Assessment of In Silico and In Vitro Selpercatinib Metabolic Stability in Human Liver Microsomes Using a Validated LC-MS/MS Method

Selpercatinib (SLP; brand name Retevmo®) is a selective and potent RE arranged during transfection (RET) inhibitor. On 21 September 2022, the FDA granted regular approval to SLP (Retevmo, Eli Lilly, and Company). It is considered the only and first RET inhibitor for adults with metastatic or locally advanced solid tumors with RET gene fusion. In the current experiment, a highly specific, sensitive, and fast liquid chromatography tandem mass spectrometry (LC-MS/MS) method for quantifying SLP in human liver microsomes (HLMs) was developed and applied to the metabolic stability evaluation of SLP. The LC-MS/MS method was validated following the bioanalytical methodology validation guidelines outlined by the FDA (linearity, selectivity, matrix effect, accuracy, precision, carryover, and extraction recovery). SLP was detected by a triple quadrupole detector (TQD) using a positive ESI source and multiple reaction monitoring (MRM) mode for mass spectrometric analysis and estimation of analytes ions. The IS-normalized matrix effect and extraction recovery were acceptable according to the FDA guidelines for the bioanalysis of SLP. The SLP calibration standards were linear from 1 to 3000 ng/mL HLMs matrix, with a regression equation (y = 1.7298x + 3.62941) and coefficient of variation (r2 = 0.9949). The intra-batch and inter-batch precision and accuracy of the developed LC-MS/MS method were −6.56–5.22% and 5.08–3.15%, respectively. SLP and filgotinib (FLG) (internal standard; IS) were chromatographically separated using a Luna 3 µm PFP (2) stationary phase (150 × 4.6 mm) with an isocratic mobile phase at 23 ± 1 °C. The limit of quantification (LOQ) was 0.78 ng/mL, revealing the LC-MS/MS method sensitivity. The intrinsic clearance and in vitro t1/2 (metabolic stability) of SLP in the HLMs matrix were 34 mL/min/kg and 23.82 min, respectively, which proposed an intermediate metabolic clearance rate of SLP, confirming the great value of this type of kinetic experiment for more accurate metabolic stability predictions. The literature review approved that the established LC-MS/MS method is the first developed and reported method for quantifying SLP metabolic stability.

[1]  A. Kadi,et al.  Development and Validation of a Rapid LC-MS/MS Method for Quantifying Alvocidib: In Silico and In Vitro Metabolic Stability Estimation in Human Liver Microsomes , 2023, Molecules.

[2]  A. Kadi,et al.  Development of an LC-MS/MS Method for Quantification of Sapitinib in Human Liver Microsomes: In Silico and In Vitro Metabolic Stability Evaluation , 2023, Molecules.

[3]  A. Kadi,et al.  A validated LC-MS/MS analytical method for the quantification of pemigatinib: metabolic stability evaluation in human liver microsomes , 2022, RSC advances.

[4]  M. Gaze,et al.  Treatment outcome with a selective RET tyrosine kinase inhibitor selpercatinib in children with multiple endocrine neoplasia type 2 and advanced medullary thyroid carcinoma. , 2021, European journal of cancer.

[5]  Kun Wang,et al.  Determination of selpercatinib, a RET kinase inhibitor, in rat plasma and its application to a pharmacokinetic study. , 2020, Biomedical chromatography : BMC.

[6]  Jiang Liu,et al.  FDA Approval Summary: Selpercatinib for the Treatment of Lung and Thyroid Cancers with RET Gene Mutations or Fusions , 2020, Clinical Cancer Research.

[7]  J. Beijnen,et al.  Quantitative bioanalytical assay for the selective RET inhibitors selpercatinib and pralsetinib in mouse plasma and tissue homogenates using liquid chromatography-tandem mass spectrometry. , 2020, Journal of chromatography. B, Analytical technologies in the biomedical and life sciences.

[8]  G. Rossi,et al.  Diagnosis and treatment of early and locally advanced non-small-cell lung cancer: The 2019 AIOM (Italian Association of Medical Oncology) clinical practice guidelines. , 2020, Critical reviews in oncology/hematology.

[9]  G. Lippi,et al.  Current Cancer Epidemiology , 2019, Journal of epidemiology and global health.

[10]  V. Adamo,et al.  RET fusions in solid tumors. , 2019, Cancer treatment reviews.

[11]  A. Gunia-Krzyżak,et al.  Metabolic stability and its role in the discovery of new chemical entities , 2019, Acta pharmaceutica.

[12]  A. Kadi,et al.  Validated LC-MS/MS assay for quantification of the newly approved tyrosine kinase inhibitor, dacomitinib, and application to investigating its metabolic stability , 2019, PloS one.

[13]  A. Kadi,et al.  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 , 2019, RSC advances.

[14]  Jonathan D. Tyzack,et al.  Computational methods and tools to predict cytochrome P450 metabolism for drug discovery , 2019, Chemical biology & drug design.

[15]  A. Kadi,et al.  Identification of reactive intermediate formation and bioactivation pathways in Abemaciclib metabolism by LC–MS/MS: in vitro metabolic investigation , 2019, Royal Society Open Science.

[16]  A. Drilon,et al.  A phase 1 study of LOXO-292, a potent and highly selective RET inhibitor, in patients with RET-altered cancers. , 2018 .

[17]  Jonathan D. Tyzack,et al.  WhichP450: a multi-class categorical model to predict the major metabolising CYP450 isoform for a compound , 2018, Journal of Computer-Aided Molecular Design.

[18]  A. Kadi,et al.  LC–MS/MS method for the quantification of masitinib in RLMs matrix and rat urine: application to metabolic stability and excretion rate , 2017, Chemistry Central Journal.

[19]  Johannes Kirchmair,et al.  Drug Metabolism Prediction , 2014 .

[20]  Hoa Le,et al.  Comparison of metabolic soft spot predictions of CYP3A4, CYP2C9 and CYP2D6 substrates using MetaSite and StarDrop. , 2011, Combinatorial chemistry & high throughput screening.

[21]  James V Belcastro,et al.  An automated liquid chromatography-mass spectrometry process to determine metabolic stability half-life and intrinsic clearance of drug candidates by substrate depletion. , 2008, Assay and drug development technologies.

[22]  D. Leahy,et al.  Integrating invitro ADMET data through generic physiologically based pharmacokinetic models , 2006, Expert opinion on drug metabolism & toxicology.

[23]  J. Brockmöller,et al.  Benzydamine N-oxidation as an index reaction reflecting FMO activity in human liver microsomes and impact of FMO3 polymorphisms on enzyme activity. , 2000, British journal of clinical pharmacology.

[24]  W. Busby,et al.  Effect of methanol, ethanol, dimethyl sulfoxide, and acetonitrile on in vitro activities of cDNA-expressed human cytochromes P-450. , 1999, Drug metabolism and disposition: the biological fate of chemicals.

[25]  M. Barinaga From Bench Top to Bedside , 1997, Science.

[26]  D J Rance,et al.  The prediction of human pharmacokinetic parameters from preclinical and in vitro metabolism data. , 1997, The Journal of pharmacology and experimental therapeutics.

[27]  J B Houston,et al.  Utility of in vitro drug metabolism data in predicting in vivo metabolic clearance. , 1994, Biochemical pharmacology.

[28]  D. Pessayre,et al.  Inactivation of cytochrome P-450 by the drug methoxsalen. , 1986, The Journal of pharmacology and experimental therapeutics.