We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×

Non-regulated LC–MS/MS bioanalysis in support of early drug development: a Novartis perspective

    Yunlin Fu

    *Author for correspondence: Tel.: +1 862 778 1726;

    E-mail Address: yunlin.fu@novartis.com

    Pharmacokinetic Sciences – Drug Disposition, Novartis Institutes for BioMedical Research, One Health Plaza, East Hanover, NJ 07936, USA

    ,
    Wenkui Li

    Pharmacokinetic Sciences – Drug Disposition, Novartis Institutes for BioMedical Research, One Health Plaza, East Hanover, NJ 07936, USA

    &
    Franck Picard

    Pharmacokinetic Sciences – Drug Disposition, Novartis Institutes for BioMedical Research, Basel, CH-4056, Switzerland

    Published Online:https://doi.org/10.4155/bio-2022-0204

    Scientifically qualified LC–MS/MS methods are essential for the determination of small molecule drug candidates and/or their metabolite(s) in support of various non-regulated safety assessment and in vivo absorption, distribution, metabolism and excretion studies in preclinical development. This article outlines an effective method development workflow to fit for this purpose. The workflow features a ‘universal’ protein precipitation solvent for efficient sample extraction, a mobile phase additive for managing chromatographic resolution and addressing carryover and an internal standard cocktail to select the best analogue internal standard to track the analyte of interest in LC–MS/MS. In addition, good practices are recommended to prevent bioanalytical pitfalls due to instability, non-specific binding and dosing vehicle-induced matrix effect. Proper handling of non-liquid matrix is also discussed.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Aubry A-F, Shen JX. Bioanalysis and where does it fit into the drug development process? In: Bioanalysis for Service Users. Future Science Limited, London, UK, 6–16 (2013).
    • 2. Bhosale A. Importance of bioanalysis in drug discovery and development: a review. International Journal of Pharmaceutical Science and Research 5(1), 55–58 (2020).
    • 3. Thakur A, Tan Z, Kameyama T et al. Bioanalytical strategies in drug discovery and development. Drug Metab. Rev. 53(3), 434–458 (2021).
    • 4. Pandey S, Pandey P, Tiwari G, Tiwari R. Bioanalysis in drug discovery and development. Pharm. Methods 1(1), 14–24 (2010).
    • 5. Unger S, Li W, Flarakos J, Tse FLS. Roles of LC–MS bioanalysis in drug discovery, development, and therapeutic drug monitoring. In: Handbook of LC‐MS Bioanalysis. Li WZhang JTse FLS (Eds). John Wiley & Sons, Inc., NJ, USA, 1–13 (2013). • A comprehensive perspectives on the application of LC–MS in bioanalysis in support of drug discovery and development, including early development.
    • 6. Yuan L, Aubry AF, Ji QC. A simple, effective approach for rapid development of high-throughput and reliable LC–MS/MS bioanalytical assays. Bioanalysis 8(17), 1809–1822 (2016).
    • 7. EMA. Guideline on bioanalytical method validation (2011). www.ema.europa.eu/en/documents/scientific-guideline/guideline-bioanalytical-method-validation_en.pdf
    • 8. US FDA. Guidance for industry: Bioanalytical Method Validation (2018). http://academy.gmp-compliance.org/guidemgr/files/UCM368107.PDF
    • 9. Liu G, Snapp HM, Ji QC, Arnold ME. Strategy of accelerated method development for high-throughput bioanalytical assays using ultra high-performance liquid chromatography coupled with mass spectrometry. Anal. Chem. 81(22), 9225–9232 (2009).
    • 10. Xue Y-J, Melo B, Vallejo M et al. An integrated bioanalytical method development and validation approach: case studies. Biomed. Chromatogr. 26(10), 1215–1227 (2012).
    • 11. Jemal M, Ouyang Z, Xia YQ. Systematic LC–MS/MS bioanalytical method development that incorporates plasma phospholipids risk avoidance, usage of incurred sample and well thought-out chromatography. Biomed. Chromatogr. 24(1), 2–19 (2010).
    • 12. Shou WZ. Discovery bioanalysis. Bioanalysis 4(9), 983–984 (2012).
    • 13. Kemper CJ, Koetzner L, Koleto M. A primer for best practices in tissue preparation for bioanalysis. Bioanalysis 4(21), 2621–2636 (2012).
