Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Clinical proteomics: translating benchside promise into bedside reality

Key Points

  • Early detection of cancer should increase the likelihood that treatments achieve a true cure. However, several cancers, such as ovarian cancer, lack a specific symptom and a specific biomarker, and accurate and reliable diagnostic non-invasive modalities.

  • The microenvironment of the tumor–host interaction is a potential source for biomarkers that could be shed into the serum proteome. However, looking for single proteins with expression levels in serum that are altered significantly as a result of the disease process is laborious and time consuming.

  • Analysis of serum proteomic patterns comprising many individual proteins, each of which independently were not able to differentiate diseased from healthy individuals, has recently been shown to provide a diagnostic end point for cancer detection.

  • Analysing the proteins that change in actual diseased human tissue offers new opportunities for the identification of therapeutic targets.

  • Laser-capture microdissection (LCM) is a technology for procuring pure cell populations from a stained tissue section under direct microscopic visualization, and has been applied to discover new protein targets that are either a cause, or consequence, of the disease process in the actual tissue.

  • A new type of protein array — the reverse-phase protein array — is particularly suited to analysing protein signalling pathways using small numbers of human tissue cells microdissected from biopsy specimens procured during clinical trials.

  • In summary, clinical proteomics could have significant potential in the following crucial elements of patient care and management:

  • early detection of the disease using proteomic patterns of body fluid samples;

  • diagnosis based on proteomic signatures as a complement to histopathology;

  • individualized selection of therapeutic combinations that best target the patient's entire disease-specific protein network;

  • real-time assessment of therapeutic efficacy and toxicity;

  • rational redirection of therapy based on changes in the diseased protein network that are associated with drug resistance.

  • Combinatorial therapy, in which the signalling pathway itself is viewed as the target rather than any single 'node' in the pathway, might offer new opportunities at increasing efficacy while decreasing toxicity.

Abstract

The ultimate goal of proteomics is to characterize the information flow through protein networks. This information can be a cause, or a consequence, of disease processes. Clinical proteomics is an exciting new subdiscipline of proteomics that involves the application of proteomic technologies at the bedside, and cancer, in particular, is a model disease for studying such applications. Here, we describe proteomic technologies that are being developed to detect cancer earlier, to discover the next generation of targets and imaging biomarkers, and finally to tailor the therapy to the patient.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Example of a protein signalling pathway.
Figure 2: Tumour–host interaction.
Figure 3: SELDI mass spectrometry.
Figure 4: Two-colour 2D-gel methodology.
Figure 5: Protein microarray.
Figure 6: Reverse-phase arrays.
Figure 7: Whole-body array.
Figure 8: Combinatorial therapy.
Figure 9: Proteomic technology applied to cancer-patient management.

Similar content being viewed by others

References

  1. Liotta, L. & Petricoin, E. Molecular profiling of human cancer. Nature Rev. Genet. 1, 48–56 (2000).A description of new genomic and proteomic technologies that are allowing the reclassification of human cancer on the basis of molecular portraits instead of histology alone.

    CAS  PubMed  Google Scholar 

  2. Ideker, T. et al. Integrated genomic and proteomic analyses of a systematically perturbed metabolic network. Science 292, 929–934 (2001).

    CAS  PubMed  Google Scholar 

  3. Schwikowski, B., Uetz, P. & Fields, S. A network of protein–protein interactions in yeast. Nature Biotechnol. 18, 1257–1261 (2000).

    CAS  Google Scholar 

  4. Legrain, P., Jestin, J. L. & Schachter, V. From the analysis of protein complexes to proteome-wide linkage maps. Curr. Opin. Biotechnol. 4, 402–407 (2000).

    Google Scholar 

  5. Blume-Jensen, P. & Hunter, T. Oncogenic kinase signaling. Nature 411, 355–365 (2001).

    CAS  PubMed  Google Scholar 

  6. Pawson, T. Protein modules and signaling networks. Nature 373, 573–580 (1995).

    CAS  PubMed  Google Scholar 

  7. Liotta, L. A. & Kohn, E. C. The microenvironment of the tumour–host interface. Nature 411, 375–379 (2001).

    CAS  PubMed  Google Scholar 

  8. Ozols, R. F., Rubin, S. C., Thomas, G. M. & Robboy, S. J. in Principles and Practice of Gynecologic Oncology (eds Hoskins, W. J., Perez, C. A. & Young, R. C.) 981–1058 (Lippincott Williams & Wilkins, Philadelphia, Pennsylvania, 2000).

