Korean J Lab Med.  2011 Apr;31(2):61-71. 10.3343/kjlm.2011.31.2.61.

The Path to Clinical Proteomics Research: Integration of Proteomics, Genomics, Clinical Laboratory and Regulatory Science

Affiliations
  • 1Office of Cancer Clinical Proteomics Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA. rodriguezh@mail.nih.gov

Abstract

Better biomarkers are urgently needed to cancer detection, diagnosis, and prognosis. While the genomics community is making significant advances in understanding the molecular basis of disease, proteomics will delineate the functional units of a cell, proteins and their intricate interaction network and signaling pathways for the underlying disease. Great progress has been made to characterize thousands of proteins qualitatively and quantitatively in complex biological systems by utilizing multi-dimensional sample fractionation strategies, mass spectrometry and protein microarrays. Comparative/quantitative analysis of high-quality clinical biospecimen (e.g., tissue and biofluids) of human cancer proteome landscape has the potential to reveal protein/peptide biomarkers responsible for this disease by means of their altered levels of expression, post-translational modifications as well as different forms of protein variants. Despite technological advances in proteomics, major hurdles still exist in every step of the biomarker development pipeline. The National Cancer Institute's Clinical Proteomic Technologies for Cancer initiative (NCI-CPTC) has taken a critical step to close the gap between biomarker discovery and qualification by introducing a pre-clinical "verification" stage in the pipeline, partnering with clinical laboratory organizations to develop and implement common standards, and developing regulatory science documents with the US Food and Drug Administration to educate the proteomics community on analytical evaluation requirements for multiplex assays in order to ensure the safety and effectiveness of these tests for their intended use.

Keyword

Quantitative proteomics; Biomarker; Multiplex protein assays; MRM-MS; Immunoassays

MeSH Terms

Biological Markers/analysis
Clinical Laboratory Techniques/standards
*Genomics
Humans
Mass Spectrometry/methods/standards
Neoplasms/*diagnosis/genetics
*Proteomics
Quality Control
United States
United States Food and Drug Administration

Figure

  • Fig. 1 Barriers between candidate biomarker discovery by proteomics and qualification.

  • Fig. 2 The envisioned National Cancer Institute-Clinical Proteomic Technologies for Cancer initiative (NCI-CPTC) development pipeline from discovery to qualification. (A) The gap in the current proteomics research pipeline. (B) The incorporation of verification into the NCI-CPTC pipeline between discovery and qualification.

  • Fig. 3 Multiple Reaction Monitoring Mass Spectrometry (MRM-MS). A schematic of a triple quadrupole mass spectrometer (QQQ-MS) commonly used in MRM-MS analysis: Q1 and Q3 represent two mass filters for precursor and fragment ion selection while Q2 (collision cell) creates fragment ions via collisionally-induced dissociation (CID). In this case, one of the three peptide precursor ions (colored in blue) is selected in Q1, fragmented in Q2 and quantitated using one of its fragment ions (transition) selected in Q3 by the relative intensity of its peak area. An MRM-MS assay offers multiplexing capability of many target analytes in a single HPLC run.

  • Fig. 4 Regulatory science education through the development of public documents that enable a more efficient flow through biomarker discovery, development and regulatory processes.

  • Fig. 5 National Cancer Institute-Clinical Proteomic Technologies for Cancer initiative proposed proteomics development and implementation process.


Cited by  1 articles

Proteomic Profiling of Serum from Patients with Tuberculosis
Sang Hoon Song, Minje Han, Yang Seon Choi, Ki Soon Dan, Man Gil Yang, Junghan Song, Sung Sup Park, Jae Ho Lee
Ann Lab Med. 2014;34(5):345-353.    doi: 10.3343/alm.2014.34.5.345.


