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:

Predictive biomarkers: a paradigm shift towards personalized cancer medicine

Abstract

Advances in our understanding of the intricate molecular mechanisms for transformation of a normal cell to a cancer cell, and the aberrant control of complementary pathways, have presented a much more complex set of challenges for the diagnostic and therapeutic disciplines than originally appreciated. The oncology field has entered an era of personalized medicine where treatment selection for each cancer patient is becoming individualized or customized. This advance reflects the molecular and genetic composition of the tumors and progress in biomarker technology, which allow us to align the most appropriate treatment according to the patient's disease. There is a worldwide acceptance that advances in our ability to identify predictive biomarkers and provide them as companion diagnostics for stratifying and subgrouping patients represents the next leap forward in improving the quality of clinical care in oncology. As such, we are progressing from a population-based empirical 'one drug fits all' treatment model, to a focused personalized approach where rational companion diagnostic tests support the drug's clinical utility by identifying the most responsive patient subgroup.

Key Points

  • Cancer is a diverse collection of diseases that have different causative factors, molecular composition, and natural histories

  • Many recently developed cancer drugs target discrete molecular aberrations or pathways in tumor cells and consequently are active on a subset of the patient population

  • Companion diagnostics that measure biomarkers that allow responsive patients to be identified and subgrouped are being increasingly integrated with the drug-development process and clinical trials

  • Most response-specific biomarkers that have reached clinical validation were identified through retrospective analysis of clinical data

  • Molecular techniques are available that allow biomarkers to be identified in a systematic prospectively driven fashion

  • The long sought after goal where therapeutic choice is guided by an informative biomarker 'code' is now upon us

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: Action of drugs targeting EGFR and HER2.
Figure 2: Design of biomarker testing clinical trials.
Figure 3: Response-specific biomarkers in cancer clinical trials.
Figure 4: A theoretical approach to personalized cancer therapy.

Similar content being viewed by others

References

  1. American Cancer Society. Lifetime Risk of Developing or Dying From Cancer [online], (2010).

  2. Yabroff, K. R., Warren, J. L. & Brown, M. L. Costs of cancer care in the USA: a descriptive review. Nat. Clin. Pract. Oncol. 4, 643–656 (2007).

    Article  PubMed  Google Scholar 

  3. Azorsa, D. O. et al. Synthetic lethal RNAi screening identifies sensitizing targets for gemcitabine therapy in pancreatic cancer. J. Transl. Med. 7, 43 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  4. Schilsky, R. L. Personalized medicine in oncology: the future is now. Nat. Rev. Drug Discov. 9, 363–366 (2010).

    Article  CAS  PubMed  Google Scholar 

  5. Fine, B. M. & Amler, L. Predictive biomarkers in the development of oncology drugs: a therapeutic industry perspective. Clin. Pharmacol. Ther. 85, 535–538 (2009).

    Article  CAS  PubMed  Google Scholar 

  6. Duffy, M. J. & Crown, J. A personalized approach to cancer treatment: how biomarkers can help. Clin. Chem. 54, 1770–1779 (2008).

    Article  CAS  PubMed  Google Scholar 

  7. Park, J. W. et al. Rationale for biomarkers and surrogate end points in mechanism-driven oncology drug development. Clin. Cancer Res. 10, 3885–3896 (2004).

    Article  CAS  PubMed  Google Scholar 

  8. Cronin, M. et al. Analytical validation of the Oncotype DX genomic diagnostic test for recurrence prognosis and therapeutic response prediction in node-negative, estrogen receptor-positive breast cancer. Clin. Chem. 53, 1084–1091 (2007).

    Article  CAS  PubMed  Google Scholar 

  9. Ward, D. G. et al. Identification of serum biomarkers for colon cancer by proteomic analysis. Br. J. Cancer 94, 1898–1905 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Sarker, D. & Workman, P. Pharmacodynamic biomarkers for molecular cancer therapeutics. Adv. Cancer Res. 96, 213–268 (2007).

    Article  CAS  PubMed  Google Scholar 

  11. Ratain, M. J., Schilsky, R. L., Conley, B. A. & Egorin, M. J. Pharmacodynamics in cancer therapy. J. Clin. Oncol. 8, 1739–1753 (1990).

    Article  CAS  PubMed  Google Scholar 

  12. August, J. Market watch: emerging companion diagnostics for cancer drugs. Nat. Rev. Drug Discov. 9, 351 (2010).

    Article  CAS  PubMed  Google Scholar 

  13. Sawyers, C. Targeted cancer therapy. Nature 432, 294–297 (2004).

    Article  CAS  PubMed  Google Scholar 

  14. Segal, N. H. & Saltz, L. B. Evolving treatment of advanced colon cancer. Annu. Rev. Med. 60, 207–219 (2009).

    Article  CAS  PubMed  Google Scholar 

  15. Badgwell, B. D. et al. Management of bevacizumab-associated bowel perforation: a case series and review of the literature. Ann. Oncol. 19, 577–582 (2008).

    Article  CAS  PubMed  Google Scholar 

  16. Goodsaid, F. & Papaluca, M. Evolution of biomarker qualification at the health authorities. Nat. Biotechnol. 28, 441–443 (2010).

    Article  CAS  PubMed  Google Scholar 

  17. Raftery, J. NICE and the challenge of cancer drugs. BMJ 338, b67 (2009).

    Article  PubMed  Google Scholar 

  18. Berns, K. et al. A functional genetic approach identifies the PI3K pathway as a major determinant of trastuzumab resistance in breast cancer. Cancer Cell 12, 395–402 (2007).

    Article  CAS  PubMed  Google Scholar 

  19. Slamon, D. J. et al. Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogene. Science 235, 177–182 (1987).

    Article  CAS  PubMed  Google Scholar 

  20. Dawood, S. et al. Prognosis of women with metastatic breast cancer by HER2 status and trastuzumab treatment: an institutional-based review. J. Clin. Oncol. 28, 92–98 (2010).

    Article  CAS  PubMed  Google Scholar 

  21. Slamon, D. J. et al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N. Engl. J. Med. 344, 783–792 (2001).

    Article  CAS  PubMed  Google Scholar 

  22. Hudis, C. A. Trastuzumab–mechanism of action and use in clinical practice. N. Engl. J. Med. 357, 39–51 (2007).

    Article  CAS  PubMed  Google Scholar 

  23. Bange, J., Zwick, E. & Ullrich, E. Molecular targets for breast cancer therapy and prevention. Nat. Med. 7, 548–552 (2001).

    Article  CAS  PubMed  Google Scholar 

  24. Romond, E. H. et al. Trastuzumab plus adjuvant chemotherapy for operable HER2-positive breast cancer. N. Engl. J. Med. 353, 1673–1684 (2005).

    Article  CAS  PubMed  Google Scholar 

  25. Piccart-Gebhart, M. J. et al. Trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer. N. Engl. J. Med. 353, 1659–1672 (2005).

    Article  CAS  PubMed  Google Scholar 

  26. Valabrega, G., Montemurro, F. & Aglietta, M. Trastuzumab: mechanism of action, resistance and future perspectives in HER2-overexpressing breast cancer. Ann. Oncol. 18, 977–984 (2007).

    Article  CAS  PubMed  Google Scholar 

  27. Suter, T. M. et al. Trastuzumab-associated cardiac adverse effects in the herceptin adjuvant trial. J. Clin. Oncol. 25, 3859–3865 (2007).

