Wu Y. Yan X. Zhu YM. Huang JH. Chen Z. Metabolic profiles of neratinib in rat by using ultra‐high‐performance liquid chromatography coupled with diode array detector and Q‐Exactive Orbitrap tandem mass spectrometry. Biomedical Chromatography. 2018; 32 (9): 42-72. https://doi.org/10.1002/bmc.4272
24. Dong J. Li S. Liu G. In vitro metabolism of ibrutinib in rat, dog and human hepatocytes using liquid chromatography combined with diode‐array detection and Q‐Exactive Orbitrap tandem mass spectrometry. Rapid Communications in Mass Spectrometry. 2019; 33(23): 1804-15. https://doi.org/10.1002/rcm.8542
25. Stoev G. Stoyanov A. Comparison of the reliability of the identification with diode array detector and mass spectrometry. Journal of Chromatography A. 2007; 1150(1-2): 302-11. https://doi.org/10.1016/j.chroma.2006.12.026
26. Carvalho DO. Curto AF. Guido LF. Determination of phenolic content in different barley varieties and corresponding malts by liquid chromatography-diode array detection-electrospray ionization tandem mass spectrometry. Antioxidants. 2015; 4(3): 563-76. https://doi.org/10.3390/antiox4030563
27. Rood JJ. Schellens JH. Beijnen JH. Sparidans RW. Recent developments in the chromatographic bioanalysis of approved kinase inhibitor drugs in oncology. Journal of Pharmaceutical and Biomedical Analysis. 2016; 130: 244-63. https://doi.org/10.1016/j.jpba.2016.06.037
28. Poitout‐Belissent F. Aulbach A. Tripathi N. Ramaiah L. Reducing blood volume requirements for clinical pathology testing in toxicologic studies points to consider. Veterinary Clinical Pathology. 2016; 45(4): 534-51. https://doi.org/10.1111/vcp.12429
29. Jiang W. Zhao T. Zhen X. Jin C. Li H. Ha J. Rapid Determination of 9 Tyrosine Kinase Inhibitors for the Treatment of Hepatocellular Carcinoma in Human Plasma by QuEChERS-UPLC-MS/MS. Frontiers in Pharmacology. 2022; 13: 920436. https://doi.org/10.3389/fphar.2022.920436
30. Alrabiah H. Kadi AA. Attwa MW. Abdelhameed AS. A simple liquid chromatography-tandem mass spectrometry method to accurately determine the novel third-generation EGFR-TKI naquotinib with its applicability to metabolic stability assessment. RSC Advances. 2019; 9(9): 4862-9. https://doi.org/10.1039/C8RA09812C
31. Khan H. Analytical Method Development in Pharmaceutical Research: Steps involved in HPLC Method Development. Asian Journal of Pharmaceutical Research. 2017; 7(3): 203-7. http://dx.doi.org/10.5958/2231-5691.2017.00031.4
32. Bao SS. Wen J. Liu TH. Zhang BW. Wang CC. Hu GX. A UHPLC–MS/MS method for the quantitation of olmutinib in rat plasma. Acta Chromatographica. 2019; 31(2): 105-8. https://doi.org/10.1556/1326.2018.00375
33. Maher HM. Alzoman NZ. Shehata SM. Abahussain AO. Comparative pharmacokinetic profiles of selected irreversible tyrosine kinase inhibitors, neratinib and pelitinib, with apigenin in rat plasma by UPLC–MS/MS. Journal of Pharmaceutical and Biomedical Analysis. 2017; 137: 258-67. https://doi.org/10.1016/j.jpba.2017.01.039
34. Abdelhameed AS. Attwa MW. Al-Shaklia NS. Kadi AA. A highly sensitive LC-MS/MS method to determine novel Bruton's tyrosine kinase inhibitor spebrutinib: application to metabolic stability evaluation. Royal Society Open Science. 2019; 6(6): 190-434. https://doi.org/10.1098/rsos.190434
35. Anastas PT. Warner JC. Green chemistry. Frontiers, 2004; 24(7-8): 775-799. http://dx.doi.org/10.1016/j.eiar.2004.06.006
36. Inturi S. Avula PR. A sensitive bioanalytical method development and validation of cabozantinib in human plasma by LC-ESI-MS/MS. Brazilian Journal of Pharmaceutical Sciences. 2018; 54(2): e17163. https://doi.or/10.1590/s2175-97902018000217163
37. Patil SD. Dugaje T. Kshirsagar SJ. Development and Validation of HPLC Method for Estimation of Pharmaceutical Drug and its Stability Studies in Simulated Biological Fluid: Comparative Study. Asian Journal of Pharmacy and Technology. 2019; 9(3): 179-84. http://dx.doi.org/10.5958/2231-5713.2019.00030.8
38. Satyanarayana L. Naidu SV. Rao MN. Latha RS. The Estimation of Nilotinib in Capsule dosage form by RP-HPLC. Asian Journal of Pharmaceutical Research. 2011; 1(3): 78-80.
39. Kalaichelvi R. Jayachandran E. Quantitative Estimation of SorafenibTosylate Its Pure Form and in Its Tablet Formulation by RP-HPLC Method. Journal of Chemistry. 2012; 2013(539264). https://doi.org/10.1155/2013/539264
40. Sandhya P. Vishnu P. Anjali N. Method development and validation of Imatinib Mesylate in Pharmaceutical dosage form by RP-HPLC. World Journal of Pharmacy and Pharmaceutical Sciences. 2013; 3 (1): 682-8.
41. Dutta D. Das S. Ghosh M. Validated HPTLC method for the determination of nintedanib in bulk drug. Multidisciplinary Digital Publishing Institute Proceedings. 2018; 9(1): 22. https://doi.org/10.3390/ecsoc-22-05675
42. Blanchet B. Saboureau C. Benichou AS. Billemont B. Taieb F. Ropert S. Dauphin A. Goldwasser F. Tod M. Development and validation of an HPLC-UV-visible method for sunitinib quantification in human plasma. Clinica Chimica Acta. 2009; 404(2): 134-9. https://doi.org/10.1016/j.cca.2009.03.042
43. Liu Y. Simultaneous and Rapid Determination of Six Tyrosine Kinase Inhibitors in Patients with Non-Small Cell Lung Cancer using HPLC-MS/MS. International Journal of Analytical Chemistry. 2021; 5524361. https://doi.org/10.1155%2F2021%2F5524361
44. Adcock IM. Chung KF. Caramori G. Ito K. Retracted: Kinase inhibitors and airway inflammation. European Journal of Pharmacology. 2006; 533(1-3): 118-32. https://doi.org/10.1016/j.ejphar.2005.12.054
45. Attimarad M. Ahmed KM. Aldhubaib BE. Harsha S. High-performance thin layer chromatography: A powerful analytical technique in pharmaceutical drug discovery. Pharmaceutical Methods. 2011; 2(2): 71-5. https://doi.org/10.4103/2229-4708.84436
46. Ivanovic D. Medenica M. Jancic B. Malenovic A. Reversed-phase liquid chromatography analysis of imatinib mesylate and impurity product in Glivec® capsules. Journal of Chromatography B.2004; 800(1-2): 253-8. https://doi.org/10.1016/j.jchromb.2003.10.018
47. Dhandhukia PC. Thakker JN. Quantitative analysis and validation of method using HPTLC. In High-performance thin-layer chromatography (HPTLC). 2010: 203-221. Berlin, Heidelberg: Springer Berlin Heidelberg. https://doi.org/10.1007/978-3-642-14025-9_12
48. Watt AP. Morrison D. Locker KL. Evans DC. Higher throughput bioanalysis by automation of a protein precipitation assay using a 96-well format with detection by LC− MS/MS. Analytical Chemistry. 2000; 72(5): 979-84. https://doi.org/10.1021/ac9906633
49. Reddy M. Swaminathan S. Review on Analytical Method Development and Validation by HPLC/LC-MS of Selected Anti-Cancer Drugs. World Journal of Pharmacy and Pharmaceutical Sciences. 2017; 6(4): 470-481. http://dx.doi.org/10.20959/wjpps20174-8876
50. Vadera N. Subramanian G. Musmade P. Stability-indicating HPTLC determination of imatinib mesylate in bulk drug and pharmaceutical dosage form. Journal of Pharmaceutical and Biomedical Analysis. 2007; 43(2): 722-6. https://doi.org/10.1016/j.jpba.2006.07.022
51. Merienne C. Rousset M. Ducint D. Castaing N. Titier K. Molimard M. Bouchet S. High throughput routine determination of 17 tyrosine kinase inhibitors by LC–MS/MS. Journal of Pharmaceutical and Biomedical Analysis. 2018; 150: 112-20. https://doi.org/10.1016/j.jpba.2017.11.060
52. Huynh HH. Pressiat C. Sauvageon H. Madelaine I. Maslanka P. Lebbé C. Thieblemont C. Goldwirt L. Mourah S. Development and validation of a simultaneous quantification method of 14 tyrosine kinase inhibitors in human plasma using LC-MS/MS. Therapeutic Drug Monitoring. 2017; 39(1): 43-54. https://doi.org/10.1097/FTD.0000000000000357
53. Koller D. Vaitsekhovich V. Mba C, Steegmann JL, Zubiaur P, Abad-Santos F, Wojnicz A. Effective quantification of 11 tyrosine kinase inhibitors and caffeine in human plasma by validated LC-MS/MS method with potent phospholipids clean-up procedure. Application to therapeutic drug monitoring. Talanta. 2020; 208: 120450. https://doi.org/10.1016/j.talanta.2019.120450
Received on 25.08.2023 Modified on 21.12.2023
Accepted on 19.02.2024 © RJPT All right reserved
Research J. Pharm. and Tech 2024; 17(6):2949-2954.
DOI: 10.52711/0974-360X.2024.00461