Reference

1. Etzioni R, Urban N, Ramsey S, McIntosh M, Schwartz S, Reid B, et al. The case for early detection. Nat Rev Cancer. 2003; 3:243–252. PMID: 12671663.
Article
2. Anderson L. Candidate-based proteomics in the search for biomarkers of cardiovascular disease. J Physiol. 2005; 563:23–60. PMID: 15611012.
Article
3. Rifai N, Gillette MA, Carr SA. Protein biomarker discovery and validation: the long and uncertain path to clinical utility. Nat Biotechnol. 2006; 24:971–983. PMID: 16900146.
Article
4. García-Foncillas J, Bandrés E, Zárate R, Remírez N. Proteomic analysis in cancer research: potential application in clinical use. Clin Transl Oncol. 2006; 8:250–261. PMID: 16648100.
Article
5. Bouchal P, Roumeliotis T, Hrstka R, Nenutil R, Vojtesek B, Garbis SD. Biomarker discovery in low-grade breast cancer using isobaric stable isotope tags and two-dimensional liquid chromatography-tandem mass spectrometry (iTRAQ-2DLC-MS/MS) based quantitative proteomic analysis. J Proteome Res. 2009; 8:362–373. PMID: 19053527.
Article
6. Wiener MC, Sachs JR, Deyanova EG, Yates NA. Differential mass spectrometry: a label-free LC-MS method for finding significant differences in complex peptide and protein mixtures. Anal Chem. 2004; 76:6085–6096. PMID: 15481957.
Article
7. Geiger T, Cox J, Ostasiewicz P, Wisniewski JR, Mann M. Super-SILAC mix for quantitative proteomics of human tumor tissue. Nat Methods. 2010; 7:383–385. PMID: 20364148.
Article
8. Anderson L, Hunter CL. Quantitative mass spectrometric multiple reaction monitoring assays for major plasma proteins. Mol Cell Proteomics. 2006; 5:573–588. PMID: 16332733.
Article
9. Wang H, Wong CH, Chin A, Kennedy J, Zhang Q, Hanash S. Quantitative serum proteomics using dual stable isotope coding and nano LC-MS/MSMS. J Proteome Res. 2009; 8:5412–5422. PMID: 19817497.
Article
10. Lee J, Soper SA, Murray KK. Microfluidic chips for mass spectrometry-based proteomics. J Mass Spectrom. 2009; 44:579–593. PMID: 19373851.
Article
11. Pierobon M, Calvert V, Belluco C, Garaci E, Deng J, Lise M, et al. Multiplexed cell signaling analysis of metastatic and nonmetastatic colorectal cancer reveals COX2-EGFR signaling activation as a potential prognostic pathway biomarker. Clin Colorectal Cancer. 2009; 8:110–117.
Article
12. Ramachandran N, Raphael JV, Hainsworth E, Demirkan G, Fuentes MG, Rolfs A, et al. Next-generation high-density self-assembling functional protein arrays. Nat Methods. 2008; 5:535–538. PMID: 18469824.
Article
13. Beirne P, Pantelidis P, Charles P, Wells AU, Abraham DJ, Denton CP, et al. Multiplex immune serum biomarker profiling in sarcoidosis and systemic sclerosis. Eur Respir J. 2009; 34:1376–1382. PMID: 19541722.
Article
14. Kelleher MT, Fruhwirth G, Patel G, Ofo E, Festy F, Barber PR, et al. The potential of optical proteomic technologies to individualize prognosis and guide rational treatment for cancer patients. Target Oncol. 2009; 4:235–252. PMID: 19756916.
Article
15. Wang P, Whiteaker JR, Paulovich AG. The evolving role of mass spectrometry in cancer biomarker discovery. Cancer Biol Ther. 2009; 8:1083–1094. PMID: 19502776.
Article
16. Whiteaker JR, Zhang H, Eng JK, Fang R, Piening BD, Feng LC, et al. Head-to-head comparison of serum fractionation techniques. J Proteome Res. 2007; 6:828–836. PMID: 17269739.
Article
17. Ernoult E, Bourreau A, Gamelin E, Guette C. A proteomic approach for plasma biomarker discovery with iTRAQ labeling and OFFGEL fractionation. J Biomed Biotechnol. 2010; 2010:927917. PMID: 19888438.
18. Nirmalan NJ, Hughes C, Peng J, McKenna T, Langridge J, Cairns DA, et al. 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. 2010; 10:896–906. PMID: 21117664.
Article