    Google Scholar 

  9. Menon, U. & Jacobs, I. in Principles and Practice of Gynecologic Oncology (eds Hoskins, W. J., Perez, C. A. &, Young, R. C.) 165–182 (Lippincott Williams & Wilkins, Philadelphia, Pennsylvania, 2000).

    Google Scholar 

  10. Bast, R. C. et al. A radioimmunoassay using a monoclonal antibody to monitor the course of epithelial ovarian cancer. N. Engl. J. Med. 309, 883–887 (1983).

    PubMed  Google Scholar 

  11. Menon, U. & Jacobs, I. J. Recent developments in ovarian cancer screening. Curr. Opin. Obstet. Gynecol. 12, 39–42 (2000).

    CAS  PubMed  Google Scholar 

  12. Jacobs, I. J. et al. Screening for ovarian cancer: a pilot randomized controlled trial. Lancet 353, 1207–1210 (1999).

    CAS  PubMed  Google Scholar 

  13. Cohen, L. S., Escobar P. F., Scharm, C., Glimco, B. & Fishman, D. A. Three-dimensional power Doppler ultrasound improves the diagnostic accuracy for ovarian cancer prediction. Gynecol. Oncol. 82, 40–48 (2001).

    CAS  PubMed  Google Scholar 

  14. Adam, B. L., Vlahou, A., Semmes, O. J. & Wright, G. L. Jr. Proteomic approaches to biomarker discovery in prostate and bladder cancers. Proteomics 1, 1264–1270 (2001).

    CAS  PubMed  Google Scholar 

  15. Carter, D. et al. Purification and characterization of the mammaglobin/lipophilin B complex, a promising diagnostic marker for breast cancer. Biochemistry 41, 6714–6722 (2002).

    CAS  PubMed  Google Scholar 

  16. Rosty, C. et al. Identification of hepatocarcinoma-intestine-pancreas/pancreatitis-associated protein I as a biomarker for pancreatic ductal adenocarcinoma by protein biochip technology. Cancer Res. 62, 1868–1875 (2002).

    CAS  PubMed  Google Scholar 

  17. Xiao, Z. et al. Quantitation of serum prostate-specific membrane antigen by a novel protein biochip immunoassay discriminates benign from malignant prostate disease. Cancer Res. 61, 6029–6033 (2001).

    CAS  PubMed  Google Scholar 

  18. Kim, J. H. et al. Osteopontin as a potential diagnostic biomarker for ovarian cancer. JAMA 287, 1671–1679 (2002).

    CAS  PubMed  Google Scholar 

  19. Petricoin, E. F. et al. Use of proteomic patterns in serum to identify ovarian cancer. Lancet 359, 572–577 (2002).This paper describes the invention of a new process and diagnostic paradigm that is based on serum proteomic pattern analysis instead of a discovery effort based on a 'one-biomarker-at-a-time' approach.

    CAS  PubMed  Google Scholar 

  20. Ball, G. et al. An integrated approach utilizing artificial neural networks and SELDI mass spectrometry for the classification of human tumours and rapid identification of potential biomarkers. Bioinformatics 18, 395–404 (2002).

    CAS  PubMed  Google Scholar 

  21. Ting, K. L., Lee, R. C., Chang, C. L. & Guarino, A. M. The relationship between the mass spectra of drugs and their biological activity — an application of artificial intelligence to chemistry. Comput. Biol. Med. 4, 301–332 (1975).

    CAS  PubMed  Google Scholar 

  22. Nicholson, J. K., Connelly, J., Lindon, J. C. & Holmes, E. Metabonomics: a platform for studying drug toxicity and gene function. Nature Rev. Drug Discov. 1, 153–161 (2002).

    CAS  Google Scholar 

  23. Alizadeh, A. A. et al. Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling. Nature 403, 503–511 (2000).

    CAS  PubMed  Google Scholar 

  24. Golub, T. R. et al. Molecular classification of cancer: class discovery and class prediction by gene expression monitoring. Science 286, 531–537 (1999).

    CAS  PubMed  Google Scholar 

  25. Lindahl, D., Palmer, J. & Edenbrandt, L. Myocardial SPET: artificial neural networks describe extent and severity of perfusion defects. Clin. Physiol. 19, 497–503 (1999).

    CAS  PubMed  Google Scholar 

  26. Lapuerta, P. et al. Neural network assessment of perioperative cardiac risk in vascular surgery patients. Med. Decis. Making 18, 70–75 (1998).

    CAS  PubMed  Google Scholar 

  27. Holland, J. H. (ed.) Adaptation in Natural and Artificial Systems: an Introductory Analysis with Applications to Biology, Control, and Artificial Intelligence 3rd edn (MIT Press, Cambridge, Massachusetts, 1994).

    Google Scholar 

  28. Kohonen, T. Self-organizing formation of topologically correct feature maps. Biol. Cybernetics 43, 59–69 (1982).

    Google Scholar 

  29. Kohonen, T. The self-organizing map. Proc. IEEE 78, 1464–1480 (1990).

    Google Scholar 

  30. Tou, J. T. & Gonzalez, R. (eds) in Pattern Recognition Principles (eds Tou, J. T. & Gonzalez, R.) 75–109 (Addison Weley, Reading, Massachusetts, 1974).

    Google Scholar 

  31. Stoeckli, M., Chaurand, P., Hallahan, D. E. & Caprioli, R. M. Imaging mass spectrometry: a new technology for the analysis of protein expression in mammalian tissues. Nature Med. 7, 493–496 (2001).

    CAS  PubMed  Google Scholar 

  32. Emmert-Buck, M. R. et al. Laser capture microdissection. Science 274, 998–1001 (1996).

    CAS  PubMed  Google Scholar 

  33. Emmert-Buck, M. R. et al. An approach to proteomic analysis of human tumors. Mol. Carcinog. 27, 158–165 (2000).

    CAS  PubMed  Google Scholar 

  34. Craven, R. A., Totty, N., Harnden, P., Selby, P. J. & Banks, R. E. Laser capture microdissection and two-dimensional polyacrylamide gel electrophoresis: evaluation of tissue preparation and sample limitations. Am. J. Pathol. 160, 815–822 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Ornstein, D. K. et al. Proteomic analysis of laser capture microdissected human prostate cancer and in vitro prostate cell lines. Electrophoresis 21, 2235–2242 (2000).

    CAS  PubMed  Google Scholar 

  36. Wulfkuhle, J. D. et al. New approaches to proteomic analysis of breast cancer. Proteomics 1, 1205–1215 (2001).

    CAS  PubMed  Google Scholar 

  37. Jones, M. B. et al. Proteomic analysis and identification of new biomarkers and therapeutic targets for invasive ovarian cancer. Proteomics 2, 76–84 (2002).

    CAS  PubMed  Google Scholar 

  38. Knezevic, V. et al. Proteomic profiling of the cancer microenvironment by antibody arrays. Proteomics 1, 1271–1278 (2001).

    CAS  PubMed  Google Scholar 

  39. Ahram, M. et al. Proteomic analysis of human prostate cancer. Mol. Carcinog. 33, 9–15 (2002).

    CAS  PubMed  Google Scholar 

  40. Gorg, A. et al. The current state of two-dimensional electrophoresis with immobilized pH gradients. Electrophoresis 21, 1037–1053 (2000).

    CAS  PubMed  Google Scholar 

  41. Hanash, S. M. Biomedical applications of two-dimensional electrophoresis using immobilized pH gradients: current status. Electrophoresis 21, 1202–1209 (2000).

    CAS  PubMed  Google Scholar 

  42. Herbert, B. R., Sanchez, J.-C. & Bini, L. in Proteome Reseach: New Frontiers in Functional Genomics Ch. 2 (eds Wilkens, M. R., Williams, K. L., Appel, R. D. & Hochstrasser, D. F.) 13–30 (Springer–Verlag, New York, 1997).

    Google Scholar 

  43. Shen, Y. et al. High-throughput proteomics using high-efficiency multiple-capillary liquid chromatography with on-line high-performance ESI FTICR mass spectrometry. Anal. Chem. 73, 3011–3021 (2001).The design and application of an automated multiple-capillary liquid chromatography (LC) system for high-throughput proteome analysis using a high-magnetic-field Fourier transform ion cyclotron resonance (FTICR) mass spectrometer for a combined resolving power of > 6 × 107 components. This set provided the characterization of 1,000 proteins from a single capillary LC–FTICR analysis.

