30,916
Views
5
CrossRef citations to date
0
Altmetric
Special Report

Protein Quantification by LC–MS: A Decade of Progress Through the Pages of Bioanalysis

Pages 629-644 | Received 12 Feb 2019, Accepted 12 Mar 2019, Published online: 15 Apr 2019

References

  • Campbell JL , LeBlanc JCY. Peptide and protein drug analysis by MS: challenges and opportunities for the discovery environment. Bioanalysis3(6), 645–657 (2011).
  • Ewles M , GoodwinL. Bioanalytical approaches to analyzing peptides and proteins by LC–MS/MS. Bioanalysis3(12), 1379–1397 (2011).
  • Li F , FastD, MichaelS. Absolute quantitation of protein therapeutics in biological matrices by enzymatic digestion and LC–MS. Bioanalysis3(21), 2459–2480 (2011).
  • Szapacs M , MenckenT, WilliamsJ, LiY. Utilizing digestion procedures in the bioanalytical laboratory. Bioanalysis8(1), 29–36 (2016).
  • Zhang G , SextonDJ, FaucetteRRet al. 2D-LC–MS/MS to measure cleaved high-molecular weight kininogen in human plasma as a biomarker for C1-INH-HAE. Bioanalysis9(19), 1477–1491 (2017).
  • Hansen K , SzarkaS, EscoffierEet al. Glu-C, an alternative digestive enzyme for the quantitative LC–MS/MS analysis of an IgG-based antibody biotherapeutic. Bioanalysis10(13), 997–1007 (2018).
  • Fung EN , ZambitoF, HaulenbeekJet al. Targeting an acid labile aspartyl-prolyl amide bond as a viable alternative to trypsin digestion to generate a surrogate peptide for LC–MS/MS analysis. Bioanalysis6(22), 2985–2998 (2014).
  • Ouyang Z , FurlongMT, WuSet al. Pellet digestion: a simple and efficient sample preparation technique for LC–MS/MS quantification of large therapeutic proteins in plasma. Bioanalysis4(10), 17–28 (2012).
  • Yuan L , ArnoldME, AubryA-F, JiQC. Simple and efficient digestion of a monoclonal antibody in serum using pellet digestion: comparison with traditional digestion methods in LC–MS/MS bioanalysis. Bioanalysis4(24), 2887–2896 (2012).
  • Shida H , NaitoT, ShibataKet al. LC–MS/MS method for denosumab quantitation in human serum with rapid protein digestion using immobilized trypsin. Bioanalysis10(18), 1501–1510 (2018).
  • Regnier FE , KimJ. Accelerating trypsin digestion: the immobilized enzyme reactor. Bioanalysis6(19), 2685–2698 (2014).
  • Iwamoto N , TakanashiM, UminoYet al. Application of nano-surface and molecular-orientation limited proteolysis to LC–MS bioanalysis of cetuximab. Bioanalysis8(10), 1009–1020 (2016).
  • Gong C , ZengJ, AkinsanyaBet al. Development and validation of an LC–MS/MS assay for the quantitation of a PEGylated anti CD28 domain antibody in human serum: overcoming interference from antidrug antibodies and soluble target. Bioanalysis6(18), 2371–2383 (2014).
  • Bronsema KJ , BischoffR, vande Merbel NC. High-sensitivity LC–MS/MS quantification of peptides and proteins in complex biological samples: the impact of enzymatic digestion and internal standard selection on method performance. Anal. Chem.85(20), 9528–9535 (2013).
  • Wright K , DufieldD. Minimalistic sample preparation strategies for LC–MS quantification of large molecule biopharmaceuticals: a case study highlighting alpha-1 antitrypsin protein. Bioanalysis6(13), 1813–1825 (2014).
  • Pu J , AnB, VazvaeiF, QuJ. Bioanalysis. Enrichment of protein therapeutics and biomarkers for LC–MS quantification. Bioanalysis10(13), 979–982 (2018).
  • Wilffert D , BischoffR, vande Merbel NC. Antibody-free workflows for protein quantification by LC–MS/MS. Bioanalysis7(6), 763–779 (2015).
  • Wilffert D , AsselmanA, DonzelliRet al. Highly sensitive antibody-free μLC–MS/MS quantification of rhTRAIL in serum. Bioanalysis8(9), 881–890 (2016).
  • Bronsema KJ , BischoffR, BoucheM-Pet al. High-sensitivity quantitation of a Nanobody® in plasma by single-cartridge multidimensional SPE and ultra-performance LC–MS/MS. Bioanalysis7(1), 53–64 (2015).
  • Wilffert D , DonzelliR, AsselmanAet al. Quantitative antibody-free LC–MS/MS analysis of sTRAIL in sputum and saliva at the sub ng/mL level. J. Chromatogr. B1032, 205–210 (2016).
  • Shi J , WongJM, MaJet al. Reagent-free LC–MS/MS-based pharmacokinetic quantification of polyhistidine-tagged therapeutic proteins. Bioanalysis9(3), 251–264 (2017).
  • Fung EN , BryanP, KozhichA. Techniques for quantitative LC–MS/MS analysis of protein therapeutics: advances in enzyme digestion and immunocapture. Bioanalysis8(8), 847–856 (2016).
  • Zhao Y , GuH, ZhengN, ZengJ. Critical considerations for immunocapture enrichment LC–MS bioanalysis of protein therapeutics and biomarkers. Bioanalysis10(13), 987–995 (2018).
  • Vialaret J , BroutinS, PugnierCet al. What sample preparation should be chosen for targeted MS monoclonal antibody quantification in human serum? Bioanalysis 10(10), 723–735 (2018).
  • Sleczka BG , MehlJT, ShusterDJet al. Quantification of human mAbs in mouse tissues using generic affinity enrichment procedures and LC–MS detection. Bioanalysis6(13), 1795–1811 (2014).
  • Lee JW . Generic method approaches for monoclonal antibody therapeutics analysis using both ligand binding and LC–MS/MS techniques. Bioanalysis8(1), 19–27 (2016).
  • Chen L , LiuS, PagelsSet al. Development of an ELISA-LC–MS hybrid assay for quantification of biotherapeutics. Bioanalysis10(17), 1427–1438 (2018).
  • Wang Q , HanJ, ShaCet al. Novel strategy using tryptic peptide immunoaffinity-based LC–MS/MS to quantify denosumab in monkey serum. Bioanalysis9(19), 1451–1463 (2017).
  • Gupta V , LassmanME, LeeAYHet al. An evaluation of an aptamer for use as an affinity ligand reagent with MS: PCSK9 as an example protein. Bioanalysis8(15), 1557–1564 (2016).
  • Klont F , HadderinghM, HorvatovichPet al. Affimers as an alternative to antibodies in an affinity LC–MS assay for the quantification of the soluble receptor of advanced glycation end-products (sRAGE) in human serum. J. Prot. Res.17(8), 2892–2899 (2018).
  • Stevenson D , El-SharifHF, ReddySM. Selective extraction of proteins and other macromolecules from biological samples using molecular imprinted polymers. Bioanalysis8(21), 2255–2263 (2016).
  • Yang W , KernstockR, SimmonsN, AlakA. ELISA microplate: a viable immunocapture platform over magnetic beads for immunoaffinity-LC–MS/MS quantitation of protein therapeutics?Bioanalysis7(3), 307–318 (2015).
  • Lanshoeft C , HeudiO, CianferaniSet al. Quantitative analysis of hIgG1 in monkey serum by LC–MS/MS using mass spectrometric immunoassay. Bioanalysis8(10), 1035–1049 (2016).
  • Becker JO , HoofnagleAN. Replacing immunoassays with tryptic digestion-peptide immunoaffinity enrichment and LC–MS/MS. Bioanalysis4(3), 281–290 (2012).
  • Sucharski FK , MeierS, MiessCet al. Development of an automated, interference-free, 2D-LC–MS/MS assay for quantification of a therapeutic mAb in human sera. Bioanalysis10(13), 1023–1037 (2018).
  • Schultz GA , McCardleK, NeubertH. Large-scale implementation of sequential protein and peptide immunoaffinity enrichment LC/nanoLC–MS/MS for human β-nerve growth factor. Bioanalysis8(8), 753–764 (2016).
  • Howard JW , KayRG, PleasanceS, CreaserCS. UHPLC for the separation of proteins and peptides. Bioanalysis4(24), 2971–2988 (2012).
  • Kleinnijenhuis AJ , IngolaM, ToerscheJHet al. Quantitative bottom up analysis of infliximab in serum using protein A purification and integrated μLC-electrospray chip IonKey MS/MS technology. Bioanalysis8(9), 891–904 (2016).
  • Lassman ME , Fernandez-MetzlerC. Applications of low-flow LC-SRM for the analysis of large molecules in pharmaceutical R&D. Bioanalysis6(13), 1859–1867 (2014).
  • Shi T , QianW-J. Antibody-free PRISM-SRM for multiplexed protein quantification: is this the new competition for immunoassays in bioanalysis?Bioanalysis5(3), 267–269 (2013).
  • Sandra K , MortierK, JorgeLet al. LC–MS/MS quantification of next-generation biotherapeutics: a case study for an IgE binding Nanobody in cynomolgus monkey plasma. Bioanalysis6(9), 1201–1213 (2014).
  • Sandra K , SandraP. The opportunities of 2D-LC in the analysis of monoclonal antibodies. Bioanalysis7(22), 2843–2847 (2015).
  • Xu K , LiuL, MaiaMet al. A multiplexed hybrid LC–MS/MS pharmacokinetic assay to measure two co-adminstered monoclonal antibodies in a clinical study. Bioanalysis6(13), 1781–1794 (2014).
  • Zhu Y , D’ArienzoC, LouZet al. LC–MS/MS multiplexed assay for the quantitation of a therapeutic protein BMS-986089 and the target protein Myostatin. Bioanalysis8(3), 193–204 (2016).
  • Mekhssian K , MessJ-N, GarofoloF. Application of high-resolution MS in the quantification of a therapeutic monoclonal antibody in human plasma. Bioanalysis6(13), 1767–1779 (2014).
  • Plumb RS , FujimotoG, MatherJet al. Comparison of the quantification of a therapeutic protein using nominal and accurate mass MS/MS. Bioanalysis4(5), 605–615 (2012).
  • Bults P , MeintsM, SonessonAet al. Improving selectivity and sensitivity of protein quantitation by LC-HR-MS/MS: determination of somatropin in rat plasma. Bioanalysis10(13), 1009–1021 (2018).
  • Kay RG , HoardJ, StenssonS. A current perspective of supercharging reagents and peptide bioanalysis. Bioanalysis8(3), 157–161 (2016).
  • Furlong MT , ZhaoS, MylottWet al. Dual universal peptide approach to bioanalysis of human monoclonal antibody protein drug candidates in animal studies. Bioanalysis5(11), 1363–1376 (2013).
  • Furlong MT , TitschC, XuWet al. An exploratory universal LC–MS/MS assay for bioanalysis of hinge region-stabilized human IgG4 mAbs in clinical studies. Bioanalysis6(13), 1747–1758 (2014).
  • Kaur S , LiuL, CortesDFet al. Validation of a biotherapeutic immunoaffinity-LC–MS/MS assay in monkey serum: ‘plug-and-play’ across seven molecules. Bioanalysis8(15), 1565–1577 (2016).
  • Van den Broek I , van DongenWD. LC–MS-based quantification of intact proteins: perspective for clinical and bioanalytical applications. Bioanalysis7(15), 1943–1958 (2015).
  • Lanshoeft C , CianferaniS, HeudiO. Generic hybrid ligand binding assay liquid chromatography high-resolution mass spectrometry-based workflow for multiplexed human immunoglobulin G1 quantification at the intact protein level: application to preclinical pharmacokinetic studies. Anal. Chem.89(4), 2628–2635 (2017).
  • Ramagiri S , GarofoloF. Large molecule bioanalysis using Q-TOF without predigestion and its data processing challenges. Bioanalysis4(5), 529–540 (2012).
  • Jian W , KangL, BurtonL, WengN. A workflow for absolute quantitation of large therapeutic proteins in biological samples at intact level using LC-HRMS. Bioanalysis8(16), 1679–1691 (2016).
  • Qiu X , KangL, CaseMet al. Quantitation of intact monoclonal antibody in biological samples: comparison of different data processing strategies. Bioanalysis10(13), 1055–1067 (2018).
  • Jin W , BurtonL, MooreI. LC-HRMS quantitation of intact antibody drug conjugate trastuzumab emtansine from rat plasma. Bioanalysis10(11), 851–862 (2018).
  • Kellie JF , KehlerJR, KarlinseyMZ, SummerfieldSG. Toward best practices in data processing and analysis for intact biotherapeutics by MS in quantitative bioanalysis. Bioanalysis9(23), 1883–1893 (2017).
  • Zhang L , VasicekLA, HsiehSCet al. Top-down LC–MS quantitation of intact denatured and native monoclonal antibodies in biological samples. Bioanalysis10(13), 1039–1054 (2018).
  • Kellie JF , KehlerJR, MenckenTet al. A whole-molecule immunocapture LC–MS approach for the in vivo quantitation of biotherapeutics. Bioanalysis8(20), 2103–2114 (2016).
  • Van de Merbel NC , BronsemaKJ, NemanskyM. Protein quantification using LC–MS: can it make a difference?Bioanalysis4(17), 2113–2116 (2012).
  • Heinrich J , StaackRF, StubenrauchK-G, PapadimitriouA. Proposal for a harmonized descriptive analyte nomenclature for quantitative large-molecule bioanalysis. Bioanalysis7(24), 3057–3062 (2015).
  • Zhang YJ , OlahTV, ZengJ. The integration of ligand binding and LC–MS-based assays into bioanalytical strategies for protein analysis. Bioanalysis6(13), 1827–1841 (2014).
  • Zhang YJ , LuoL, DesaiDD. Overview on biotherapeutic proteins: impact on bioanalysis. Bioanalysis8(1), 1–9 (2016).
  • White JT , BonillaLE. Free and total biotherapeutic evaluation in chromatographic assays: interference from targets and immunogenicity. Bioanalysis4(19), 2401–2411 (2012).
  • Staack RF , JordanG, DahlG, HeinrichJ. Free analyte concept: a novel approach to prove correct quantification of free therapeutic protein drug/biomarker concentrations. Bioanalysis6(4), 485–496 (2014).
  • Jordan G , OnamiI, HeinrichJ, StaackRF. Evaluation of the potential use of hybrid LC–MS/MS for active drug quantification applying the ‘free analyte QC concept’. Bioanalysis9(21), 1705–1717 (2017).
  • Ewles M , MannuR, FoxCet al. LC–MS/MS strategies for therapeutic antibodies and investigation into the quantitative impact of anti-drug antibodies. Bioanalysis8(24), 2565–2579 (2016).
  • Zhang H , GuH, ShipkovaPet al. Immunoaffinity LC–MS/MS for quantitative determination of a free and total protein target as a target engagement biomarker. Bioanalysis9(20), 1573–1588 (2017).
  • Bronsema KJ , BischoffR, PijnappelWWMPet al. Absolute quantification of the total and antidrug antibody-bound concentrations of recombinant human α-glucosidase in human plasma using protein G extraction and LC–MS/MS. Anal. Chem.87(8), 4394–4401 (2015).
  • Hamuro LL , KishnaniNS. Metabolism of biologics: biotherapeutic proteins. Bioanalysis4(2), 189–195 (2012).
  • Kellie JF , KarlinseyMZ. Review of approaches and examples for monitoring biotransformation in protein and peptide therapeutics by MS. Bioanalysis10(22), 1877–1890 (2018).
  • Bults P , BischoffR, BakkerHet al. LC–MS/MS-based monitoring of in vivo protein biotransformation: quantitative determination of tratstuzumab and its deamidation products in human plasma. Anal. Chem.88(3), 1871–1877 (2016).
  • Mehl JT , SleczkaBG, CiccimaroFet al. Quantification of in vivo site-specific Asp isomerization and Asn deamidation of mAbs in animal serum using IP-LC–MS. Bioanalysis8(15), 1611–1622 (2016).
  • Liu L , XuK, LiJet al. Optimizing hybrid LC–MS/MS binding conditions is critical: impact of biotransformation on quantification of trastuzumab. Bioanalysis10(22), 1819–1831 (2018).
  • Kang L , CamachoRC, LiWet al. Simultaneous catabolite identification and quantitation of large therapeutic protein at the intact level by immunoaffinity capture liquid chromatography–high-resolution mass spectrometry. Anal. Chem.89(11), 6065–6075 (2017).
  • Shi C , GoldbergS, LinTet al. Bioanalytical workflow for novel scaffold protein-drug conjugates: quantitation of total centyrin protein, conjugated centyrin and free payload for centyrin-drug conjugate in plasma and tissue samples using liquid chromatography–tandem mass spectrometry. Bioanalysis10(20), 1651–1665 (2018).
  • Zell M , HusserC, StaackRFet al. In vivo biotransformation of the fusion protein tetranectin-apolipoprotein A1 analyzed by ligand-binding mass spectrometry combined with quantitation by ELISA. Anal. Chem.88(23), 11670–11677 (2016).
  • Saad OM , ShenB-Q, XuKet al. Bioanalytical approaches for characterizing catabolism of antibody-drug conjugates. Bioanalysis7(13), 1583–1604 (2015).
  • Tumey LN , RagoB, HanX. In vivo biotransformation of antibody-drug conjugates. Bioanalysis7(13), 1649–1664 (2015).
  • Myler H , RanganVS, WangJet al. An integrated multiplatform bioanalytical strategy for antibody-drug conjugates: a novel case study. Bioanalysis7(13), 1569–1582 (2015).
  • Sanderson RJ , NicholasND, BakerLee Cet al. Antibody-conjugated drug assay for protease-cleavable antibody-drug conjugates. Bioanalysis8(1), 55–63 (2016).
  • Wang J , GuH, LiuAet al. Antibody-drug conjugate bioanalysis using LB-LC–MS/MS hybrid assays: strategies, methodology and correlation to ligand-binding assays. Bioanalysis8(13), 1383–1401 (2016).
  • Kaur S , XuK, SaadOMet al. Bioanalytical assay strategies for the development of antibody-drug conjugate biotherapeutics. Bioanalysis5(2), 201–226 (2013).
  • Xu K , LiuL, DereRet al. Characterization of the drug-to-antibody ratio distribution for antibody-drug conjugates in plasma/serum. Bioanalysis5(9), 1057–1071 (2013).
  • Grafmuller L , WeiC, RamanathanRet al. Unconjugated payload quantification and DAR characterization of antibody-drug conjugates using high-resolution MS. Bioanalysis8(16), 1663–1678 (2016).
  • Davis JA , KaganM, ReadJet al. Immunoprecipitation middle-up LC–MS for in vivo drug-to-antibody ratio determination for antibody-drug conjugates. Bioanalysis9(20), 1535–1549 (2017).
  • Trenchevska O , NelsonRW, NedelkovD. Mass spectrometric immunoassays for discovery, screening and quantification of clinically relevant proteoforms. Bioanalysis8(15), 1623–1633 (2016).
  • Hansbauer E-M , WorbsS, VollandHet al. Rapid detection of abrin toxin and its isoforms in complex matrices by immuno-extraction and quantitative high resolution targeted mass spectrometry. Anal. Chem.89(21), 11719–11727 (2017).
  • Duggan J , RenB, MaoYet al. LC–MS quantification of protein drugs: validating protein LC–MS methods with predigestion immunocapture. Bioanalysis8(18), 1951–1964 (2016).
  • Knutsson M , SchmidtR, TimmermanP. LC–MS/MS of large molecules in a regulated environment – which acceptance criteria to apply?Bioanalysis5(18), 2211–2214 (2013).
  • Duggan JX , VazvaeiF, JenkinsR. Bioanalytical method validation considerations for LC–MS/MS assays of therapeutic proteins. Bioanalysis7(11), 1389–1395 (2015).

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.