19. Krishhan VV, Khan IH, Luciw PA. Multiplexed microbead immunoassays by flow cytometry for molecular profiling: basic concepts and proteomics applications. Crit Rev Biotechnol. 2009; 29:29–43. PMID: 19514901.
Article
20. Cha S, Imielinski MB, Rejtar T, Richardson EA, Thakur D, Sgroi DC, et al. In situ proteomic analysis of human breast cancer epithelial cells using laser capture microdissection: annotation by protein set enrichment analysis and gene ontology. Mol Cell Proteomics. 2010; 9:2529–2544. PMID: 20739354.
Article
21. Anderson KS, Sibani S, Wallstrom G, Qiu J, Mendoza EA, Raphael J, et al. Protein microarray signature of autoantibody biomarkers for the early detection of breast cancer. J Proteome Res. 2011; 10:85–96. PMID: 20977275.
Article
22. Bateman NW, Sun M, Hood BL, Flint MS, Conrads TP. Defining central themes in breast cancer biology by differential proteomics: conserved regulation of cell spreading and focal adhesion kinase. J Proteome Res. 2010; 9:5311–5324. PMID: 20681588.
Article
23. Kristiansen TZ, Harsha HC, Grønborg M, Maitra A, Pandey A. Differential membrane proteomics using 18O-labeling to identify biomarkers for cholangiocarcinoma. J Proteome Res. 2008; 7:4670–4677. PMID: 18839982.
24. An HJ, Lebrilla CB. A glycomics approach to the discovery of potential cancer biomarkers. Methods Mol Biol. 2010; 600:199–213. PMID: 19882130.
Article
25. Choudhary C, Mann M. Decoding signalling networks by mass spectrometry-based proteomics. Nat Rev Mol Cell Biol. 2010; 11:427–439. PMID: 20461098.
Article
26. Madian AG, Regnier FE. Profiling carbonylated proteins in human plasma. J Proteome Res. 2010; 9:1330–1343. PMID: 20121119.
Article
27. Iwabata H, Yoshida M, Komatsu Y. Proteomic analysis of organ-specific post-translational lysine-acetylation and -methylation in mice by use of anti-acetyllysine and -methyllysine mouse monoclonal antibodies. Proteomics. 2005; 5:4653–4664. PMID: 16247734.
Article
28. Ceroni A, Sibani S, Baiker A, Pothineni VR, Bailer SM, LaBaer J, et al. Systematic analysis of the IgG antibody immune response against varicella zoster virus (VZV) using a self-assembled protein microarray. Mol Biosyst. 2010; 6:1604–1610. PMID: 20514382.
Article
29. Wong J, Sibani S, Lokko NN, LaBaer J, Anderson KS. Rapid detection of antibodies in sera using multiplexed self-assembling bead arrays. J Immunol Methods. 2009; 350:171–182. PMID: 19732778.
Article
30. Anderson NL. The clinical plasma proteome: a survey of clinical assays for proteins in plasma and serum. Clin Chem. 2010; 56:177–185. PMID: 19884488.
Article
31. Osterfeld SJ, Yu H, Gaster RS, Caramuta S, Xu L, Han SJ, et al. Multiplex protein assays based on real-time magnetic nanotag sensing. Proc Natl Acad Sci USA. 2008; 105:20637–20640. PMID: 19074273.
Article
32. Paulovich AG, Billheimer D, Ham AJ, Vega-Montoto L, Rudnick PA, Tabb DL, et al. Interlaboratory study characterizing a yeast performance standard for benchmarking LC-MS platform performance. Mol Cell Proteomics. 2010; 9:242–254. PMID: 19858499.
Article
33. Rudnick PA, Clauser KR, Kilpatrick LE, Tchekhovskoi DV, Neta P, Blonder N, et al. Performance metrics for liquid chromatography-tandem mass spectrometry systems in proteomics analyses. Mol Cell Proteomics. 2010; 9:225–241. PMID: 19837981.
Article
34. Addona TA, Abbatiello SE, Schilling B, Skates SJ, Mani DR, Bunk DM, et al. Multi-site assessment of the precision and reproducibility of multiple reaction monitoring-based measurements of proteins in plasma. Nat Biotechnol. 2009; 27:633–641. PMID: 19561596.
Article
35. Tabb DL, Vega-Montoto L, Rudnick PA, Variyath AM, Ham AJ, Bunk DM, et al. Repeatability and reproducibility in proteomic identifications by liquid chromatography-tandem mass spectrometry. J Proteome Res. 2010; 9:761–776. PMID: 19921851.
Article
