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:

Technology Insight: pharmacoproteomics for cancer—promises of patient-tailored medicine using protein microarrays

Abstract

Patient-tailored medicine can be defined as the selection of specific therapeutics to treat disease in a particular individual based on genetic, genomic or proteomic information. While individualized treatments have been used in medicine for years, advances in cancer treatment have now generated a need to more precisely define and identify those patients who will derive the most benefit from new-targeted agents. Cellular signaling pathways are a protein-based network, and the intended drug effect is to disrupt aberrant protein phosphorylation-based enzymatic activity and epigenetic phenomena. Pharmacoproteomics, or the tailoring of therapy based on proteomic knowledge, will begin to take a central role in this process. A new type of protein array platform, the reverse-phase protein microarray, shows potential for providing detailed information about the state of the cellular 'circuitry' from small samples such as patient biopsy specimens. Measurements of hundreds of specific phosphorylated proteins that span large classes of important signaling pathways can be obtained at once from only a few thousand cells. Clinical implementation of these new proteomic tools to aid the clinical, medical and surgical oncologist in making decisions about patient care will now require thoughtful communication between practicing clinicians and research scientists.

Key Points

  • An ever-increasing number of molecularly targeted agents for treating cancer are entering clinical use, generating a new need to better define and identify those patients who will most benefit from them

  • Proteomic technologies may eventually hold a dominant position in tailored medicine as we transition from pharmacogenomics to pharmacoproteomics, since gene transcript profiling cannot be used effectively to elucidate or monitor protein–protein interaction networks and signal transduction pathways

  • Protein microarrays that examine post-translational modifications, such as phosphorylation, in a global, high-throughput manner, can be used to profile the working state of cellular signaling pathways in human tissue

  • The reverse-phase protein microarray (RPA) format is uniquely suited to signal transduction profiling of small samples (e.g. biopsy specimens) such that rational selection and monitoring of patients for targeted medicine is now technically possible

  • Clinical implementation of RPAs for profiling patient tissue specimens before, during and after therapy could provide important diagnostic and prognostic information, and could help therapeutic decision making and monitoring of response or resistance

  • Establishment of RPAs or any stratification tool as routine clinical practice will require standardization of tissue collection and processing, and the cooperation and collaboration of surgeons, oncologists, pathologists and scientists

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

Access options

Buy this article

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

Figure 1: A roadmap for individualized cancer therapy.
Figure 2: Reverse-phase protein microarray construction.
Figure 3: New paradigm for treating advanced-stage metastatic disease using pharmacoproteomics.
Figure 4: Tailored combination therapy offers the hope of greater efficacy with reduced toxicity.

Similar content being viewed by others

References

  1. Jain KK (2002) Personalized medicine. Curr Opin Mol Ther 4: 548–558

    CAS  PubMed  Google Scholar 

  2. Baak JPA et al. (2003) Genomics and proteomics in cancer. Eur J Cancer 39: 1199–1215

    CAS  PubMed  Google Scholar 

  3. Segal E et al. (2005) From signatures to models: understanding cancer using microarrays. Nat Genet 37 (Suppl) S38–S45

    CAS  PubMed  Google Scholar 

  4. Brennan DJ et al. (2005) Application of DNA microarray technology in determining breast cancer prognosis and therapeutic response. Expert Opin Biol Ther 5: 1069–1083

    CAS  PubMed  Google Scholar 

  5. Nishizuka S et al. (2003) Proteomic profiling of the NCI-60 cancer cell lines using new high-density reverse-phase lysate microarrays. Proc Natl Acad Sci USA 100: 14229–14234

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Celis JE and Gromov P (2003) Proteomics in translational cancer research: toward an integrated approach. Cancer Cell 3: 9–15

    CAS  PubMed  Google Scholar 

  7. Hunter T (2000) Signaling—2000 and beyond. Cell 100: 113–127

    CAS  PubMed  Google Scholar 

  8. Gorg A et al. (2004) Current two-dimensional electrophoresis technology for proteomics. Proteomics 4: 3665–3685

    PubMed  Google Scholar 

  9. Gygi SP et al. (1999) Quantitative analysis of complex protein mixtures using isotope-coded affinity tags. Nat Biotechnol 17: 994–999

    CAS  PubMed  Google Scholar 

  10. Krutchinsky AN et al. (2001) Automatic identification of proteins with a MALDI-quadrupole ion trap mass spectrometer. Anal Chem 73: 5066–5077

    CAS  PubMed  Google Scholar 

  11. Washburn MP et al. (2001) Large scale analysis of the yeast proteome by multidimensional protein identification technology. Nat Biotechnol 19: 242–247

    CAS  PubMed  Google Scholar 

  12. Zhou G et al. (2002) 2D differential in-gel electrophoresis for the identification of esophageal scans cell cancer-specific protein markers. Mol Cell Proteomics 1: 117–124

    CAS  PubMed  Google Scholar 

  13. Eckel-Passow JE et al. (2005) Experimental design and analysis of antibody microarrays: applying methods from cDNA arrays. Cancer Res 65: 2985–2989

    CAS  PubMed  Google Scholar 

  14. Haab BB (2005) Antibody arrays in cancer research. Mol Cell Proteomics 4: 377–383

    CAS  PubMed  Google Scholar 

  15. Leuking A et al. (2005) Protein biochips: a new and versatile platform technology for molecular medicine. Drug Disc Today 10: 789–794

    Google Scholar 

  16. Liotta LA et al. (2001) Clinical proteomics: personalized molecular medicine. JAMA 286: 2211–2214

    CAS  PubMed  Google Scholar 

  17. Petricoin EF et al. (2002) Clinical proteomics: translating benchside promise into bedside reality. Nat Rev Drug Discov 1: 683–695

    CAS  PubMed  Google Scholar 

  18. Paweletz CP et al. (2001) Reverse phase protein microarrays which capture disease progression show activation of pro-survival pathways at the cancer invasion front. Oncogene 20: 1981–1989

    CAS  PubMed  Google Scholar 

  19. Zhu H and Snyder M (2003) Protein chip technology. Curr Opin Chem Biol 7: 55–63

    CAS  PubMed  Google Scholar 

  20. Cutler P (2003) Protein arrays: the current state-of-the-art. Proteomics 3: 3–18

    CAS  PubMed  Google Scholar 

  21. Ge H (2000) UPA, a universal protein array system for quantitative detection of protein–protein, protein–DNA, protein–RNA and protein–ligand interactions. Nucleic Acids Res 28: e3

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Liotta L and Petricoin E (2000) Molecular profiling of human cancer. Nat Rev Genet 1: 48–56

    CAS  PubMed  Google Scholar 

  23. MacBeath G (2002) Protein microarrays and proteomics. Nat Genet 32 (Suppl) 526–532

    CAS  PubMed  Google Scholar 

  24. Miller JC et al. (2003) Antibody microarray profiling of human prostate cancer sera: antibody screening and identification of potential biomarkers. Proteomics 3: 56–63

    CAS  PubMed  Google Scholar 

  25. Liotta LA et al. (2003) Protein microarrays: meeting analytical challenges for clinical applications. Cancer Cell 3: 317–325

    CAS  PubMed  Google Scholar 

  26. Angenendt P (2005) Progress in protein and antibody microarray technology. Drug Disc Today 10: 503–511

    CAS  Google Scholar 

  27. LaBaer J and Ramachandran N (2005) Protein microarrays as tools for functional proteomics. Curr Opin Chem Biol 9: 14–19

    CAS  PubMed  Google Scholar 

  28. Humphery-Smith I et al. (2002) Protein arrays for assessment of target selectivity. Drug Discov World 4: 17–27

    Google Scholar 

  29. MacBeath G and Schreiber SL (2000) Printing proteins as microarrays for high-throughput function determination. Science 289: 1760–1763

    CAS  PubMed  Google Scholar 

  30. Petach H and Gold L (2002) Dimensionality is the issue: use of photoaptamers in protein microarrays. Curr Opin Biotechnol 13: 309–314

    CAS  PubMed  Google Scholar 

  31. Weng S et al. (2002) Generating addressable protein microarrays with PROfusion covalent mRNA-protein fusion technology. Proteomics 2: 48–57

    CAS  PubMed  Google Scholar 

  32. Hultschig C et al. (2006) Recent advances of protein microarrays. Curr Opin Chem Biol 10: 4–10

    CAS  PubMed  Google Scholar 

  33. Robinson WH (2006) Antigen arrays for antibody profiling. Curr Opin Chem Biol 10: 67–72

    CAS  PubMed  Google Scholar 

  34. Sreekumar A et al. (2001) Profiling of cancer cells using protein microarrays: discovery of novel radiation-regulated proteins. Cancer Res 61: 7585–7593

    CAS  PubMed  Google Scholar 

  35. Sanchez-Carbayo M et al. (2006) Profiling bladder cancer using targeted antibody arrays. Am J Pathol 168: 93–103

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Grubb RL et al. (2003) Signal pathway profiling of prostate cancer using reverse phase protein microarrays. Proteomics 3: 2142–2146

    CAS  PubMed  Google Scholar 

  37. Petricoin III EF et al. (2005) Mapping molecular networks using proteomics: a vision for patient-tailored combination therapy. J Clin Oncol 23: 3614–3621

    CAS  PubMed  Google Scholar 

  38. Wulfkuhle JD et al. (2003) Signal pathway profiling of ovarian cancer from human tissue specimens using reverse-phase protein microarrays. Proteomics 3: 2085–2090

    CAS  PubMed  Google Scholar 

  39. Gulmann C et al. (2005) Proteomic analysis of apoptotic pathways reveals prognostic factors in follicular lymphoma. Clin Cancer Res 11: 5847–5855

    CAS  PubMed  Google Scholar 

  40. Sheehan KM et al. (2005) Use of reverse-phase protein microarrays and reference standard development for molecular network analysis of metastatic ovarian carcinoma. Mol Cell Proteomics 4: 346–355

    CAS  PubMed  Google Scholar 

  41. Emmert-Buck MR et al. (1996) Laser capture microdissection. Science 274: 998–1001

    CAS  PubMed  Google Scholar 

  42. Zha H et al. (2004) Similarities of prosurvival signals in Bcl-2-positive and Bcl-2-negative follicular lymhomas identified by reverse phase protein microarray. Lab Invest 84: 235–244

    CAS  PubMed  Google Scholar 

  43. Giltrane JM and Rimm DL (2005) Technology insight: identification of biomarkers with tissue microarray technology. Nat Clin Pract Oncol 1: 104–111

    Google Scholar 

  44. Espina V et al. (2003) Protein microarrays: molecular profiling technologies for clinical specimens. Proteomics 3: 2091–2100

    CAS  PubMed  Google Scholar 

  45. Wulfkuhle J et al. (2004) Genomic and proteomic technologies for individualisation and improvement of cancer treatment. Eur J Cancer 40: 2623–2632

    CAS  PubMed  Google Scholar 

  46. Espina V et al. (2004) Use of proteomic analysis to monitor responses to biological therapies. Expert Opin Biol Ther 4: 83–93

    CAS  PubMed  Google Scholar 

  47. Posadas EM et al. (2004) Proteomics and ovarian cancer: implications for diagnosis and treatment: a critical review of the recent literature. Curr Opin Oncol 16: 478–484

    CAS  PubMed  Google Scholar 

  48. Krause DS and Van Etten RA (2005) Tyrosine kinases as targets for cancer therapy. N Engl J Med 353: 172–187

    CAS  PubMed  Google Scholar 

  49. Awada A et al. (2004) New anticancer agents and therapeutic strategies in development for solid cancers: a clinical perspective. Expert Rev Anticancer Ther 4: 53–60

    CAS  PubMed  Google Scholar 

  50. Harari PM and Huang S (2004) Combining EGFR inhibitors with radiation or chemotherapy: will preclinical studies predict clinical results? Int J Radiat Oncol Biol Phys 58: 976–983

    CAS  PubMed  Google Scholar 

  51. Raben D et al. (2002) ZD1839, a selective epidermal growth factor receptor tyrosine kinase inhibitor, alone and in combination with radiation and chemotherapy as a new therapeutic strategy in non-small cell lung cancer. Semin Oncol 29: 37–46

    CAS  PubMed  Google Scholar 

  52. Araujo RP et al. (2005) A mathematical model of combination therapy using the EGFR signaling network. Biosystems 80: 57–69

    CAS  PubMed  Google Scholar 

  53. Araujo RP et al. (2004) Network-targeted combination therapy: a new concept in cancer treatment. Drug Disc Today 1: 425–433

    CAS  Google Scholar 

  54. Arteaga CL and Baselga J (2003) Clinical trial design and end points for epidermal growth factor receptor-targeted therapies: implications for drug development and practice. Clin Cancer Res 9: 1579–1589

    CAS  PubMed  Google Scholar 

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

    CAS  Google Scholar 

  56. Jain KK (2005) Personalised medicine for cancer: from drug development into clinical practice. Expert Opin Pharmacother 6: 1463–1476

    CAS  PubMed  Google Scholar 

  57. von Mehren M (2003) Gastrointestinal stromal tumors: a paradigm for molecularly targeted therapy. Cancer Invest 21: 553–563

    CAS  PubMed  Google Scholar 

  58. Wiestner A and Staudt LM (2003) Towards molecular diagnosis and targeted therapy of lymphoid malignancies. Semin Hematol 40: 296–307

    CAS  PubMed  Google Scholar 

  59. Hynes NE and Lane HA (2005) ERBB receptors and cancer: the complexity of targeted inhibitors. Nat Rev Cancer 5: 341–354

    CAS  PubMed  Google Scholar 

  60. Emens LA (2005) Trastuzumab: targeted therapy for the management of HER-2/neu-overexpressing metastatic breast cancer. Am J Therapeutics 12: 243–253

    Google Scholar 

  61. Dowsett M (2003) Correlation between immunohistochemistry (HercepTest) and fluorescence in situ hybridization (FISH) for HER2 in 426 breast carcinomas from 37 centres. J Pathol 199: 418–423

    CAS  PubMed  Google Scholar 

  62. Vogel DL et al. (2002) Efficacy and safety of trastuzumab as a single agent in first-line treatment of HER2-overexpressing metastatic breast cancer. J Clin Oncol 20: 719–726

    CAS  PubMed  Google Scholar 

  63. DiGiovanna MP et al. (2005) Relationship of epidermal growth factor receptor expression to ErbB-2 signaling activity and prognosis in breast cancer patients. J Clin Oncol 23: 1152–1160

    CAS  PubMed  Google Scholar 

  64. Thor AD et al. (2000) Activation (tyrosine phosphorylation) of ErbB-2 (HER-2/neu): a study of incidence and correlation with outcome in breast cancer. J Clin Oncol 18: 3230–3239

    CAS  PubMed  Google Scholar 

  65. Tripathy D (2005) Targeted therapies in breast cancer. Breast J 11 (Suppl 1): S30–S35

    PubMed  Google Scholar 

  66. Engelman JA and Janne PA (2005) Factors predicting response to EGFR tyrosine kinase inhibitors. Semin Resp Crit Care Med 26: 314–322

    Google Scholar 

  67. Giaccone G (2005) Epidermal growth factor receptor inhibitors in the treatment of non-small-cell lung cancer. J Clin Oncol 23: 3235–3242

    CAS  PubMed  Google Scholar 

  68. Baselga J et al. (2002) Phase I safety, pharmacokinetic, and pharmacodynamic trial of ZD1839, a selective, oral epidermal growth factor receptor tyrosine kinase inhibitor, in patients with five selected solid tumor types. J Clin Oncol 20: 4292–4302

    CAS  PubMed  Google Scholar 

  69. Herbst RS et al. (2002) Selective oral epidermal growth factor receptor tyrosine kinase inhibitor ZD1839 is generally well-tolerated and has activity in non-small-cell lung cancer and other solid tumors: results of a phase I trial. J Clin Oncol 20: 3815–3825

    CAS  PubMed  Google Scholar 

  70. Fukuoka M et al. (2003) Multi-institutional randomized phase II trial of gefitinib for previously treated patients with advanced non-small-cell lung cancer. J Clin Oncol 21: 2237–2246

    CAS  PubMed  Google Scholar 

  71. Kris MG et al. (2003) Efficacy of gefitinib, an inhibitor of the epidermal growth factor receptor tyrosine kinase, in symptomatic patients with non-small cell lung cancer: a randomized trial. JAMA 290: 2149–2158

    CAS  PubMed  Google Scholar 

  72. Bailey R et al. (2003) Gefitinib, (“Iressa”, ZD1839) monotherapy for pretreated advance non-small cell lung cancer in IDEAL 1 and 2: tumor response is not clinically relevantly predictable from tumor EGFR membrane staining alone. Lung Cancer 41: 71

    Google Scholar 

  73. Giaccone G et al. (2004) Gefitinib in combination with gemcitabine and cisplatin in advanced non-small-cell lung cancer: a phase III trial—INTACT 1. J Clin Oncol 22: 777–784

    CAS  PubMed  Google Scholar 

  74. Lynch TJ et al. (2004) Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med 350: 2129–2139

    CAS  PubMed  Google Scholar 

  75. Paez JG et al. (2004) EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science 304: 1497–1500

    CAS  PubMed  Google Scholar 

  76. Cappuzzo F et al. (2004) Akt phosphorylation and gefitinib efficacy in patients with advance non-small-cell lung cancer. J Natl Cancer Inst 96: 1133–1141

    CAS  PubMed  Google Scholar 

  77. Narayanan S (2000) The preanalytic phase: an important component of laboratory medicine. Am J Clin Pathol 113: 429–452

    CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Julia D Wulfkuhle or Emanuel F Petricoin III.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Wulfkuhle, J., Edmiston, K., Liotta, L. et al. Technology Insight: pharmacoproteomics for cancer—promises of patient-tailored medicine using protein microarrays. Nat Rev Clin Oncol 3, 256–268 (2006). https://doi.org/10.1038/ncponc0485

Download citation

  • Received:

  • Accepted:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncponc0485

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