    • 14. Xue YJ, Gao H, Ji QC et al. Bioanalysis of drug in tissue: current status and challenges. Bioanalysis 4(21), 2637–2653 (2012). •• A comprehensive overview on the best practice on tissue sample handling and bioanalysis.
    • 15. Fu Y, Li W, Flarakos J. Recommendations and best practices for calibration curves in quantitative LC–MS bioanalysis. Bioanalysis 11(15), 1375–1377 (2019).
    • 16. Gu H, Liu G, Wang J et al. Selecting the correct weighting factors for linear and quadratic calibration curves with least-squares regression algorithm in bioanalytical LC–MS/MS assays and impacts of using incorrect weighting factors on curve stability, data quality, and assay performance. Anal. Chem. 86(18), 8959–8966 (2014).
    • 17. Li W, Jian W, Fu Y. Basic sample preparation techniques in LC–MS bioanalysis. In: Sample Preparation in LC‐MS Bioanalysis. Li WJian WFu Y (Eds). John Wiley & Sons, Inc., NJ, USA, 1–30 (2019). •• A comprehensive overview on the three key sample preparation methods for LC–MS bioanalysis
    • 18. Jemal M, Xia YQ. LC–MS Development strategies for quantitative bioanalysis. Curr Drug Metab 7(5), 491–502 (2006).
    • 19. Fu Y, Barkley D, Li W et al. Evaluation, identification and impact assessment of abnormal internal standard response variability in regulated LC–MS bioanalysis. Bioanalysis 12(8), 545–559 (2020). •• A comprehensive overview of internal standard response monitoring and associated trouble-shooting.
    • 20. Li W, Li Y, Francisco DT, Naidong W. Hydrophilic interaction liquid chromatographic tandem mass spectrometric determination of atenolol in human plasma. Biomed. Chromatogr. 19(5), 385–393 (2005).
    • 21. Li W, Luo S, Li S et al. Simultaneous determination of ribavirin and ribavirin base in monkey plasma by high performance liquid chromatography with tandem mass spectrometry. J. Chromatogr B Anal. Technol. Biomed. Life Sci. 846(1-2), 57–68 (2007).
    • 22. García MC. The effect of the mobile phase additives on sensitivity in the analysis of peptides and proteins by high-performance liquid chromatography-electrospray mass spectrometry. J. Chromatogr B Anal. Technol. Biomed. Life Sci. 825(2), 111–123 (2005).
    • 23. Chen Y, Mehok AR, Mant CT, Hodges RS. Optimum concentration of trifluoroacetic acid for reversed-phase liquid chromatography of peptides revisited. J Chromatogr A 1043(1), 9–18 (2004).
    • 24. Bock MJ, Neilson KL, Dudley A. Use of trifluoroacetic acid to quantify small, polar compounds in rat plasma during discovery-phase pharmacokinetic evaluation. J. Chromatogr B Anal. Technol. Biomed. Life Sci. 856(1-2), 165–170 (2007).
    • 25. Back HM, Lee JH, Chae JW et al. A novel HPLC–MS/MS method for the simultaneous determination of astemizole and its major metabolite in dog or monkey plasma and application to pharmacokinetics. J Pharm Biomed Anal 114, 121–126 (2015).
    • 26. Heudi O, Barteau S, Picard F, Kretz O. A sensitive LC–MS/MS method for quantification of a nucleoside analog in plasma: Application to in vivo rat pharmacokinetic studies. J. Chromatogr B Anal. Technol. Biomed. Life Sci. 877, 1887–1893 (2009).
    • 27. Shou WZ, Naidong W. Simple means to alleviate sensitivity loss by trifluoroacetic acid (TFA) mobile phases in the hydrophilic interaction chromatography-electrospray tandem mass spectrometric (HILIC-ESI/MS/MS) bioanalysis of basic compounds. J. Chromatogr B Anal. Technol. Biomed. Life Sci. 825(2), 186–192 (2005).
    • 28. Li W, Luo S, Rebello S et al. A semi-automated LC–MS/MS method for the determination of LCI699, a steroid 11β-hydroxylase inhibitor, in human plasma. J. Chromatogr B Anal. Technol. Biomed. Life Sci. 960, 182–193 (2014). • A good demonstration of addition of acetic acid (with higher boiling temperature than trifluoroacetic acid [TFA]) to alleviate sensitivity loss due to the use of TFA in mobile phase in electrospray ionization LC–MS bioanalysis.
    • 29. Yamamoto E, Ishihama Y, Asakawa N. Application of partially fluorinated carboxylic acids as ion-pairing reagents in LC/ESI-MS. Talanta 127, 219–224 (2014).
    • 30. Fu Y, Li W, Flarakos J, Tse FLS. Quantitative analysis of pasireotide (SOM230), a cyclic peptide, in monkey plasma using liquid chromatography in combination with tandem mass spectrometry. J. Chromatogr B Anal. Technol. Biomed. Life Sci. 1008, 242–249 (2016).
    • 31. Smith KA, Merrigan SD, Johnson-Davis KL. Selecting a structural analog as an internal standard for the quantification of 6-methylmercaptopurine by LC–MS/MS. J. Appl. Lab. Med. 3(3), 384–396 (2018).
    • 32. Hill HM, Smith GT. Evaluation and elimination of carryover and/or contamination in LC–MS bioanalysis. In: Handbook of LC‐MS Bioanalysis. Li WZhang JTse FLS (Eds). John Wiley & Sons, Inc., NJ, USA, 259–273 (2013).
    • 33. Williams JS, Donahue SH, Gao H, Brummel CL. Universal LC–MS method for minimized carryover in a discovery bioanalytical setting. Bioanalysis 4(9), 1025–1037 (2012).
    • 34. Briscoe CJ, Hage DS. Factors affecting the stability of drugs and drug metabolites in biological matrices. Bioanalysis 1(1), 205–220 (2009).
    • 35. Yuan L, Sophia xu X, Ji QC. Challenges and recommendations in developing LC–MS/MS bioanalytical assays of labile glucuronides and parent compounds in the presence of glucuronide metabolites. Bioanalysis 12(9), 615–624 (2020).
    • 36. Li W, Zhang J, Tse FLS. Strategies in quantitative LC–MS/MS analysis of unstable small molecules in biological matrices. Biomed. Chromatogr. 25(1-2), 258–277 (2011).
    • 37. Hilhorst M, Van Amsterdam P, Heinig K et al. Stabilization of clinical samples collected for quantitative bioanalysis – a reflection from the European Bioanalysis Forum. Bioanalysis 7(3), 333–343 (2015).
    • 38. Yuan L, Fu Y, Zhang D et al. Use of a carboxylesterase inhibitor of phenylmethanesulfonyl fluoride to stabilize epothilone D in rat plasma for a validated UHPLC–MS/MS assay. J. Chromatogr B Anal. Technol. Biomed. Life Sci. 969, 60–68 (2014).
    • 39. Fung EN, Zheng N, Arnold ME, Zeng J. Effective screening approach to select esterase inhibitors used for stabilizing ester-containing prodrugs analyzed by LC–MS/MS. Bioanalysis 2(4), 733–743 (2010).
    • 40. Ji AJ, Jiang Z, Livson Y et al. Challenges in urine bioanalytical assays: overcoming nonspecific binding. Bioanalysis 2(9), 1573–1586 (2010).
    • 41. Li W, Luo S, Smith HT, Tse FL. Quantitative determination of BAF312, a S1P-R modulator, in human urine by LC–MS/MS: prevention and recovery of lost analyte due to container surface adsorption. J. Chromatogr B Anal. Technol. Biomed. Life Sci. 878(5-6), 583–589 (2010).
    • 42. Fu Y, Li W, Smith HT, Tse FL. An investigation of incurred human urine sample reanalysis failure. Bioanalysis 3(9), 967–972 (2011).
    • 43. Li F, Ewles M, Pelzer M et al. Case studies: the impact of nonanalyte components on LC–MS/MS-based bioanalysis: strategies for identifying and overcoming matrix effects. Bioanalysis 5(19), 2409–2441 (2013).
    • 44. Schuhmacher J, Zimmer D, Tesche F, Pickard V. Matrix effects during analysis of plasma samples by electrospray and atmospheric pressure chemical ionization mass spectrometry: practical approaches to their elimination. Rapid Commun. Mass Spectrom. 17(17), 1950–1957 (2003).
    • 45. Shou WZ, Naidong W. Post-column infusion study of the ‘dosing vehicle effect’ in the liquid chromatography/tandem mass spectrometric analysis of discovery pharmacokinetic samples. Rapid Commun. Mass Spectrom. 17(6), 589–597 (2003).
    • 46. Tong XS, Wang J, Zheng S et al. Effect of signal interference from dosing excipients on pharmacokinetic screening of drug candidates by liquid chromatography/mass spectrometry. Anal. Chem. 74(24), 6305–6313 (2002).
    • 47. Bruenner B, James C. Evaluation and elimination of matrix effects in LC–MS bioanalysis. 249–258 (2013).
    • 48. Liang X, Ubhayakar S, Liederer BM et al. Evaluation of homogenization techniques for the preparation of mouse tissue samples to support drug discovery. Bioanalysis 3(17), 1923–1933 (2011).
    • 49. Gao H, Williams J. Tissue sample preparation in LC‐MS bioanalysis. 201–216 (2019).
    • 50. Chen S, Wu JT, Huang R. Evaluation of surrogate matrices for standard curve preparation in tissue bioanalysis. Bioanalysis 4(21), 2579–2587 (2012).