    Article  CAS  PubMed  Google Scholar 

  28. Muthuswamy, S. K. Trastuzumab resistance: all roads lead to SRC. Nat. Med. 17, 416–418 (2011).

    Article  CAS  PubMed  Google Scholar 

  29. Allison, M. The HER2 testing conundrum. Nat. Biotechnol. 28, 117–119 (2010).

    Article  CAS  PubMed  Google Scholar 

  30. Phillips, K. A. et al. Clinical practice patterns and cost effectiveness of human epidermal growth receptor 2 testing strategies in breast cancer patients. Cancer 115, 5166–5174 (2009).

    Article  PubMed  Google Scholar 

  31. Cuadros, M. & Villegas, M. Systematic review of HER2 breast cancer testing. Appl. Immunohistochem. Mol. Morphol. 17, 1–7 (2009).

    Article  CAS  PubMed  Google Scholar 

  32. Perez, E. A. et al. HER2 testing in patients with breast cancer: poor correlation between weak positivity by immunohistochemistry and gene amplification by fluorescence in situ hybridization. Mayo Clin. Proc. 77, 148–154 (2002).

    Article  PubMed  Google Scholar 

  33. Stone, R. M. Optimizing treatment of chronic myeloid leukemia: a rational approach. Oncologist 9, 259–270 (2004).

    Article  CAS  PubMed  Google Scholar 

  34. Deininger, M. W. & Druker, B. J. Specific targeted therapy of chronic myelogenous leukemia with imatinib. Pharmacol. Rev. 55, 401–423 (2003).

    Article  CAS  PubMed  Google Scholar 

  35. Wodarz, D. Heterogeneity in chronic myeloid leukaemia dynamics during imatinib treatment: role of immune responses. Proc. Biol. Sci. 277, 1875–1880 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Goozner, M. Drug developers unveil strategies aimed at imatinib-resistant CML. J. Natl Cancer Inst. 102, 593–595 (2010).

    Article  CAS  PubMed  Google Scholar 

  37. Breccia, M. Hematology: Nilotinib and dasatinib–new 'magic bullets' for CML? Nat. Rev. Clin. Oncol. 7, 557–558 (2010).

    Article  CAS  PubMed  Google Scholar 

  38. Fuerst, M. L. FDA approves dasatinib for imatinib resistance and intolerance 3 weeks after enthusiastic recommendation from ODAC. Oncol. Times 28, 9–10 (2006).

    Article  Google Scholar 

  39. Kantarjian, H. et al. Dasatinib versus imatinib in newly diagnosed chronic-phase chronic myeloid leukemia. N. Engl. J. Med. 362, 2260–2270 (2010).

    Article  CAS  PubMed  Google Scholar 

  40. Breccia, M. & Alimena, G. Nilotinib: a second-generation tyrosine kinase inhibitor for chronic myeloid leukemia. Leuk. Res. 34, 129–134 (2010).

    Article  CAS  PubMed  Google Scholar 

  41. Kantarjian, H. et al. Dasatinib. Nat. Rev. Drug Discov. 5, 717–718 (2006).

    Article  CAS  PubMed  Google Scholar 

  42. Sawyers, C. L. Chronic myeloid leukemia. N. Engl. J. Med. 340, 1330–1340 (1999).

    Article  CAS  PubMed  Google Scholar 

  43. Harris, T. Gene and drug matrix for personalized cancer therapy. Nat. Rev. Drug Discov. 9, 660 (2010).

    Article  PubMed  CAS  Google Scholar 

  44. Weisberg, E., Manley, P. W., Cowan-Jacob, S. W., Hochhaus, A. & Griffin, J. D. Second generation inhibitors of BCR-ABL for the treatment of imatinib-resistant chronic myeloid leukaemia. Nat. Rev. Cancer 7, 345–356 (2007).

    Article  CAS  PubMed  Google Scholar 

  45. Terasawa, T., Dahabreh, I. & Trikalinos, T. A. BCR-ABL mutation testing to predict response to tyrosine kinase inhibitors in patients with chronic myeloid leukemia. PLoS Curr. 2, RRN1204 (2010).

    PubMed  Google Scholar 

  46. Papadopoulos, N., Kinzler, K. W. & Vogelstein, B. The role of companion diagnostics in the development and use of mutation-targeted cancer therapies. Nat. Biotechnol. 24, 985–995 (2006).

    Article  CAS  PubMed  Google Scholar 

  47. Siddiqui, M. A. & Scott, L. J. Imatinib: a review of its use in the management of gastrointestinal stromal tumours. Drugs 67, 805–820 (2007).

    Article  CAS  PubMed  Google Scholar 

  48. Heinrich, M. C. Imatinib treatment of metastatic GIST: don't stop (believing). Lancet Oncol. 11, 910–911 (2010).

    Article  PubMed  Google Scholar 

  49. Heinrich, M. C. et al. Kinase mutations and imatinib response in patients with metastatic gastrointestinal stromal tumor. J. Clin. Oncol. 21, 4342–4349 (2003).

    Article  CAS  PubMed  Google Scholar 

  50. Ciardiello, F. & Tortora, G. EGFR antagonists in cancer treatment. N. Engl. J. Med. 358, 1160–1174 (2008).

    Article  CAS  PubMed  Google Scholar 

  51. Saltz, L. B. et al. Phase II trial of cetuximab in patients with refractory colorectal cancer that expresses the epidermal growth factor receptor. J. Clin. Oncol. 22, 1201–1208 (2004).

    Article  CAS  PubMed  Google Scholar 

  52. Lenz, H. J. et al. Multicenter phase II and translational study of cetuximab in metastatic colorectal carcinoma refractory to irinotecan, oxaliplatin, and fluoropyrimidines. J. Clin. Oncol. 24, 4914–4921 (2006).

    Article  CAS  PubMed  Google Scholar 

  53. Santini, D. Molecular predictive factors of response to anti-EGFR antibodies in colorectal cancer patients. Eur. J. Cancer Suppl. 6, 86–90 (2008).

    Article  Google Scholar 

  54. Saif, M. W. Colorectal cancer in review: the role of the EGFR pathway. Expert Opin. Investig. Drugs 19, 357–369 (2010).

    Article  CAS  PubMed  Google Scholar 

  55. Lièvre, A., Blons, H. & Laurent-Puig, P. Oncogenic mutations as predictive factors in colorectal cancer. Oncogene 29, 3033–3043 (2010).

    Article  PubMed  CAS  Google Scholar 

  56. Linardou, H. et al. Assessment of somatic k-RAS mutations as a mechanism associated with resistance to EGFR-targeted agents: a systematic review and meta-analysis of studies in advanced non-small-cell lung cancer and metastatic colorectal cancer. Lancet Oncol. 9, 962–972 (2008).

    Article  CAS  PubMed  Google Scholar 

  57. Karapetis, C. S. et al. K-ras mutations and benefit from cetuximab in advanced colorectal cancer. N. Engl. J. Med. 359, 1757–1765 (2008).

    Article  CAS  PubMed  Google Scholar 

  58. Shankaran, V., Obel, J. & Benson, A. B. 3rd. Predicting response to EGFR inhibitors in metastatic colorectal cancer: current practice and future directions. Oncologist 15, 157–167 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Mack, G. S. FDA holds court on post hoc data linking KRAS status to drug response. Nat. Biotechnol. 27, 110–112 (2009).

    Article  CAS  PubMed  Google Scholar 

  60. Wolff, A. C. et al. American Society of Clinical Oncology/College of American Pathologists guideline recommendations for human epidermal growth factor receptor 2 testing in breast cancer. J. Clin. Oncol. 25, 118–145 (2007).

    Article  CAS  PubMed  Google Scholar 

  61. Gridelli, C. et al. Erlotinib in non-small cell lung cancer treatment: current status and future development. Oncologist 12, 840–849 (2007).

    Article  CAS  PubMed  Google Scholar 

  62. Oxnard, G. R. & Miller, V. A. Use of erlotinib or gefitinib as initial therapy in advanced NSCLC. Oncology (Williston Park) 24, 392–399 (2010).

    Google Scholar 

  63. Saijo, N. Targeted therapies: Tyrosine-kinase inhibitors–new standard for NSCLC therapy. Nat. Rev. Clin. Oncol. 7, 618–619 (2010).

    Article  CAS  PubMed  Google Scholar 

  64. Maemondo, M. et al. Gefitinib or chemotherapy for non-small-cell lung cancer with mutated EGFR. N. Engl. J. Med. 362, 2380–2388 (2010).

    Article  CAS  PubMed  Google Scholar 

  65. Mitsudomi, T. et al. Gefitinib versus cisplatin plus docetaxel in patients with non-small-cell lung cancer harbouring mutations of the epidermal growth factor receptor (WJTOG3405): an open label, randomised phase 3 trial. Lancet Oncol. 11, 121–128 (2010).

    Article  CAS  PubMed  Google Scholar 

  66. Lopez-Chavez, A. & Giaccone, G. Targeted therapies: Importance of patient selection for EGFR TKIs in lung cancer. Nat. Rev. Clin. Oncol. 7, 360–362 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Massarelli, E. et al. KRAS mutation is an important predictor of resistance to therapy with epidermal growth factor receptor tyrosine kinase inhibitors in non-small-cell lung cancer. Clin. Cancer Res. 13, 2890–2896 (2007).

    Article  CAS  PubMed  Google Scholar 

  68. Sasaki, T., Rodig, S. J., Chirieac, L. R. & Jänne, P. A. The biology and treatment of EML4-ALK non-small cell lung cancer. Eur. J. Cancer 46, 1773–1780 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Butrynski, J. E. et al. Crizotinib in ALK-rearranged inflammatory myofibroblastic tumor. N. Engl. J. Med. 363, 1727–1733 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. De Witt Hamer, P. C. Small molecule kinase inhibitors in glioblastoma: a systematic review of clinical studies. Neuro. Oncol. 12, 304–316 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Mellinghoff, I. K., Cloughesy, T. F. & Mischel, P. S. PTEN-mediated resistance to epidermal growth factor receptor kinase inhibitors. Clin. Cancer Res. 13, 378–381 (2007).

    Article  CAS  PubMed  Google Scholar 

  72. Mellinghoff, I. K. et al. Molecular determinants of the response of glioblastomas to EGFR kinase inhibitors. N. Engl. J. Med. 353, 2012–2024 (2005).

    Article  CAS  PubMed  Google Scholar 

  73. Diverio, D., Riccioni, R., Mandelli, F. & Lo Coco, F. The PML/RAR alpha fusion gene in the diagnosis and monitoring of acute promyelocytic leukemia. Haematologica 80, 155–160 (1995).

    CAS  PubMed  Google Scholar 

  74. Soprano, D. R., Qin, P. & Soprano, K. J. Retinoic acid receptors and cancers. Annu. Rev. Nutr. 24, 201–221 (2004).

    Article  CAS  PubMed  Google Scholar 

  75. Lin, R. J. & Evans, R. M. Acquisition of oncogenic potential by RAR chimeras in acute promyelocytic leukemia through formation of homodimers. Mol. Cell 5, 821–830 (2000).

    Article  CAS  PubMed  Google Scholar 

  76. Freemantle, S. J., Spinella, M. J. & Dmitrovsky, E. Retinoids in cancer therapy and chemoprevention: promise meets resistance. Oncogene 22, 7305–7315 (2003).

    Article  CAS  PubMed  Google Scholar 

  77. Zhou, D. C. et al. Frequent mutations in the ligand-binding domain of PML-RARalpha after multiple relapses of acute promyelocytic leukemia: analysis for functional relationship to response to all-trans retinoic acid and histone deacetylase inhibitors in vitro and in vivo. Blood 99, 1356–1363 (2002).