    CAS  PubMed  Google Scholar 

  44. Li, J., Wang, C., Kelly, J. F., Harrison, D. J. & Thibault, P. Rapid and sensitive separation of trace level protein digests using microfabricated devices coupled to a quadrupole-time-of-flight mass spectrometer. Electrophoresis 21, 198–210 (2000).

    CAS  PubMed  Google Scholar 

  45. Gygi, S. P. et al. Quantitative analysis of complex protein mixtures using isotope coded affinity tags. Nature Biotechnol. 17, 994–999 (1999).

    CAS  Google Scholar 

  46. Washburn, M. P., Wolters, D. & Yates, J. R. Large scale analysis of the yeast proteome by multidimensional protein identification technology. Nature Biotechnol. 19, 242–247 (2001).A method for rapid and large-scale proteome analysis by multidimensional LC coupled with tandem mass spectrometry, termed multidimensional protein identification technology (MudPIT). This approach was applied to yeast proteomic analysis, and a total of 1,484 proteins were detected and identified, including the identification of 131 proteins that are membrane localized — a highly problematic class of proteins due to their intrinsic hydrophobic nature.

    CAS  Google Scholar 

  47. Krutchinsky, A. N., Kalkum, M. & Chait, B. T. Automatic identification of proteins with a MALDI-quadrupole ion trap mass spectrometer. Anal. Chem. 73, 5066–5077 (2001).

    CAS  PubMed  Google Scholar 

  48. Washburn, M. P., Ulaszek, R., Deciu, C., Schieltz, D. M. & Yates, J. R. Analysis of quantitative proteomic data generated via multidimensional protein identification technology. Anal. Chem. 74, 1650–1657 (2002).

    CAS  PubMed  Google Scholar 

  49. Zhou, H., Ranish, J. A., Watts, J. D. & Aebersold, R. Quantitative proteome analysis by solid-phase isotope tagging and mass spectrometry. Nature Biotechnol. 20, 512–515 (2002).A method for directly tagging complex mixtures of peptides and proteins using a solid-phase capture and release process with the captured peptides analysed by microcapillary LC and tandem mass spectrometry (microLC–MS/MS).

    CAS  Google Scholar 

  50. Zhou, G. et al. 2D differential in-gel electrophoresis for the identification of human esophageal squamous cell cancer-specific protein markers Mol. Cell. Proteomics 1, 117–123 (2002).

    CAS  PubMed  Google Scholar 

  51. Sreekumar, A. et al. Profiling of cancer cells using protein microarrays: discovery of novel radiation-regulated proteins. Cancer Res. 61, 7585–7593 (2001).This paper describes an antibody-microarray-based platform consisting of 146 distinct antibodies to monitor alterations of protein levels induced by exposure of colon carcinoma cell lines to ionizing radiation.

    CAS  PubMed  Google Scholar 

  52. MacBeath, G. Proteomics comes to the surface. Nature Biotechnol. 19, 828–829 (2001).

    CAS  Google Scholar 

  53. Walter, G., Bussow, K., Lueking, A. & Glokler, J. High-throughput protein arrays: prospects for molecular diagnostics. Trends Mol. Med. 8, 250–253 (2002).

    CAS  PubMed  Google Scholar 

  54. Kuruvilla, F. G., Shamji, A. F., Sternson, S. M., Hergenrother, P. J. & Schreiber, S. L. Dissecting glucose signalling with diversity-oriented synthesis and small-molecule microarrays. Nature 416, 653–657 (2002).

    CAS  PubMed  Google Scholar 

  55. Paweletz, C. P. et al. Reverse-phase protein microarrays which capture disease progression show activation of pro-survival pathways at the cancer invasion front. Oncogene 20, 1981–1989 (2001).The invention of a new type of protein array in which extremely small amounts of clinical material can be immobilized and used to study signalling pathway activation.

    CAS  PubMed  Google Scholar 

  56. Torhorst, J. et al. Tissue microarrays for rapid linking of molecular changes to clinical endpoints. Am. J. Pathol. 159, 2249–2256 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Vile, R. G., Russell, S. J. & Lemoine, N. R. Cancer gene therapy: hard lessons and new courses. Gene Ther. 7, 2–8 (2000).

    CAS  PubMed  Google Scholar 

  58. Wiebe, L. I. & Knaus, E. E. Enzyme-targeted, nucleoside-based radiopharmaceuticals for scintigraphic monitoring of gene transfer and expression. Curr. Pharm. Des. 7, 1893–1906 (2001).