36. Rodriguez H, Snyder M, Uhlén M, Andrews P, Beavis R, Borchers C, et al. Recommendations from the 2008 international summit on proteomics data release and sharing policy: the amsterdam principles. J Proteome Res. 2009; 8:3689–3692. PMID: 19344107.
Article
37. Gloriam DE, Orchard S, Bertinetti D, Björling E, Bongcam-Rudloff E, Borrebaeck CA, et al. A community standard format for the representation of protein affinity reagents. Mol Cell Proteomics. 2010; 9:1–10. PMID: 19674966.
Article
38. Xu X, Zhang J, Zhang L, Liu W, Weisel CP. Selective detection of monohydroxy metabolites of polycyclic aromatic hydrocarbons in urine using liquid chromatography/triple quadrupole tandem mass spectrometry. Rapid Commun Mass Spectrom. 2004; 18:2299–2308. PMID: 15384151.
Article
39. Xu X, Roman JM, Issaq HJ, Keefer LK, Veenstra TD, Ziegler RG. Quantitative measurement of endogenous estrogens and estrogen metabolites in human serum by liquid chromatography-tandem mass spectrometry. Anal Chem. 2007; 79:7813–7821. PMID: 17848096.
Article
40. James A, Jorgensen C. Basic design of MRM assays for peptide quantification. Methods Mol Biol. 2010; 658:167–185. PMID: 20839104.
Article
41. Kiyonami R, Schoen A, Prakash A, Peterman S, Zabrouskov V, Picotti P, et al. Increased selectivity, analytical precision, and throughput in targeted proteomics. Mol Cell Proteomics. 2011; 10:M110.002931. PMID: 20664071.
Article
42. Gerszten RE, Carr SA, Sabatine M. Integration of proteomic-based tools for improved biomarkers of myocardial injury. Clin Chem. 2010; 56:194–201. PMID: 20022985.
Article
43. Kuhn E, Wu J, Karl J, Liao H, Zolg W, Guild B. Quantification of C-reactive protein in the serum of patients with rheumatoid arthritis using multiple reaction monitoring mass spectrometry and 13C-labeled peptide standards. Proteomics. 2004; 4:1175–1186. PMID: 15048997.
44. Kuhn E, Addona T, Keshishian H, Burgess M, Mani DR, Lee RT, et al. Developing multiplexed assays for troponin I and interleukin-33 in plasma by peptide immunoaffinity enrichment and targeted mass spectrometry. Clin Chem. 2009; 55:1108–1117. PMID: 19372185.
Article
45. 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. 2004; 3:235–244. PMID: 15113099.
Article
46. Ahn YH, Lee JY, Lee JY, Kim YS, Ko JH, Yoo JS. Quantitative analysis of an aberrant glycoform of TIMP1 from colon cancer serum by L-PHA-enrichment and SISCAPA with MRM mass spectrometry. J Proteome Res. 2009; 8:4216–4224. PMID: 19645485.
Article
47. Reid JD, Holmes DT, Mason DR, Shah B, Borchers CH. Towards the development of an immuno MALDI (iMALDI) mass spectrometry assay for the diagnosis of hypertension. J Am Soc Mass Spectrom. 2010; 21:1680–1686. PMID: 20199871.
Article
48. Jiang J, Parker CE, Fuller JR, Kawula TH, Borchers CH. An immunoaffinity tandem mass spectrometry (iMALDI) assay for detection of Francisella tularensis. Anal Chim Acta. 2007; 605:70–79. PMID: 18022413.
Article
49. Mansfield E, O'Leary TJ, Gutman SI. Food and drug administration regulation of in vitro diagnostic devices. J Mol Diagn. 2005; 7:2–7. PMID: 15681468.
Article
50. FDA/CDRH Public Meeting: oversight of Laboratory Developed Tests (LDTs), July 19-20, 2010. U.S. Food and Drug Administration (FDA). Updated on Sep 2010. http://www.fda.gov/MedicalDevices/NewsEvents/WorkshopsConferences/ucm212830.htm.
51. Shi L, Campbell G, Jones WD, Campagne F, Wen Z, Walker SJ, et al. The MicroArray Quality Control (MAQC)-II study of common practices for the development and validation of microarray-based predictive models. Nat Biotechnol. 2010; 28:827–838. PMID: 20676074.
Article
52. Draft guidance for industry, clinical laboratories, and FDA staff: in vitro diagnostic multivariate index assays. Center for Devices and Radiological Health (CDRH). Updated on Jul 2007. http://www.fda.gov/downloads/MedicalDevices/DeviceRegulationandGuidance/GuidanceDocuments/ucm071455.pdf.
53. 510(k) Substantial equivalence determination decision summary. U.S. Food and drug administration. Updated on Mar 2011. http://www.accessdata.fda.gov/cdrh_docs/reviews/K081754.pdf.
54. Rodriguez H, Tezak Z, Mesri M, Carr SA, Liebler DC, Fisher SJ, et al. Analytical validation of protein-based multiplex assays: a workshop report by the NCI-FDA interagency oncology task force on molecular diagnostics. Clin Chem. 2010; 56:237–243. PMID: 20007859.
Article
55. Regnier FE, Skates SJ, Mesri M, Rodriguez H, Tezak Z, Kondratovich MV, et al. Protein-based multiplex assays: mock presubmissions to the US food and drug administration. Clin Chem. 2010; 56:165–171. PMID: 20007858.
Article
56. Pacini F. Follow-up of differentiated thyroid cancer. Eur J Nucl Med Mol Imaging. 2002; 29(S2):S492–S496. PMID: 12192551.
Article
57. Saghari M, Gholamrezanezhad A, Mirpour S, Eftekhari M, Takavar A, Fard-Esfahani A, et al. Efficacy of radioiodine therapy in the treatment of elevated serum thyroglobulin in patients with differentiated thyroid carcinoma and negative whole-body iodine scan. Nucl Med Commun. 2006; 27:567–572. PMID: 16794517.
Article
58. Spencer CA. Recoveries cannot be used to authenticate thyroglobulin (Tg) measurements when sera contain Tg autoantibodies. Clin Chem. 1996; 42:661–663. PMID: 8653888.
Article
59. Spencer CA. Challenges of serum thyroglobulin (Tg) measurement in the presence of Tg autoantibodies. J Clin Endocrinol Metab. 2004; 89:3702–3704. PMID: 15292292.
Article
60. Preissner CM, O'Kane DJ, Singh RJ, Morris JC, Grebe SK. Phantoms in the assay tube: heterophile antibody interferences in serum thyroglobulin assays. J Clin Endocrinol Metab. 2003; 88:3069–3074. PMID: 12843145.
Article
61. Hoofnagle AN, Becker JO, Wener MH, Heinecke JW. Quantification of thyroglobulin, a low-abundance serum protein, by immunoaffinity peptide enrichment and tandem mass spectrometry. Clin Chem. 2008; 54:1796–1804. PMID: 18801935.
Article
62. Hoofnagle AN. Peptide lost and found: internal standards and the mass spectrometric quantification of peptides. Clin Chem. 2010; 56:1515–1517. PMID: 20739635.
Article
63. Ciccimaro E, Hanks SK, Yu KH, Blair IA. Absolute quantification of phosphorylation on the kinase activation loop of cellular focal adhesion kinase by stable isotope dilution liquid chromatography/mass spectrometry. Anal Chem. 2009; 81:3304–3313. PMID: 19354260.
Article
64. CFR-Code federal regulations title 21. U.S. Food and Drug Administration (FDA). Updated on Apr 2010. http://www.accessdata.fda.gov/scripts/cdrh/cfdocs/cfcfr/CFRSearch.cfm?fr=862.2860.
65. Clinical and Laboratory Standards Institute (CLSI). Updated on Feb 2011. http://www.clsi.org.
66. Spitz MR, Bondy ML. The evolving discipline of molecular epidemiology of cancer. Carcinogenesis. 2010; 31:127–134. PMID: 20022891.
Article
67. Rosa DD, Ismael G, Lago LD, Awada A. Molecular-targeted therapies: lessons from years of clinical development. Cancer Treat Rev. 2008; 34:61–80. PMID: 17826917.
Article
68. Bredel M, Scholtens DM, Harsh GR, Bredel C, Chandler JP, Renfrow JJ, et al. A network model of a cooperative genetic landscape in brain tumors. JAMA. 2009; 302:261–275. PMID: 19602686.
Article
69. International Cancer Genome Consortium. International network of cancer genome projects. Nature. 2010; 464:993–998. PMID: 20393554.
70. Cancer Genome Atlas Research Network. Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature. 2008; 455:1061–1068. PMID: 18772890.
71. Verhaak RG, Hoadley KA, Purdom E, Wang V, Qi Y, Wilkerson MD, et al. Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell. 2010; 17:98–110. PMID: 20129251.
Article
Full Text Links
  • KJLM
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr