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

Systematic optimization of targeted and multiplexed MS-based screening workflows for protein biomarkers

    Neelam Khanal‡

    Bioanalysis, Immunogenicity & Biomarkers, In-vitro/In-vivo Translation, Research, GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA 19426, USA

    Scientific Operations, Waters Corporation, 34 Maple Street, Milford, MA 01757, USA

    ‡These authors contributed equally to this work

    Search for more papers by this author

    ,
    Zhuo Chen‡

    Bioanalysis, Immunogenicity & Biomarkers, In-vitro/In-vivo Translation, Research, GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA 19426, USA

    ‡These authors contributed equally to this work

    Search for more papers by this author

    ,
    Yun W Alelyunas

    Scientific Operations, Waters Corporation, 34 Maple Street, Milford, MA 01757, USA

    ,
    Matthew E Szapacs

    Bioanalysis, Immunogenicity & Biomarkers, In-vitro/In-vivo Translation, Research, GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA 19426, USA

    ,
    Mark D Wrona

    Scientific Operations, Waters Corporation, 34 Maple Street, Milford, MA 01757, USA

    &
    Timothy W Sikorski

    *Author for correspondence: Tel.: +1 610 270 4978;

    E-mail Address: timothy.w.sikorski@gsk.com

    Bioanalysis, Immunogenicity & Biomarkers, In-vitro/In-vivo Translation, Research, GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA 19426, USA

    Published Online:https://doi.org/10.4155/bio-2021-0245

    Background: The capability of targeted MS-based methods to simultaneously measure multiple analytes with high selectivity and sensitivity greatly facilitates the discovery and quantitation of novel biomarkers. However, the complexity of biological samples is a major bottleneck that requires extensive sample preparation. Results: This paper reports a generic workflow to optimize surrogate peptide-based protein biomarker screening for seven human proteins in a multiplexed manner without the need for any specific affinity reagents. Each step of the sample processing and LC–MS methods is systematically assessed and optimized for better analytical performance. Conclusion: The established method is used for the screening of multiple myeloma patient samples to determine which proteins could be robustly measured and serve as potential biomarkers of the disease.

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

    References

    • 1. Biomarkers Definitions Working Group. Biomarkers and surrogate endpoints: preferred definitions and conceptual framework. Clin. Pharmacol. Ther. 69(3), 89–95 (2001).
    • 2. Haab BB, Paulovich AG, Anderson NL et al. A reagent resource to identify proteins and peptides of interest for the cancer community: a workshop report. Mol. Cell. Proteom. 5(10), 1996–2007 (2006).
    • 3. Anderson NL, Anderson NG. The human plasma proteome: history, character, and diagnostic prospects. Mol. Cell. Proteom. 1(11), 845–867 (2002).
    • 4. Domon B, Aebersold R. Mass spectrometry and protein analysis. Science 312(5771), 212–217 (2006).
    • 5. Gillette MA, Carr SA. Quantitative analysis of peptides and proteins in biomedicine by targeted mass spectrometry. Nat. Methods 10(1), 28 (2013).
    • 6. Hüttenhain R, Malmström J, Picotti P, Aebersold R. Perspectives of targeted mass spectrometry for protein biomarker verification. Curr. Opin. Chem. Bio. 13(5–6), 518–525 (2009).
    • 7. Han X, Aslanian A, Yates JR III. Mass spectrometry for proteomics. Curr. Opin. Chem. Bio. 12(5), 483–490 (2008).
    • 8. Anderson L, Hunter CL. Quantitative mass spectrometric multiple reaction monitoring assays for major plasma proteins. Mol. Cell. Proteom. 5(4), 573–588 (2006).
    • 9. Whiteaker JR, Zhao L, Anderson L, Paulovich AG. An automated and multiplexed method for high throughput peptide immunoaffinity enrichment and multiple reaction monitoring mass spectrometry-based quantification of protein biomarkers. Mol. Cell. Proteom. 9(1), 184–196 (2010).
    • 10. Addona TA, Abbatiello SE, Schilling B et al. Multi-site assessment of the precision and reproducibility of multiple reaction monitoring-based measurements of proteins in plasma. Nature Biotechnol. 27(7), 633–641 (2009).
    • 11. Anderson NL, Anderson NG, Haines LR, Hardie DB, Olafson RW, Pearson TW. Mass spectrometric quantitation of peptides and proteins using Stable Isotope Standards and Capture by Anti-Peptide Antibodies (SISCAPA). J. Proteome Res. 3(2), 235–244 (2004).
    • 12. Burkhart JM, Schumbrutzki C, Wortelkamp S, Sickmann A, Zahedi RP. Systematic and quantitative comparison of digest efficiency and specificity reveals the impact of trypsin quality on MS-based proteomics. J. Proteomics. 75(4), 1454–1462 (2012).
    • 13. Ferńndez Ocaña M, James IT, Kabir M et al. Clinical pharmacokinetic assessment of an anti-MAdCAM monoclonal antibody therapeutic by LC-MS/MS. Anal. Chem. 84(14), 5959–5967 (2012).
    • 14. Carr SA, Abbatiello SE, Ackermann BL et al. Targeted peptide measurements in biology and medicine: best practices for mass spectrometry-based assay development using a fit-for-purpose approach. Mol. Cell. Proteomics 13(3), 907–917 (2014). • A comprehensive guide for targeted MS assay categorization based on the assay purpose and the required analytical characteristics.
    • 15. Furukawa Y, Kikuchi J. Molecular pathogenesis of multiple myeloma. Int. J. Clin. Oncol. 20(3), 413–422 (2015).
    • 16. Plumb RS, Johnson KA, Rainville P et al. UPLC/MS(E); a new approach for generating molecular fragment information for biomarker structure elucidation. Rapid Commun. Mass Spectrom. 20(13), 1989–1994 (2006).
    • 17. Strader MB, Tabb DL, Hervey WJ, Pan C, Hurst GB. Efficient and specific trypsin digestion of microgram to nanogram quantities of proteins in organic-aqueous solvent systems. Anal. Chem. 78(1), 125–134 (2006).
    • 18. Kusebauch U, Deutsch EW, Campbell DS, Sun Z, Farrah T, Moritz RL. Using PeptideAtlas, SRMAtlas, and PASSEL: comprehensive resources for discovery and targeted proteomics. Curr. Protoc. Bioinform. 46(1), 13.25.11–13.25.28 (2014).
    • 19. Chen Z, Alelyunas YW, Wrona MD, Kehler JR, Szapacs ME, Evans CA. Microflow UPLC and high-resolution MS as a sensitive and robust platform for quantitation of intact peptide hormones. Bioanalysis 11(13), 1275–1289 (2019). •• Real-world example indicating the benefits of microflow ultra performance liquid chromatography and high-resolution MS on quantitative bioanalysis.
    • 20. Matuszewski BK, Constanzer ML, Chavez-Eng CM. Strategies for the assessment of matrix effect in quantitative bioanalytical methods based on HPLC-MS/MS. Anal. Chem. 75(13), 3019–3030 (2003).
    • 21. Kloepfer A, Quintana JB, Reemtsma T. Operational options to reduce matrix effects in liquid chromatography – electrospray ionisation–mass spectrometry analysis of aqueous environmental samples. J. Chromatogr. A 1067(1–2), 153–160 (2005).
    • 22. Gangl ET, Annan M, Spooner N, Vouros P. Reduction of signal suppression effects in ESI-MS using a nanosplitting device. Anal. Chem. 73(23), 5635–5644 (2001).
    • 23. Van Hoof N, Courtheyn D, Antignac JP et al. Multi-residue liquid chromatography/tandem mass spectrometric analysis of beta-agonists in urine using molecular imprinted polymers. Rapid Commun. 19(19), 2801–2808 (2005).
    • 24. Guo B, Li C, Deng Z et al. A new method for measurement of (-)-sophocarpine, a candidate therapeutic for viral myocarditis, in plasma: application to a toxicokinetic study in beagle dogs. Rapid Commun. 19(19), 2840–2848 (2005).
    • 25. Quintana JB, Rodil R, Reemtsma T. Suitability of hollow fibre liquid-phase microextraction for the determination of acidic pharmaceuticals in wastewater by liquid chromatography–electrospray tandem mass spectrometry without matrix effects. J. Chromatogr. A 1061(1), 19–26 (2004).
    • 26. Lien GW, Chen CY, Wang GS. Comparison of electrospray ionization, atmospheric pressure chemical ionization and atmospheric pressure photoionization for determining estrogenic chemicals in water by liquid chromatography tandem mass spectrometry with chemical derivatizations. J. Chromatogr. A 1216(6), 956–966 (2009).
    • 27. Zhou W, Yang S, Wang PG. Matrix effects and application of matrix effect factor. Bioanalysis 9(23), 1839–1844 (2017).
    • 28. Chiva C, Ortega M, Sabido E. Influence of the digestion technique, protease, and missed cleavage peptides in protein quantitation. J. Proteome Res. 13(9), 3979–3986 (2014).
    • 29. Siepen JA, Keevil EJ, Knight D, Hubbard SJ. Prediction of missed cleavage sites in tryptic peptides aids protein identification in proteomics. J. Proteome Res. 6(1), 399–408 (2007).
    • 30. Hervey WJT, Strader MB, Hurst GB. Comparison of digestion protocols for microgram quantities of enriched protein samples. J. Proteome Res. 6(8), 3054–3061 (2007).
    • 31. Waas M, Bhattacharya S, Chuppa S et al. Combine and conquer: surfactants, solvents, and chaotropes for robust mass spectrometry based analyses of membrane proteins. Anal. Chem. 86(3), 1551–1559 (2014).
    • 32. Freije JR, Mulder PP, Werkman W et al. Chemically modified, immobilized trypsin reactor with improved digestion efficiency. J. Proteome Res. 4(5), 1805–1813 (2005).
    • 33. Ludwig KR, Schroll MM, Hummon AB. Comparison of in-solution, FASP, and S-Trap based digestion methods for bottom-up proteomic studies. J. Proteome Res. 17(7), 2480–2490 (2018).
    • 34. Leon IR, Schwammle V, Jensen ON, Sprenger RR. Quantitative assessment of in-solution digestion efficiency identifies optimal protocols for unbiased protein analysis. Mol. Cell. Proteomics 12(10), 2992–3005 (2013).
    • 35. Tanca A, Abbondio M, Pisanu S, Pagnozzi D, Uzzau S, Addis MF. Critical comparison of sample preparation strategies for shotgun proteomic analysis of formalin-fixed, paraffin-embedded samples: insights from liver tissue. Clin. Proteomics. 11(1), 28 (2014).
    • 36. Twomey JD, Brahme NN, Zhang B. Drug-biomarker co-development in oncology – 20 years and counting. Drug Resist. Updat. 30, 48–62 (2017).
    • 37. Tan J, Chen S, Huang J et al. High PD-1 and Tim-3 expression concurrent with exhausted CD4+ and CD8+ T cells in bone marrow compared with peripheral blood from patients with multiple myeloma. Blood 130(Suppl. 1), 4391 (2017).
    • 38. Shah N, Chari A, Scott E, Mezzi K, Usmani SZ. B-cell maturation antigen (BCMA) in multiple myeloma: rationale for targeting and current therapeutic approaches. Leukemia 34(4), 985–1005 (2020).
    • 39. Bandow JE. Comparison of protein enrichment strategies for proteome analysis of plasma. Proteomics 10(7), 1416–1425 (2010). • A nice review of the most popular protein enrichment strategies for protein analysis.
    • 40. Björhall K, Miliotis T, Davidsson P. Comparison of different depletion strategies for improved resolution in proteomic analysis of human serum samples. Proteomics 5(1), 307–317 (2005).
    • 41. Betancourt LH, De Bock PJ, Staes A et al. SCX charge state selective separation of tryptic peptides combined with 2D-RP-HPLC allows for detailed proteome mapping. J. Proteomics. 91, 164–171 (2013).
    • 42. Shi T, Fillmore TL, Sun X et al. Antibody-free, targeted mass-spectrometric approach for quantification of proteins at low picogram per milliliter levels in human plasma/serum. PNAS 109(38), 15395–15400 (2012).
    • 43. Pu J, An B, Vazvaei F, Qu J. Enrichment of protein therapeutics and biomarkers for LC–MS quantification. Bioanalysis 10(13), 979–982 (2018).