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

Applications of high-resolution MS in bioanalysis

    Mike-Qingtao Huang

    * Author for correspondence

    Janssen Research & Development, LLC, Pharmaceutical Companies of Johnson & Johnson, 1000 Route 202 South, Raritan, NJ 08869, USA.

    ,
    Zhongping (John) Lin

    Frontage Laboratories Inc., 700 Pennsylvania Drive, Exton, PA 19341, USA

    &
    Naidong Weng

    Janssen Research & Development, LLC, Pharmaceutical Companies of Johnson & Johnson, 1000 Route 202 South, Raritan, NJ 08869, USA

    Published Online:https://doi.org/10.4155/bio.13.100

    High-resolution MS (HRMS) in conjunction with LC (LC–HRMS) has become available to many laboratories in the pharmaceutical industry. Due to its enhanced, though sometime perceived, specificity using the high-resolution power and its capability of simultaneous quantitation and structural elucidation using the post-acquisition data mining feature, utilization of LC–HRMS for bioanalysis could lead to potential rapid and reliable method development as well as sample analysis, thus generating both cost and resource savings. Here, we would like to share our perspectives about several current and future applications of LC–HRMS in bioanalysis. We will also discuss the factors influencing the quality of method establishment and potential pitfalls that need to be considered for the utilization of LC–HRMS in the field of regulated bioanalysis. We believe when utilized appropriately, LC–HRMS will play a significant role in the future landscape of quantitative bioanalysis.