    CAS  PubMed  Google Scholar 

  59. Liotta, L. A., Kohn, E. C. & Petricoin, E. F. Clinical proteomics: personalized molecular medicine. JAMA 286, 2211–2214 (2001).

    CAS  PubMed  Google Scholar 

  60. Liotta, L. & Petricoin, E. Molecular profiling of human cancer. Nature Rev. Genet. 1, 48–56 (2000).

    CAS  PubMed  Google Scholar 

  61. Karpati, G., Li, H. & Nalbantoglu, J. Molecular therapy for glioblastoma. Curr. Opin. Mol. Ther. 1, 545–552 (1999).

    CAS  PubMed  Google Scholar 

  62. Brown, C. K. & Kirkwood, J. M. Targeted therapy for malignant melanoma. Curr. Oncol. Rep. 3, 344–352 (2001).

    CAS  PubMed  Google Scholar 

  63. Frankel, A. E., Sievers, E. L. & Scheinberg, D. A. Cell surface receptor-targeted therapy of acute myeloid leukemia: a review. Cancer Biother. Radiopharm. 15, 459–476 (2000).

    CAS  PubMed  Google Scholar 

  64. Cheng, J. D., Rieger, P. T., von Mehren, M., Adams, G. P. & Weiner, L. M. Recent advances in immunotherapy and monoclonal antibody treatment of cancer. Semin. Oncol. Nurs. 16 (Suppl. 1), 2–12 (2000).

    CAS  PubMed  Google Scholar 

  65. Gasparini, G. & Gion, M. Molecular-targeted anticancer therapy: challenges related to study design and choice of proper endpoints. Cancer J. Sci. Am. 6, 117–131 (2000).

    CAS  Google Scholar 

  66. Cimoli, G. et al. Signaling proteins as innovative targets for antineoplastic therapy: our experience with the signaling protein c-myc. Tumori. 87, S20–S23 (2001).

    CAS  PubMed  Google Scholar 

  67. Kolonin, M., Pasqualini, R. & Arap, W. Molecular addresses in blood vessels as targets for therapy. Curr. Opin. Chem. Biol. 5, 308–313 (2001).

    CAS  PubMed  Google Scholar 

  68. Rosenwald, A. et al. The use of molecular profiling to predict survival after chemotherapy for diffuse large-B-cell lymphoma. N. Engl. J. Med. 346, 1937–1947 (2002).This paper describes the use of hierarchical clustering analysis of gene-expression profiles of biopsy samples from 240 diffuse large-B-cell lymphoma patients to build a molecular predictor of survival after chemotherapy.

    PubMed  Google Scholar 

  69. Ponder, B. A. Cancer genetics. Nature 411, 337–341 (2001).

    Google Scholar 

  70. Evan, G. I. & Vousden, K. H. Proliferation, cell cycle and apoptosis in cancer. Nature 411, 342–348 (2001).

    CAS  PubMed  Google Scholar 

  71. Kaptain, S., Tan, L. K. & Chen, B. HER2/NEU and breast cancer. Diagn. Mol. Pathol. 10, 139–152 (2001).

    CAS  PubMed  Google Scholar 

  72. Leyland-Jones, B. Trastuzumab: hopes and realities. Lancet Oncol. 3, 137–144 (2002).

    CAS  PubMed  Google Scholar 

  73. Sebolt-Leopold, J. S. Development of anticancer drugs targeting the MAP kinase pathway. Oncogene 19, 6594–6599 (2000).

    CAS  PubMed  Google Scholar 

  74. Santen, R. J. et al. The role of mitogen-activated protein (MAP) kinase in breast cancer. J. Steroid Biochem. Mol. Biol. 80, 239–256 (2002).

    CAS  PubMed  Google Scholar 

  75. Thiesing, J. T., Ohno-Jones, S., Kolibaba, K. S. & Druker, B. J. Efficacy of STI571, an ABL tyrosine kinase inhibitor, in conjunction with other antileukemic agents against BCR–ABL-positive cells. Blood 96, 3195–3199 (2000).These experiments show that combinations of Gleevec with interferon-α, hydroxyurea, daunorubicin and cytosine arabinoside have increased antiproliferative effects compared with STI571 alone, suggesting that clinical trials to test these combinations might be worthwhile.

    CAS  PubMed  Google Scholar 

  76. Druker, B. J. et al. Efficacy and safety of a specific inhibitor of the BCR–ABL tyrosine kinase in chronic myeloid leukemia. N. Engl. J. Med. 344, 1031–1037 (2001).

    CAS  PubMed  Google Scholar 

  77. Vlahos, C. J. & Stancato, L. F. in Platelets and Megakaryoctyes: Methods and Protocols (eds Gibbons, J. M. & Mahaut-Smith, M. P.) (Humana, Totowa, New Jersey, 2002).

    Google Scholar 

  78. Traxler, P. et al. Tyrosine kinase inhibitors: from rational design to clinical trials. Med. Res. Rev. 21, 499–512 (2001).

    CAS  PubMed  Google Scholar 

  79. Zwick, E., Bange, J. & Ullrich, A. Receptor tyrosine kinases as targets for anticancer drugs. Trends Mol. Med. 8, 17–23 (2002).

    CAS  PubMed  Google Scholar 

  80. Normanno, N. et al. Cooperative inhibitory effect of ZD1839 (Iressa) in combination with trastuzumab (Herceptin) on human breast cancer cell growth. Ann. Oncol. 13, 65–72 (2002).

    CAS  PubMed  Google Scholar 

  81. Moasser, M. M., Basso, A., Averbuch, S. D. & Rosen, N. The tyrosine kinase inhibitor ZD1839 ('Iressa') inhibits HER2-driven signaling and suppresses the growth of HER2-overexpressing tumor cells. Cancer Res. 61, 7184–7188 (2001).An analysis of the ability of ZD1839 (Iressa) to inhibit the phosphorylation of EGFR, HER2, HER3 and AKT and growth inhibition in a series of human breast cancer cell lines. These studies indicate that HER2-overexpressing tumours might be especially susceptible to Iressa.

    CAS  PubMed  Google Scholar 

  82. Cuello, M. et al. Down-regulation of the ERBB-2 receptor by trastuzumab (Herceptin) enhances tumor necrosis factor-related apoptosis-inducing ligand-mediated apoptosis in breast and ovarian cancer cell lines that overexpress ERBB-2. Cancer Res. 61, 4892–4900 (2001).

    CAS  PubMed  Google Scholar 

  83. Paweletz, C. P. et al. Rapid protein display profiling of cancer progression directly from human tissue using a protein biochip. Drug Dev. Res. 49, 34–42 (2000).

    CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Emanuel F. Petricoin.

Related links

Related links

DATABASES

Cancer.gov

breast cancer

colon cancer

lung cancer

ovarian cancer

prostate cancer

LocusLink

ABL

AKT

aurora 2

ERBB1

ERBB2

GFAP

myoglobin

PSA

Medscape DrugInfo

Gleevec

Herceptin

FURTHER INFORMATION

Clinical Proteomics Program Databank

National Cancer Institute

Glossary

METASTASIS

The movement or spreading of cancer cells from one organ or tissue to another. Cancer cells usually spread through the bloodstream or lymph system.

PERITONEAL CAVITY

The peritoneum is the thin membrane that lines the abdominal cavity.

TRAINING

A process in which a computer-driven system is provided data from a training set in which the outcome is known and is unblinded.

2D-PAGE

A method for separating proteins by mass and charge.

pI GRADIENTS

Isoelectric gradients are formed by subjecting a defined set of small molecules with specific net charges to an electric current, which allows the separation of proteins within the gradient, even if the proteins only differ in charge by one-thousandth of a pH unit.

CY3/CY5

Cy3 and Cy5 are water-soluble cyanine dyes that can be used as fluorescent labels for proteins and modified oligonucleotides.

REMATING

A computer-driven data-analysis process that is performed by combining solutions to select and retain the best elements of each solution, and discard those elements that do not solve the problem.

SELECTIVE PRESSURE

The ability of a computer-driven data-analysis process to restrict the solution output to those that pass a fixed criteria — in this case, the ability to distinguish normal from cancerous samples.

ICAT

A method in which proteins are labelled using isotope-coded affinity tags, which allows them to be systematically identified and quantified.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Petricoin, E., Zoon, K., Kohn, E. et al. Clinical proteomics: translating benchside promise into bedside reality. Nat Rev Drug Discov 1, 683–695 (2002). https://doi.org/10.1038/nrd891

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrd891

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing