1932

Abstract

Transporters are key to understanding how an individual will respond to a particular dose of a drug. Two patients with similar systemic concentrations may have quite different local concentrations of a drug at the required site. The transporter profile of any individual depends upon a variety of genetic and environmental factors, including genotype, age, and diet status. Robust models (virtual patients) are therefore required and these models are data hungry. Necessary data include quantitative transporter profiles at the relevant organ. Liquid chromatography with tandem mass spectrometry (LC-MS/MS) is currently the most powerful method available for obtaining this information. Challenges include sourcing the tissue, isolating the hydrophobic membrane-embedded transporter proteins, preparing the samples for MS (including proteolytic digestion), choosing appropriate quantification methodology, and optimizing the LC-MS/MS conditions. Great progress has been made with all of these, especially within the last few years, and is discussed here.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-anchem-091719-024553
2020-06-12
2024-04-24
Loading full text...

Full text loading...

/deliver/fulltext/anchem/13/1/annurev-anchem-091719-024553.html?itemId=/content/journals/10.1146/annurev-anchem-091719-024553&mimeType=html&fmt=ahah

Literature Cited

  1. 1. 
    Di L, Artursson P, Avdeef A, Ecker GF, Faller B et al. 2012. Evidence-based approach to assess passive diffusion and carrier-mediated drug transport. Drug Discov. Today 17:15–16905–12
    [Google Scholar]
  2. 2. 
    Kell DB, Dobson PD, Oliver SG 2011. Pharmaceutical drug transport: the issues and the implications that it is essentially carrier-mediated only. Drug Discov. Today 16:15–16704–14
    [Google Scholar]
  3. 3. 
    Mendes P, Oliver SG, Kell DB 2015. Fitting transporter activities to cellular drug concentrations and fluxes: why the bumblebee can fly. Trends Pharmacol. Sci. 36:11710–23
    [Google Scholar]
  4. 4. 
    Scott DO, Ghosh A, Di L, Maurer TS 2017. Passive drug permeation through membranes and cellular distribution. Pharmacol. Res. 117:94–102
    [Google Scholar]
  5. 5. 
    Hillgren KM, Keppler D, Zur AA, Giacomini KM, Stieger B et al. 2013. Emerging transporters of clinical importance: an update from the International Transporter Consortium. Clin. Pharmacol. Ther. 94:152–63
    [Google Scholar]
  6. 6. 
    Bi YA, Costales C, Mathialagan S, West M, Eatemadpour S et al. 2019. Quantitative contribution of six major transporters to the hepatic uptake of drugs: “SLC-phenotyping” using primary human hepatocytes. J. Pharmacol. Exp. Ther. 370:172–83
    [Google Scholar]
  7. 7. 
    Giacomini KM, Balimane PV, Cho SK, Eadon M, Edeki T et al. 2013. International Transporter Consortium commentary on clinically important transporter polymorphisms. Clin. Pharmacol. Ther. 94:123–26
    [Google Scholar]
  8. 8. 
    Kalvass JC, Polli JW, Bourdet DL, Feng B, Huang SM et al. 2013. Why clinical modulation of efflux transport at the human blood-brain barrier is unlikely: the ITC evidence-based position. Clin. Pharmacol. Ther. 94:180–94
    [Google Scholar]
  9. 9. 
    Rose RH, Neuhoff S, Abduljalil K, Chetty M, Rostami-Hodjegan A, Jamei M 2014. Application of a physiologically based pharmacokinetic model to predict OATP1B1-related variability in pharmacodynamics of rosuvastatin. CPT Pharmacomet. Syst. Pharmacol. 3:e124
    [Google Scholar]
  10. 10. 
    Rostami-Hodjegan A. 2013. Response to “The link between pharmacodynamics and physiologically based pharmacokinetic models. .” Clin. Pharmacol. Ther. 93:2152
    [Google Scholar]
  11. 11. 
    Elmorsi Y, Barber J, Rostami-Hodjegan A 2016. Ontogeny of hepatic drug transporters and relevance to drugs used in pediatrics. Drug Metab. Dispos. 44:7992–98
    [Google Scholar]
  12. 12. 
    Giacomini K, Huang S, Tweedie D, Benet LZ, Brouwer LR et al. 2010. Membrane transporters in drug development. Nat. Rev. Drug Discov. 9:215–36
    [Google Scholar]
  13. 13. 
    Prasad B, Achour B, Artursson P, Hop C, Lai Y et al. 2019. Toward a consensus on applying quantitative liquid chromatography-tandem mass spectrometry proteomics in translational pharmacology research: a white paper. Clin. Pharmacol. Ther. 106:3525–43
    [Google Scholar]
  14. 14. 
    Kumar V, Salphati L, Hop C, Xiao G, Lai Y et al. 2019. A comparison of total and plasma membrane abundance of transporters in suspended, plated, sandwich-cultured human hepatocytes versus human liver tissue using quantitative targeted proteomics and cell surface biotinylation. Drug Metab. Dispos. 47:4350–57
    [Google Scholar]
  15. 15. 
    Harwood MD, Achour B, Neuhoff S, Russell MR, Carlson G et al. 2016. In vitro–in vivo extrapolation scaling factors for intestinal P-glycoprotein and breast cancer resistance protein: Part I: A cross-laboratory comparison of transporter-protein abundances and relative expression factors in human intestine and Caco-2 cells. Drug Metab. Dispos. 44:3297–307
    [Google Scholar]
  16. 16. 
    Gertz M, Tsamandouras N, Sall C, Houston JB, Galetin A 2014. Reduced physiologically-based pharmacokinetic model of repaglinide: impact of OATP1B1 and CYP2C8 genotype and source of in vitro data on the prediction of drug-drug interaction risk. Pharm. Res. 31:92367–82
    [Google Scholar]
  17. 17. 
    Bosgra S, van de Steeg E, Vlaming ML, Verhoeckx KC, Huisman MT et al. 2014. Predicting carrier-mediated hepatic disposition of rosuvastatin in man by scaling from individual transfected cell-lines in vitro using absolute transporter protein quantification and PBPK modeling. Eur. J. Pharm. Sci. 65:156–66
    [Google Scholar]
  18. 18. 
    Neuhoff S, Yeo KR, Barter Z, Jamei M, Turner DB, Rostami-Hodjegan A 2013. Application of permeability-limited physiologically-based pharmacokinetic models: part II—prediction of p-glycoprotein mediated drug-drug interactions with digoxin. J. Pharm. Sci. 102:93161–73
    [Google Scholar]
  19. 19. 
    Kim SJ, Lee KR, Miyauchi S, Sugiyama Y 2019. Extrapolation of in vivo hepatic clearance from in vitro uptake clearance by suspended human hepatocytes for anionic drugs with high binding to human albumin: improvement of in vitro-to-in vivo extrapolation by considering the “albumin-mediated” hepatic uptake mechanism on the basis of the “facilitated-dissociation model. .” Drug Metab. Dispos. 47:294–103
    [Google Scholar]
  20. 20. 
    Di Marco A, Gonzalez Paz O, Fini I, Vignone D, Cellucci A et al. 2019. Application of an in vitro blood-brain barrier model in the selection of experimental drug candidates for the treatment of Huntington's disease. Mol. Pharm. 16:52069–82
    [Google Scholar]
  21. 21. 
    Ball K, Bouzom F, Scherrmann JM, Walther B, Decleves X 2013. Physiologically based pharmacokinetic modelling of drug penetration across the blood-brain barrier—towards a mechanistic IVIVE-based approach. AAPS J 15:4913–32
    [Google Scholar]
  22. 22. 
    Gaohua L, Neuhoff S, Johnson TN, Rostami-Hodjegan A, Jamei M 2016. Development of a permeability-limited model of the human brain and cerebrospinal fluid (CSF) to integrate known physiological and biological knowledge: estimating time varying CSF drug concentrations and their variability using in vitro data. Drug Metab. Pharmacokinet. 31:3224–33
    [Google Scholar]
  23. 23. 
    Galetin A. 2014. Rationalizing underprediction of drug clearance from enzyme and transporter kinetic data: from in vitro tools to mechanistic modeling. Methods Mol. Biol. 1113:255–88
    [Google Scholar]
  24. 24. 
    Heikkinen AT, Parrott N, Dunkley T, Cutler P 2016. Targeted proteomics to support transporter IVIVE and PBPK. Drug Transporters: Volume 2: Recent Advances and Emerging Technologies: Royal Society of Chemistry KY Glynis Nicholls 44–72 London: Royal Soc. Chem.
    [Google Scholar]
  25. 25. 
    Bi YA, Kimoto E, Sevidal S, Jones HM, Barton HA et al. 2012. In vitro evaluation of hepatic transporter-mediated clinical drug-drug interactions: hepatocyte model optimization and retrospective investigation. Drug Metab. Dispos. 40:61085–92
    [Google Scholar]
  26. 26. 
    Li N, Bi YA, Duignan DB, Lai Y 2009. Quantitative expression profile of hepatobiliary transporters in sandwich cultured rat and human hepatocytes. Mol. Pharm. 6:41180–89
    [Google Scholar]
  27. 27. 
    Harwood MD, Neuhoff S, Carlson GL, Warhurst G, Rostami-Hodjegan A 2013. Absolute abundance and function of intestinal drug transporters: a prerequisite for fully mechanistic in vitro-in vivo extrapolation of oral drug absorption. Biopharm. Drug Dispos. 34:12–28
    [Google Scholar]
  28. 28. 
    Kumar V, Yin J, Billington S, Prasad B, Brown CDA et al. 2018. The importance of incorporating OCT2 plasma membrane expression and membrane potential in IVIVE of metformin renal secretory clearance. Drug Metab. Dispos. 46:101441–45
    [Google Scholar]
  29. 29. 
    Couto N, Al-Majdoub ZM, Achour B, Wright PC, Rostami-Hodjegan A, Barber J 2019. Quantification of proteins involved in drug metabolism and disposition in the human liver using label-free global proteomics. Mol. Pharm. 16:2632–47
    [Google Scholar]
  30. 30. 
    Sasaki K, Tachikawa M, Uchida Y, Hirano S, Kadowaki F et al. 2018. ATP-binding cassette transporter A subfamily 8 is a sinusoidal efflux transporter for cholesterol and taurocholate in mouse and human liver. Mol. Pharm. 15:2343–55
    [Google Scholar]
  31. 31. 
    Aretz I, Hardt C, Wittig I, Meierhofer D 2016. An impaired respiratory electron chain triggers down-regulation of the energy metabolism and de-ubiquitination of solute carrier amino acid transporters. Mol. Cell. Proteom. 15:51526–38
    [Google Scholar]
  32. 32. 
    Tsybovsky Y, Wang B, Quazi F, Molday RS, Palczewski K 2011. Posttranslational modifications of the photoreceptor-specific ABC transporter ABCA4. Biochemistry 50:326855–66
    [Google Scholar]
  33. 33. 
    Riddell N, Faou P, Crewther SG 2018. Short term optical defocus perturbs normal developmental shifts in retina/RPE protein abundance. BMC Dev. Biol. 18:118
    [Google Scholar]
  34. 34. 
    Pelkonen L, Sato K, Reinisalo M, Kidron H, Tachikawa M et al. 2017. LC-MS/MS based quantitation of ABC and SLC transporter proteins in plasma membranes of cultured primary human retinal pigment epithelium cells and immortalized ARPE19 cell line. Mol. Pharm. 14:3605–13
    [Google Scholar]
  35. 35. 
    Shawahna R, Uchida Y, Declèves X, Ohtsuki S, Yousif S et al. 2011. Transcriptomic and quantitative proteomic analysis of transporters and drug metabolizing enzymes in freshly isolated human brain microvessels. Mol. Pharm. 8:41332–41
    [Google Scholar]
  36. 36. 
    Al Feteisi H, Al-Majdoub ZM, Achour B, Couto N, Rostami-Hodjegan A, Barber J 2018. Identification and quantification of blood-brain barrier transporters in isolated rat brain microvessels. J. Neurochem. 146:6670–85
    [Google Scholar]
  37. 37. 
    Billington S, Salphati L, Hop C, Chu X, Evers R et al. 2019. Interindividual and regional variability in drug transporter abundance at the human blood-brain barrier measured by quantitative targeted proteomics. Clin. Pharmacol. Ther. 106:1228–37
    [Google Scholar]
  38. 38. 
    Al-Majdoub ZM, Al Feteisi H, Achour B, Warwood S, Neuhoff S et al. 2019. Proteomic quantification of human blood-brain barrier SLC and ABC transporters in healthy individuals and dementia patients. Mol. Pharm. 16:31220–33
    [Google Scholar]
  39. 39. 
    Drozdzik M, Busch D, Lapczuk J, Muller J, Ostrowski M et al. 2019. Protein abundance of clinically relevant drug transporters in the human liver and intestine: a comparative analysis in paired tissue specimens. Clin. Pharmacol. Ther. 105:51204–12
    [Google Scholar]
  40. 40. 
    Vildhede A, Kimoto E, Rodrigues AD, Varma MVS 2018. Quantification of hepatic organic anion transport proteins OAT2 and OAT7 in human liver tissue and primary hepatocytes. Mol. Pharm. 15:83227–35
    [Google Scholar]
  41. 41. 
    Ohtsuki S, Schaefer O, Kawakami H, Inoue T, Liehner S et al. 2012. Simultaneous absolute protein quantification of transporters, cytochromes P450, and UDP-glucuronosyltransferases as a novel approach for the characterization of individual human liver: comparison with mRNA levels and activities. Drug Metab. Dispos. 40:183–92
    [Google Scholar]
  42. 42. 
    Fallon JK, Smith PC, Xia CQ, Kim MS 2016. Quantification of four efflux drug transporters in liver and kidney across species using targeted quantitative proteomics by isotope dilution nanoLC-MS/MS. Pharm. Res. 33:92280–88
    [Google Scholar]
  43. 43. 
    Wegler C, Gaugaz FZ, Andersson TB, Wiśniewski JR, Busch D et al. 2017. Variability in mass spectrometry-based quantification of clinically relevant drug transporters and drug metabolizing enzymes. Mol. Pharm. 14:93142–51
    [Google Scholar]
  44. 44. 
    Prasad B, Johnson K, Billington S, Lee C, Chung GW et al. 2016. Abundance of drug transporters in the human kidney cortex as quantified by quantitative targeted proteomics. Drug Metab. Dispos. 44:121920–24
    [Google Scholar]
  45. 45. 
    Li CY, Hosey-Cojocari C, Basit A, Unadkat JD, Leeder JS Prasad B. 2019. Optimized renal transporter quantification by using aquaporin 1 and aquaporin 2 as anatomical markers: application in characterizing the ontogeny of renal transporters and its correlation with hepatic transporters in paired human samples. Clin. Pharmacol. Ther 21:88
    [Google Scholar]
  46. 46. 
    Cheung KWK, van Groen BD, Spaans E, van Borselon MD, de Bruijn ACJM et al. 2019. A comprehensive analysis of ontogeny of renal drug transporters: mRNA analyses, quantitative proteomics, and localization. Clin. Pharmacol. Ther. 106:51083–92
    [Google Scholar]
  47. 47. 
    Fallon JK, Houvig N, Booth-Genthe CL, Smith PC 2018. Quantification of membrane transporter proteins in human lung and immortalized cell lines using targeted quantitative proteomic analysis by isotope dilution nanoLC-MS/MS. J. Pharm. Biomed. Anal. 154:150–57
    [Google Scholar]
  48. 48. 
    Gröer C, Brück S, Lai Y, Paulick A, Busemann A et al. 2013. LC-MS/MS-based quantification of clinically relevant intestinal uptake and efflux transporter proteins. J. Pharm. Biomed. Anal. 85:253–61
    [Google Scholar]
  49. 49. 
    Liao MZ, Gao C, Bhatt DK, Prasad B, Mao Q 2018. Quantitative proteomics reveals changes in transporter protein abundance in liver, kidney and brain of mice by pregnancy. Drug Metab. Lett. 12:2145–52
    [Google Scholar]
  50. 50. 
    Braun C, Sakamoto A, Fuchs H, Ishiguro N, Suzuki S et al. 2017. Quantification of transporter and receptor proteins in dog brain capillaries and choroid plexus: relevance for the distribution in brain and CSF of selected BCRP and P-gp substrates. Mol. Pharm. 14:103436–47
    [Google Scholar]
  51. 51. 
    Wang L, Prasad B, Salphati L, Chu X, Gupta A et al. 2015. Interspecies variability in expression of hepatobiliary transporters across human, dog, monkey, and rat as determined by quantitative proteomics. Drug Metab. Dispos. 43:3367–74
    [Google Scholar]
  52. 52. 
    Malinen MM, Ito K, Kang HE, Honkakoski P, Brouwer KLR 2019. Protein expression and function of organic anion transporters in short-term and long-term cultures of Huh7 human hepatoma cells. Eur. J. Pharm. Sci. 130:186–95
    [Google Scholar]
  53. 53. 
    Li N, Palandra J, Nemirovskiy OV, Lai Y 2009. LC-MS/MS mediated absolute quantification and comparison of bile salt export pump and breast cancer resistance protein in livers and hepatocytes across species. Anal. Chem. 81:62251–59
    [Google Scholar]
  54. 54. 
    Li N, Singh P, Mandrell KM, Lai Y 2010. Improved extrapolation of hepatobiliary clearance from in vitro sandwich cultured rat hepatocytes through absolute quantification of hepatobiliary transporters. Mol. Pharm. 7:3630–41
    [Google Scholar]
  55. 55. 
    Jiang L, Meng F, Qiu Z, Zhang K, Ding Y et al. 2019. Comparison of UPLC-MS/MS-based targeted quantitation and conventional quantitative methods for the analysis of MRP1 expression in tumor cell lines. J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 1109:10–18
    [Google Scholar]
  56. 56. 
    Pan Q, Zhang X, Zhang L, Cheng Y, Zhao N et al. 2018. Solute carrier organic anion transporter family member 3a1 is a bile acid efflux transporter in cholestasis. Gastroenterology 155:51578–92.e1516
    [Google Scholar]
  57. 57. 
    Schroll MM, Liu X, Herzog SK, Skube SB, Hummon AB 2016. Nutrient restriction of glucose or serum results in similar proteomic expression changes in 3D colon cancer cell cultures. Nutr. Res. 36:101068–80
    [Google Scholar]
  58. 58. 
    Shi P, Liao M, Chuang BC, Griffin R, Shi J et al. 2018. Efflux transporter breast cancer resistance protein dominantly expresses on the membrane of red blood cells, hinders partitioning of its substrates into the cells, and alters drug-drug interaction profiles. Xenobiotica 48:111173–83
    [Google Scholar]
  59. 59. 
    Vildhede A, Nguyen C, Erickson BK, Kunz RC, Jones R et al. 2018. Comparison of proteomic quantification approaches for hepatic drug transporters: multiplexed global quantitation correlates with targeted proteomic quantitation. Drug Metab. Dispos. 46:5692–96
    [Google Scholar]
  60. 60. 
    Drozdzik M, Gröer C, Penski J, Lapczuk J, Ostrowski M et al. 2014. Protein abundance of clinically relevant multidrug transporters along the entire length of the human intestine. Mol. Pharm. 11:103547–55
    [Google Scholar]
  61. 61. 
    Hoshi Y, Uchida Y, Tachikawa M, Inoue T, Ohtsuki S, Terasaki T 2013. Quantitative atlas of blood-brain barrier transporters, receptors, and tight junction proteins in rats and common marmoset. J. Pharm. Sci. 102:93343–55
    [Google Scholar]
  62. 62. 
    Uchida Y, Tachikawa M, Obuchi W, Hoshi Y, Tomioka Y et al. 2013. A study protocol for quantitative targeted absolute proteomics (QTAP) by LC-MS/MS: application for inter-strain differences in protein expression levels of transporters, receptors, claudin-5, and marker proteins at the blood-brain barrier in ddY, FVB, and C57BL/6J mice. Fluids Barriers CNS 10:121
    [Google Scholar]
  63. 63. 
    Ohtsuki S, Ikeda C, Uchida Y, Sakamoto Y, Miller F et al. 2013. Quantitative targeted absolute proteomic analysis of transporters, receptors and junction proteins for validation of human cerebral micro-vascular endothelial cell line HCMEC/D3 as a human blood-brain barrier model. Mol. Pharm. 10:1289–96
    [Google Scholar]
  64. 64. 
    Nirmalan NJ, Hughes C, Peng J, McKenna T, Langridge J et al. 2011. Initial development and validation of a novel extraction method for quantitative mining of the formalin-fixed, paraffin-embedded tissue proteome for biomarker investigations. J. Proteome Res. 10:2896–906
    [Google Scholar]
  65. 65. 
    Whitelegge JP. 2013. Integral membrane proteins and bilayer proteomics. Anal. Chem. 85:52558–68
    [Google Scholar]
  66. 66. 
    Seigneurin-Berny D, Rolland N, Garin J, Joyard J 1999. Technical advance: differential extraction of hydrophobic proteins from chloroplast envelope membranes: a subcellular-specific proteomic approach to identify rare intrinsic membrane proteins. Plant J 19:2217–28
    [Google Scholar]
  67. 67. 
    Whitelegge J, Halgand F, Souda P, Zabrouskov V 2006. Top-down mass spectrometry of integral membrane proteins. Expert Rev. Proteom. 3:6585–96
    [Google Scholar]
  68. 68. 
    Vuckovic D, Dagley LF, Purcell AW, Emili A 2013. Membrane proteomics by high performance liquid chromatography-tandem mass spectrometry: analytical approaches and challenges. Proteomics 13:3–4404–23
    [Google Scholar]
  69. 69. 
    Yu YQ, Gilar M, Lee PJ, Bouvier ES, Gebler JC 2003. Enzyme-friendly, mass spectrometry-compatible surfactant for in-solution enzymatic digestion of proteins. Anal. Chem. 75:216023–28
    [Google Scholar]
  70. 70. 
    Feist P, Hummon AB. 2015. Proteomic challenges: sample preparation techniques for microgram-quantity protein analysis from biological samples. Int. J. Mol. Sci. 16:23537–63
    [Google Scholar]
  71. 71. 
    Mbeunkui F, Goshe MB. 2011. Investigation of solubilization and digestion methods for microsomal membrane proteome analysis using data-independent LC-MSE. Proteomics 11:5898–911
    [Google Scholar]
  72. 72. 
    Botelho D, Wall MJ, Vieira DB, Fitzsimmons S, Liu F, Doucette A 2010. Top-down and bottom-up proteomics of SDS-containing solutions following mass-based separation. J. Proteome Res. 9:62863–70
    [Google Scholar]
  73. 73. 
    Wiśniewski JR, Zougman A, Nagaraj N, Mann M 2009. Universal sample preparation method for proteome analysis. Nat. Methods 6:5359–62
    [Google Scholar]
  74. 74. 
    Bryk AH, Wiśniewski JR. 2017. Quantitative analysis of human red blood cell proteome. J. Proteome Res. 16:82752–61
    [Google Scholar]
  75. 75. 
    Fischer R, Kessler BM. 2015. Gel-aided sample preparation (GASP)–a simplified method for gel-assisted proteomic sample generation from protein extracts and intact cells. Proteomics 15:71224–29
    [Google Scholar]
  76. 76. 
    Pirmoradian M, Budamgunta H, Chingin K, Zhang B, Astorga-Wells J, Zubarev RA 2013. Rapid and deep human proteome analysis by single-dimension shotgun proteomics. Mol. Cell. Proteom. 12:113330–38
    [Google Scholar]
  77. 77. 
    Waas M, Bhattacharya S, Chuppa S, Wu X, Jensen DR et al. 2014. Combine and conquer: surfactants, solvents, and chaotropes for robust mass spectrometry based analyses of membrane proteins. Anal. Chem. 86:31551–59
    [Google Scholar]
  78. 78. 
    Zhao Q, Liang Y, Yuan H, Sui Z, Wu Q et al. 2013. Biphasic microreactor for efficient membrane protein pretreatment with a combination of formic acid assisted solubilization, on-column pH adjustment, reduction, alkylation, and tryptic digestion. Anal. Chem. 85:188507–12
    [Google Scholar]
  79. 79. 
    Doucette AA, Vieira DB, Orton DJ, Wall MJ 2014. Resolubilization of precipitated intact membrane proteins with cold formic acid for analysis by mass spectrometry. J. Proteome Res. 13:126001–12
    [Google Scholar]
  80. 80. 
    Couto N, Barber J, Gaskell SJ 2011. Matrix-assisted laser desorption/ionisation mass spectrometric response factors of peptides generated using different proteolytic enzymes. J. Mass Spectrom. 46:121233–40
    [Google Scholar]
  81. 81. 
    Kozlowski LP. 2017. Proteome-pI: proteome isoelectric point database. Nucleic Acids. Res. 45:D1D1112–16
    [Google Scholar]
  82. 82. 
    Le Maire M, Champeil P, Moller JV 2000. Interaction of membrane proteins and lipids with solubilizing detergents. Biochim. Biophys. Acta 1508:1–286–111
    [Google Scholar]
  83. 83. 
    Molinari M. 2007. N-glycan structure dictates extension of protein folding or onset of disposal. Nat. Chem. Biol. 3:6313–20
    [Google Scholar]
  84. 84. 
    Al-Majdoub ZM, Carroll KM, Gaskell SJ, Barber J 2014. Quantification of the proteins of the bacterial ribosome using QconCAT technology. J. Proteome Res. 13:31211–22
    [Google Scholar]
  85. 85. 
    Dormeyer W, van Hoof D, Mummery CL, Krijgsveld J, Heck AJ 2008. A practical guide for the identification of membrane and plasma membrane proteins in human embryonic stem cells and human embryonal carcinoma cells. Proteomics 8:194036–53
    [Google Scholar]
  86. 86. 
    Bhatt DK, Prasad B. 2018. Critical issues and optimized practices in quantification of protein abundance level to determine interindividual variability in DMET proteins by LC-MS/MS proteomics. Clin. Pharmacol. Ther. 103:4619–30
    [Google Scholar]
  87. 87. 
    Shen X, Shen S, Li J, Hu Q, Nie L et al. 2018. IonStar enables high-precision, low-missing-data proteomics quantification in large biological cohorts. PNAS 115:21E4767–76
    [Google Scholar]
  88. 88. 
    Couvillion SP, Zhu Y, Nagy G, Adkins JN, Ansong C et al. 2019. New mass spectrometry technologies contributing towards comprehensive and high throughput omics analyses of single cells. Analyst 144:3794–807
    [Google Scholar]
  89. 89. 
    Wilm M, Mann M. 1996. Analytical properties of the nanoelectrospray ion source. Anal. Chem. 68:11–8
    [Google Scholar]
  90. 90. 
    Marginean I, Tang K, Smith RD, Kelly RT 2014. Picoelectrospray ionization mass spectrometry using narrow-bore chemically etched emitters. J. Am. Soc. Mass Spectrom. 25:130–36
    [Google Scholar]
  91. 91. 
    Sun L, Zhu G, Zhao Y, Yan X, Mou S, Dovichi NJ 2013. Ultrasensitive and fast bottom-up analysis of femtogram amounts of complex proteome digests. Angew. Chem. Int. Ed. 52:5113661–64
    [Google Scholar]
  92. 92. 
    Harbourt DE, Fallon JK, Ito S, Baba T, Ritter JK et al. 2012. Quantification of human uridine-diphosphate glucuronosyl transferase 1A isoforms in liver, intestine, and kidney using nanobore liquid chromatography-tandem mass spectrometry. Anal. Chem. 84:198–105
    [Google Scholar]
  93. 93. 
    Li S, Plouffe BD, Belov AM, Ray S, Wang X et al. 2015. An integrated platform for isolation, processing, and mass spectrometry-based proteomic profiling of rare cells in whole blood. Mol. Cell. Proteom. 14:61672–83
    [Google Scholar]
  94. 94. 
    Luo Q, Yue G, Valaskovic GA, Gu Y, Wu SL, Karger BL 2007. On-line 1D and 2D porous layer open tubular/LC-ESI-MS using 10-μm-i.d. poly(styrene-divinylbenzene) columns for ultrasensitive proteomic analysis. Anal. Chem. 79:166174–81
    [Google Scholar]
  95. 95. 
    Lanucara F, Holman SW, Gray CJ, Eyers CE 2014. The power of ion mobility-mass spectrometry for structural characterization and the study of conformational dynamics. Nat. Chem. 6:4281–94
    [Google Scholar]
  96. 96. 
    Parson WB, Schneider BB, Kertesz V, Corr JJ, Covey TR, Van Berkel GJ 2011. Rapid analysis of isomeric exogenous metabolites by differential mobility spectrometry-mass spectrometry. Rapid. Commun. Mass Spectrom. 25:223382–86
    [Google Scholar]
  97. 97. 
    Gabelica V, Marklund E. 2018. Fundamentals of ion mobility spectrometry. Curr. Opin. Chem. Biol. 42:51–59
    [Google Scholar]
  98. 98. 
    Creese AJ, Cooper HJ. 2012. Separation and identification of isomeric glycopeptides by high field asymmetric waveform ion mobility spectrometry. Anal. Chem. 84:52597–601
    [Google Scholar]
  99. 99. 
    Morris CB, May JC, Leaptrot KL, McLean JA 2019. Evaluating separation selectivity and collision cross section measurement reproducibility in helium, nitrogen, argon, and carbon dioxide drift gases for drift tube ion mobility-mass spectrometry. J. Am. Soc. Mass Spectrom. 30:61059–68
    [Google Scholar]
  100. 100. 
    Fernandez-Maestre R, Wu C, Hill HH 2015. Separation of asparagine, valine and tetraethylammonium ions overlapping in an ion mobility spectrum by clustering with methanol introduced as a modifier into the buffer gas. Anal. Methods 7:3863–69
    [Google Scholar]
  101. 101. 
    Bridon G, Bonneil E, Muratore-Schroeder T, Caron-Lizotte O, Thibault P 2012. Improvement of phosphoproteome analyses using FAIMS and decision tree fragmentation. application to the insulin signaling pathway in Drosophila melanogaster S2 cells. J. Proteome Res. 11:2927–40
    [Google Scholar]
  102. 102. 
    Shliaha PV, Bond NJ, Gatto L, Lilley KS 2013. Effects of traveling wave ion mobility separation on data independent acquisition in proteomics studies. J. Proteome Res. 12:62323–39
    [Google Scholar]
  103. 103. 
    MacLean BX, Pratt BS, Egertson JD, MacCoss MJ, Smith RD, Baker ES 2018. Using skyline to analyze data-containing liquid chromatography, ion mobility spectrometry, and mass spectrometry dimensions. J. Am. Soc. Mass Spectrom. 29:112182–88
    [Google Scholar]
  104. 104. 
    Gethings LA, Connolly JB. 2014. Simplifying the proteome: analytical strategies for improving peak capacity. Adv. Exp. Med. Biol. 806:59–77
    [Google Scholar]
  105. 105. 
    Achour B, Al Feteisi H, Lanucara F, Rostami-Hodjegan A, Barber J 2017. Global proteomic analysis of human liver microsomes: rapid characterization and quantification of hepatic drug-metabolizing enzymes. Drug Metab. Dispos. 45:6666–75
    [Google Scholar]
  106. 106. 
    Fournier ML, Gilmore JM, Martin-Brown SA, Washburn MP 2007. Multidimensional separations-based shotgun proteomics. Chem. Rev. 107:83654–86
    [Google Scholar]
  107. 107. 
    Perkins DN, Pappin DJ, Creasy DM, Cottrell JS 1999. Probability-based protein identification by searching sequence databases using mass spectrometry data. Electrophoresis 20:183551–67
    [Google Scholar]
  108. 108. 
    Eng JK, McCormack AL, Yates JR 1994. An approach to correlate tandem mass spectral data of peptides with amino acid sequences in a protein database. J. Am. Soc. Mass Spectrom. 5:11976–89
    [Google Scholar]
  109. 109. 
    Craig R, Beavis RC. 2003. A method for reducing the time required to match protein sequences with tandem mass spectra. Rapid. Commun. Mass Spectrom. 17:202310–16
    [Google Scholar]
  110. 110. 
    El-Khateeb E, Vasilogianni AM, Alrubia S, Al-Majdoub ZM, Couto N et al. 2019. Quantitative mass spectrometry-based proteomics in the era of model-informed drug development: applications in translational pharmacology and recommendations for best practice. Pharmacol. Ther. 203:107397
    [Google Scholar]
  111. 111. 
    Carr SA, Abbatiello SE, Ackermann BL, Borchers C, Domon B et al. 2014. Targeted peptide measurements in biology and medicine: best practices for mass spectrometry-based assay development using a fit-for-purpose approach. Mol. Cell. Proteom. 13:3907–17
    [Google Scholar]
  112. 112. 
    Kamiie J, Ohtsuki S, Iwase R, Ohmine K, Katsukura Y et al. 2008. Quantitative atlas of membrane transporter proteins: development and application of a highly sensitive simultaneous LC/MS/MS method combined with novel in-silico peptide selection criteria. Pharm. Res. 25:61469–83
    [Google Scholar]
  113. 113. 
    Uchida Y, Ohtsuki S, Katsukura Y, Ikeda C, Suzuki T et al. 2011. Quantitative targeted absolute proteomics of human blood-brain barrier transporters and receptors. J. Neurochem. 117:2333–45
    [Google Scholar]
  114. 114. 
    Niessen J, Jedlitschky G, Grube M, Bien S, Schwertz H et al. 2009. Human platelets express organic anion-transporting peptide 2B1, an uptake transporter for atorvastatin. Drug Metab. Dispos. 37:51129–37
    [Google Scholar]
  115. 115. 
    Niessen J, Jedlitschky G, Grube M, Kawakami H, Kamiie J et al. 2010. Expression of ABC-type transport proteins in human platelets. Pharmacogenet. Genom. 20:6396–400
    [Google Scholar]
  116. 116. 
    Picotti P, Bodenmiller B, Mueller LN, Domon B, Aebersold R 2009. Full dynamic range proteome analysis of S. cerevisiae by targeted proteomics. Cell 138:4795–806
    [Google Scholar]
  117. 117. 
    Oswald S, Gröer C, Drozdzik M, Siegmund W 2013. Mass spectrometry-based targeted proteomics as a tool to elucidate the expression and function of intestinal drug transporters. AAPS J 15:41128–40
    [Google Scholar]
  118. 118. 
    Prasad B, Evers R, Gupta A, Hop CE, Salphati L et al. 2014. Interindividual variability in hepatic organic anion-transporting polypeptides and P-glycoprotein (ABCB1) protein expression: quantification by liquid chromatography tandem mass spectroscopy and influence of genotype, age, and sex. Drug Metab. Dispos. 42:178–88
    [Google Scholar]
  119. 119. 
    Harwood MD, Achour B, Russell MR, Carlson GL, Warhurst G, Rostami-Hodjegan A 2015. Application of an LC-MS/MS method for the simultaneous quantification of human intestinal transporter proteins absolute abundance using a QconCAT technique. J. Pharm. Biomed. Anal. 110:27–33
    [Google Scholar]
  120. 120. 
    Nat. Methods 2013. Method of the year 2012. Nat. Methods 10:1
    [Google Scholar]
  121. 121. 
    Abbatiello S, Ackermann BL, Borchers C, Bradshaw RA, Carr SA et al. 2017. New guidelines for publication of manuscripts describing development and application of targeted mass spectrometry measurements of peptides and proteins. Mol. Cell. Proteom. 16:3327–28
    [Google Scholar]
  122. 122. 
    Whiteaker JR, Halusa GN, Hoofnagle AN, Sharma V, MacLean B et al. 2014. CPTAC Assay Portal: a repository of targeted proteomic assays. Nat. Methods 11:7703–4
    [Google Scholar]
  123. 123. 
    Abbatiello SE, Schilling B, Mani DR, Zimmerman LJ, Hall SC et al. 2015. Large-scale interlaboratory study to develop, analytically validate and apply highly multiplexed, quantitative peptide assays to measure cancer-relevant proteins in plasma. Mol. Cell. Proteom. 14:92357–74
    [Google Scholar]
  124. 124. 
    Vrana M, Whittington D, Nautiyal V, Prasad B 2017. Database of optimized proteomic quantitative methods for human drug disposition-related proteins for applications in physiologically based pharmacokinetic modeling. CPT Pharmacomet. Syst. Pharmacol. 6:4267–76
    [Google Scholar]
  125. 125. 
    Ladumor MK, Thakur A, Sharma S, Rachapally A, Mishra S et al. 2019. A repository of protein abundance data of drug metabolizing enzymes and transporters for applications in physiologically based pharmacokinetic (PBPK) modelling and simulation. Sci. Rep. 9:19709
    [Google Scholar]
  126. 126. 
    Kusebauch U, Campbell DS, Deutsch EW, Chu CS, Spicer DA et al. 2016. Human SRMAtlas: a resource of targeted assays to quantify the complete human proteome. Cell 166:3766–78
    [Google Scholar]
  127. 127. 
    US Dep. Health Hum. Serv., Cent. Drug Eval. Res., Cent. Vet. Med 2018. Bioanalytical method validation: guidance for industry Rep., May, US Food Drug Admin Silver Spring, MD: https://www.fda.gov/files/drugs/published/Bioanalytical-Method-Validation-Guidance-for-Industry.pdf
  128. 128. 
    Eur. Med. Agency 2005. ICH Topic Q 2 (R1). Validation of analytical procedures: text and methodology Rep. CPMP/ICH/381/95, June, Eur. Med. Agency London: https://www.ema.europa.eu/en/documents/scientific-guideline/ich-q-2-r1-validation-analytical-procedures-text-methodology-step-5_en.pdf
  129. 129. 
    Al Feteisi H, Achour B, Barber J, Rostami-Hodjegan A 2015. Choice of LC-MS methods for the absolute quantification of drug-metabolizing enzymes and transporters in human tissue: a comparative cost analysis. AAPS J 17:2438–46
    [Google Scholar]
  130. 130. 
    Russell MR, Achour B, McKenzie EA, Lopez R, Harwood MD et al. 2013. Alternative fusion protein strategies to express recalcitrant QconCAT proteins for quantitative proteomics of human drug metabolizing enzymes and transporters. J. Proteome Res. 12:125934–42
    [Google Scholar]
  131. 131. 
    Gallien S, Duriez E, Crone C, Kellmann M, Moehring T, Domon B 2012. Targeted proteomic quantification on quadrupole-Orbitrap mass spectrometer. Mol. Cell. Proteom. 11:121709–23
    [Google Scholar]
  132. 132. 
    Peterson AC, Russell JD, Bailey DJ, Westphall MS, Coon JJ 2012. Parallel reaction monitoring for high resolution and high mass accuracy quantitative, targeted proteomics. Mol. Cell. Proteom. 11:111475–88
    [Google Scholar]
  133. 133. 
    Schilling B, MacLean B, Held JM, Sahu AK, Rardin MJ et al. 2015. Multiplexed, scheduled, high-resolution parallel reaction monitoring on a full scan QqTOF instrument with integrated data-dependent and targeted mass spectrometric workflows. Anal. Chem. 87:2010222–29
    [Google Scholar]
  134. 134. 
    Nakamura K, Hirayama-Kurogi M, Ito S, Kuno T, Yoneyama T et al. 2016. Large-scale multiplex absolute protein quantification of drug-metabolizing enzymes and transporters in human intestine, liver, and kidney microsomes by SWATH-MS: comparison with MRM/SRM and HR-MRM/PRM. Proteomics 16:15–162106–17
    [Google Scholar]
  135. 135. 
    Hu A, Noble WS, Wolf-Yadlin A 2016. Technical advances in proteomics: new developments in data-independent acquisition. F1000Research 2016:5
    [Google Scholar]
  136. 136. 
    Michalski A, Cox J, Mann M 2011. More than 100,000 detectable peptide species elute in single shotgun proteomics runs but the majority is inaccessible to data-dependent LC-MS/MS. J. Proteome Res. 10:41785–93
    [Google Scholar]
  137. 137. 
    Ong SE, Blagoev B, Kratchmarova I, Kristensen DB, Steen H et al. 2002. Stable isotope labeling by amino acids in cell culture, SILAC, as a simple and accurate approach to expression proteomics. Mol. Cell. Proteom. 1:5376–86
    [Google Scholar]
  138. 138. 
    Thompson A, Schafer J, Kuhn K, Kienle S, Schwarz J et al. 2003. Tandem mass tags: a novel quantification strategy for comparative analysis of complex protein mixtures by MS/MS. Anal. Chem. 75:81895–904
    [Google Scholar]
  139. 139. 
    Prasad B, Unadkat JD. 2014. Comparison of heavy labeled (SIL) peptide versus SILAC protein internal standards for LC-MS/MS quantification of hepatic drug transporters. Int. J. Proteom. 2014:451510
    [Google Scholar]
  140. 140. 
    Cox J, Mann M. 2008. MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification. Nat. Biotechnol. 26:121367–72
    [Google Scholar]
  141. 141. 
    Ludwig C, Gillet L, Rosenberger G, Amon S, Collins BC, Aebersold R 2018. Data-independent acquisition-based SWATH-MS for quantitative proteomics: a tutorial. Mol. Syst. Biol. 14:8e8126
    [Google Scholar]
  142. 142. 
    Bilbao A, Varesio E, Luban J, Strambio-De-Castillia C, Hopfgartner G et al. 2015. Processing strategies and software solutions for data-independent acquisition in mass spectrometry. Proteomics 15:5–6964–80
    [Google Scholar]
  143. 143. 
    Gillet LC, Navarro P, Tate S, Röst H, Selevsek N et al. 2012. Targeted data extraction of the MS/MS spectra generated by data-independent acquisition: a new concept for consistent and accurate proteome analysis. Mol. Cell. Proteom. 11:6O111.016717
    [Google Scholar]
  144. 144. 
    Gillet LC, Leitner A, Aebersold R 2016. Mass spectrometry applied to bottom-up proteomics: entering the high-throughput era for hypothesis testing. Annu. Rev. Anal. Chem. 9:449–72
    [Google Scholar]
  145. 145. 
    Rőst HL, Rosenberger G, Navarro P, Gillet L, Miladinović SM et al. 2014. OpenSWATH enables automated, targeted analysis of data-independent acquisition MS data. Nat. Biotechnol. 32:3219–23
    [Google Scholar]
  146. 146. 
    Keller A, Bader SL, Kusebauch U, Shteynberg D, Hood L, Moritz RL 2016. Opening a SWATH window on posttranslational modifications: automated pursuit of modified peptides. Mol. Cell Proteom. 15:31151–63
    [Google Scholar]
  147. 147. 
    Guo T, Kouvonen P, Koh CC, Gillet LC, Wolski WE et al. 2015. Rapid mass spectrometric conversion of tissue biopsy samples into permanent quantitative digital proteome maps. Nat. Med. 21:4407–13
    [Google Scholar]
  148. 148. 
    Jamwal R, Barlock BJ, Adusumalli S, Ogasawara K, Simons BL, Akhlaghi F 2017. Multiplex and label-free relative quantification approach for studying protein abundance of drug metabolizing enzymes in human liver microsomes using SWATH-MS. J. Proteome Res. 16:114134–43
    [Google Scholar]
  149. 149. 
    Howard M, Achour B, Al‐Majdoub Z, Rostami‐Hodjegan A, Barber J 2018. GASP and FASP are complementary for LC-MS/MS proteomics analysis of drug-metabolizing enzymes and transporters in pig liver. Proteomics 18:24e1800200
    [Google Scholar]
  150. 150. 
    Golghalyani V, Neupartl M, Wittig I, Bahr U, Karas M 2017. ArgC-like digestion: Complementary or alternative to tryptic digestion?. J. Proteome Res. 16:2978–87
    [Google Scholar]
  151. 151. 
    Wiśniewski JR, Wegler C, Artursson P 2019. Multiple-enzyme-digestion strategy improves accuracy and sensitivity of label- and standard-free absolute quantification to a level that is achievable by analysis with stable isotope-labeled standard spiking. J. Proteome Res. 18:1217–24
    [Google Scholar]
/content/journals/10.1146/annurev-anchem-091719-024553
Loading
/content/journals/10.1146/annurev-anchem-091719-024553
Loading

Data & Media loading...

  • Article Type: Review Article
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error