    References

    • Maurer HH. Perspectives of liquid chromatography coupled to low- and high-resolution mass spectrometry for screening, identification, and quantification of drugs in clinical and forensic toxicology. Ther. Drug Monit.32,324–327 (2010).
    • Zhu M, Ma L, Zhang D et al. Detection and characterization of metabolites in biological matrices using mass defect filtering of liquid chromatography/high resolution mass spectrometry data. Drug Metab. Dispos.34,1722–1733 (2006).
    • Zhu M, Ma L, Zhang H, Humphreys WG. Detection and structural characterization of glutathione-trapped reactivemetabolites using liquid chromatography–high-resolution mass spectrometry and mass defect filtering. Anal. Chem.79,8333–8341 (2007).
    • Ma S, Zhu M. Recent advances in applications of liquid chromatography–tandem mass spectrometry to the analysis of reactive drug metabolites. Chem. Biol. Interact.179,25–37 (2009).
    • Zhang H, Zhang D, Ray K, Zhu M. Mass defect filter technique and its applications to drug metabolite identification by high-resolution mass spectrometry. J. Mass Spectrom.44,999–1016 (2009).
    • Jemal M. High-throughput quantitative bioanalysis by LC–MS/MS. Biomed. Chromatogr.14,422–429 (2000).
    • Henion J, Brewer E, Rule G. Preparation for LC–MS/MS: analyzing biological and environmental samples – knowing the basic requirements and the big picture of an LC–MS system can ensure success in most instances. Anal. Chem.70,650A–656A (1998).
    • Weng N, Chen YL, Shou W, Jiang X. Importance of injection solution composition for LC–MS/MS methods. J. Pharm. Biomed. Anal.26,753–767 (2001).
    • Korfmacher W. Principles and applications of LC–MS in new drug discovery. Drug Discov. Today10,1357–1367 (2005).
    • 10  Zhou S, Song Q, Tang Y, Weng N. Critical review of development, validation, and transfer for high throughput bioanalytical LC–MS/MS methods. Curr. Pharm. Anal.1,3–14 (2005).
    • 11  Korfmacher W. High-resolution mass spectrometry will dramatically change our drug-discovery bioanalytical procedures. Bioanalysis3,1169–1171 (2011).
    • 12  Josephs JL. HRMS: current usage, future directions and the promise of integration with unified data streams suited to post-acquisition mining. Bioanalysis4,471–476 (2012).
    • 13  Ma S, Chowdhury SK. Application of LC-high-resolution MS with ‘intelligent’ data mining tools for screening reactive drug metabolites. Bioanalysis4,501–510 (2012).
    • 14  Campbell JL, Le Blanc JC. Using high-resolution quadrupole TOF technology in DMPK analyses. Bioanalysis4,487–500 (2012).
    • 15  Miller VP. SPE-MS analysis of absorption, distribution, metabolism and excretion assays: a tool to increase throughput and streamline workflow. Bioanalysis4,111–1121 (2012).
    • 16  Ramanathan R, Korfmacher W. The emergence of high-resolution MS as the premier analytical tool in the pharmaceutical bioanalysis arena. Bioanalysis4,467–469 (2012).
    • 17  Andrews GL, Simons BL, Young JB, Hawkridge AM, Muddiman DC. Performance characteristics of a new hybrid quadrupole time-of-flight tandem mass spectrometer (TripleTOF® 5600). Anal. Chem.83,5442–5446 (2011).
    • 18  Junot C, Madalinski G, Tabet JC, Ezan E. Fourier transform mass spectrometry for metabolome analysis. Analyst135,2203–2219 (2010).
    • 19  Makarov A, Scigelova M. Coupling liquid chromatography to Orbitrap mass spectrometry. J. Chromatogr. A1217,3938–3945 (2010).
    • 20  Scigelova M, Makarov A. Advances in bioanalytical LC–MS using the Orbitrap mass analyzer. Bioanalysis1,741–754 (2009).
    • 21  Zhang NR, Yu S, Tiller P, Yeh S, Mahan E, Emary WB. Quantitation of small molecules using high-resolution accurate mass spectrometers-a different approach for analysis of biological samples. Rapid Commun. Mass Spectrom.23,1085–1094 (2009).
    • 22  Bateman KP, Kellmann M, Muenster H, Papp R, Taylorc L. Quantitative–qualitative data acquisition using a benchtop Orbitrap mass spectrometer. J. Am. Soc. Mass Spectrom.20,1441–1450 (2009).
    • 23  Kaufmann A, Butcher P, Maden K, Walker S, Widmer M. Quantitative and confirmative performance of liquid chromatography coupled to high-resolution mass spectrometry compared to tandem mass spectrometry. Rapid Commun. Mass Spectrom.25,979–992 (2011).
    • 24  Kaufmann A, Widmer M, Maden K. Post-interface signal suppression, a phenomenon observed in a single-stage Orbitrap mass spectrometer coupled to an electrospray interfaced liquid chromatograph. Rapid Commun. Mass Spectrom.24,2162–2170 (2010).
    • 25  Xia YQ, Lau J, Olah T, Jemal M. Targeted Quantitative bioanalysis in plasma using liquid chromatography/high-resolution accurate mass spectrometry: an evaluation of global selectivity as a function of mass resolving power and extraction window, with comparison of centroid and profile modes. Rapid Commun. Mass Spectrom.25,2863–2878 (2011).
    • 26  Wei C, Grace JE, Zvyaga TA, Drexler DM. Utility of high-resolution accurate MS to eliminate interferences in the bioanalysis of ribavirin and its phosphate metabolites. Bioanalysis4,1895–1905 (2012).
    • 27  Ramanathan R, Jemal M, Ramagiri S et al. It is time for a paradigm shift in drug discovery bioanalysis: from SRM to HRMS. J. Mass Spectrom.46,595–601 (2011).
    • 28  Ranasinghe A, Ramanathan R, Jemal M et al. Integrated quantitative and qualitative workflow for in vivo bioanalytical support in drug discovery using hybrid Q-TOF-MS. Bioanalysis4,511–528 (2012).
    • 29  Zhang J, Maloney J, Drexler DM et al. Cassette incubation followed by bioanalysis using high-resolution MS for in vitro ADME screening assays. Bioanalysis4,581–593 (2012).
    • 30  Fung EN, Xia YQ, Aubry AF, Zeng J, Olah T, Jemal M. Full-scan high resolution accurate mass spectrometry (HRMS) in regulated bioanalysis: LC–HRMS for the quantitation of prednisone and prednisolone in human plasma. J. Chromatogr. B.879,2919–2927 (2011).
    • 31  Han B, Copeland M, Geiser AG et al. Development of a highly sensitive, high-throughput, mass spectrometry-based assay for rat procollagen type-I N-terminal propeptide (PINP) to measure bone formation activity. J. Proteome Res.6,4218–4229 (2007).
    • 32  Ji C, Sadagopan N, Zhang Y, Lepsy C. A universal strategy for development of a method for absolute quantification of therapeutic monoclonal antibodies in biological matrices using differential dimethyl labeling coupled with ultra performance liquid chromatography–tandem mass spectrometry. Anal. Chem.81,9321–9328 (2009).
    • 33  Hoofnagle AN, Wener MH. The fundamental flaws of immunoassays and potential solutions using tandem mass spectrometry. J. Immunol. Methods347,3–11 (2009).
    • 34  Wu ST, Ouyang Z, Olah TV, Jemal ML. A strategy for liquid chromatography/tandem mass spectrometry based quantitation of pegylated protein drugs in plasma using plasma protein precipitation with water-miscible organic solvents and subsequent trypsin digestion to generate surrogate peptides for detection. Rapid Commun. Mass Spectrom.25,281–290 (2011).
    • 35  Plumb RS, Fujimoto G, Mather J et al. Comparison of the quantification of a therapeutic protein using nominal and accurate mass MS/MS. Bioanalysis4,605–615 (2012).
    • 36  Dillen L, Cools W, Vereyken L et al. Comparison of triple quadrupole and high-resolution TOF-MS for quantification of peptides. Bioanalysis4,565–579 (2012).
    • 37  Ramagiri S, Garofolo F. Large molecule bioanalysis using Q-TOF without predigestion and its data processing challenges. Bioanalysis4,529–540 (2012).
    • 38  Ruan Q, Ji QC, Arnold ME, Humphreys WG, Zhu M. Strategy and its implications of protein bioanalysis utilizing high-resolution mass spectrometric detection of intact protein. Anal. Chem.83,8937–8944 (2011).
    • 39  Ji QC, Rodila R, Gage EM, EL-Shourbagy TA. A strategy of plasma protein quantitation by selective reaction monitoring of an intact protein. Anal. Chem.75,7008–7014 (2003).
    • 40  Ji QC, Rodila R, Morgan S, Humerickhouse RA, EL-Shourbagy TA. Investigation of the immunogenicity of a protein drug using equilibrium dialysis and liquid chromatography tandem mass spectrometry detection. Anal. Chem.77,5529–5533 (2005).
    • 41  Gucinski AC, Boyne II MT. Evaluation of intact mass spectrometry for the quantitative analysis of protein therapeutics. Anal. Chem.84,8045–8051 (2011).
    • 42  Jian W, Edom RW, Wang D, Weng N, Zhang S. Relative quantitation of glycoisoforms of intact apolipoprotein C3 in human plasma by liquid chromatography–high-resolution mass spectrometry. Anal. Chem.85,2867–2874 (2013).
    • 43  Lin ZJ, Li W, Dai G. Application of LC–MS for quantitative analysis and metabolite identification of therapeutic oligonucleotides. J. Pharm. Biomed. Anal.44,330–341 (2007).
    • 44  Hopfgartner G, Varesio E. New approaches for quantitative analysis in biological fluids using mass spectrometric detection. Trends Anal. Chem.24,583–589 (2005).
    • 45  Jian W, Edom R, Weng N, Zannikos P, Zhang Z, Wang H. Validation and application of an LC–MS/MS method for quantitation of three fatty acid ethanolamides as biomarkers for fatty acid hydrolase inhibition in human plasma. J. Chromatogr. B878,1687–1699 (2010).
    • 46  Jian W, Edom RW, Weng N. Important considerations for quantitation of biomarkers using liquid chromatography and mass spectrometry (LC–MS), Bioanalysis4,2431–2434 (2012).
    • 47  Jemal M, Xia YQ. LC–MS development strategies for quantitative bioanalysis, Curr. Drug Metab.7,491–502 (2006).
    • 48  Gao S, Zhang ZP, Karnes HT. Sensitivity enhancement in liquid chromatography/atmospheric pressure ionization mass spectrometry using derivatization and mobile phase additives, J. Chromatogr. B825,98–110 (2005).
    • 49  Zhu LS, Lapko VN, Lee JW et al. A General approach for the quantitative analysis of bisphosphonates in human serum and urine by high-performance liquid chromatography/tandem mass spectrometry. Rapid Commun. Mass Spectrom.20,3421–3426 (2006).
    • 50  Li W, Li YH, Li AC, Zhou S, Weng N. Simultaneous determination of norethindrone and ethinyl estradiol in human plasma by high performance liquid chromatography with tandem mass spectrometry – experiences on developing a highly selective method using derivatization reagent for enhancing sensitivity. J. Chromatogr. B825,223–232 (2005).
    • 101  Murray KK, Boyd RK, Eberlin MN, Langley GJ, Li L, Naito Y. Standard definitions of terms relating to mass spectrometry, international union of pure and applied chemistry (IUPAC), Analytical Chemistry Division (2006). http://old.iupac.org/reports/provisional/abstract06/murray_310107.html (Accessed 21 November 2010)