    Article  CAS  PubMed  Google Scholar 

  78. Tang, X. H. & Gudas, L. J. Retinoids, retinoic acid receptors, and cancer. Annu. Rev. Pathol. 6, 345–364 (2011).

    Article  CAS  PubMed  Google Scholar 

  79. Kuendgen, A. et al. The histone deacetylase (HDAC) inhibitor valproic acid as monotherapy or in combination with all-trans retinoic acid in patients with acute myeloid leukemia. Cancer 106, 112–119 (2006).

    Article  CAS  PubMed  Google Scholar 

  80. De los Santos, M., Zambrano, A., Sánchez-Pacheco, A. & Aranda, A. Histone deacetylase inhibitors regulate retinoic acid receptor beta expression in neuroblastoma cells by both transcriptional and posttranscriptional mechanisms. Mol. Endocrinol. 21, 2416–2426 (2007).

    Article  CAS  PubMed  Google Scholar 

  81. Wooster, R. & Weber, B. L. Breast and ovarian cancer. N. Engl. J. Med. 348, 2339–2347 (2003).

    Article  CAS  PubMed  Google Scholar 

  82. Kaelin, W. G. Jr. The concept of synthetic lethality in the context of anticancer therapy. Nat. Rev. Cancer 5, 689–698 (2005).

    Article  CAS  PubMed  Google Scholar 

  83. Ashworth, A. A synthetic lethal therapeutic approach: poly(ADP) ribose polymerase inhibitors for the treatment of cancers deficient in DNA double-strand break repair. J. Clin. Oncol. 26, 3785–3790 (2008).

    Article  CAS  PubMed  Google Scholar 

  84. Amé, J. C., Spenlehauer, C. & de Murcia, G. The PARP superfamily. Bioessays 26, 882–893 (2004).

    Article  PubMed  CAS  Google Scholar 

  85. Farmer, H. et al. Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy. Nature 434, 917–921 (2005).

    Article  CAS  PubMed  Google Scholar 

  86. Yap, T. A. et al. First in human phase I pharmacokinetic (PK) and pharmacodynamic (PD) study of KU-0059436 (Ku), a small molecule inhibitor of poly ADP-ribose polymerase (PARP) in cancer patients (p), including BRCA1/2 mutation carriers [abstract]. J. Clin. Oncol. 25 (Suppl.), a3529 (2007).

    Google Scholar 

  87. Ratnam, K. & Low, J. A. Current development of clinical inhibitors of poly(ADP-ribose) polymerase in oncology. Clin. Cancer Res. 13, 1383–1388 (2007).

    Article  CAS  PubMed  Google Scholar 

  88. Foulkes, W. D., Smith, I. E. & Rein-Filho, J. S. Triple-negative breast cancer. N. Engl. J. Med. 363, 1938–1948 (2010).

    Article  CAS  PubMed  Google Scholar 

  89. O'Shaughnessy, J. et al. Iniparib plus chemotherapy in metastatic triple-negative breast cancer. N. Engl. J. Med. 364, 205–214 (2011).

    Article  CAS  PubMed  Google Scholar 

  90. Bath, C. Phase III results for PARP inhibitor iniparib quell optimism about new option for triple-negative metastatic breast cancer. The Asco Post [online], (2011).

    Google Scholar 

  91. Hutchinson, L. Targeted therapies: PARP inhibitor olaparib is safe and effective in patients with BRCA1 and BRCA2 mutations. Nat. Rev. Clin. Oncol. 7, 549 (2010).

    Article  PubMed  Google Scholar 

  92. Miller, A. J. & Mihm, M. C. Jr. Melanoma. N. Engl. J. Med. 355, 51–65 (2006).

    Article  CAS  PubMed  Google Scholar 

  93. Flaherty, K. T. et al. Inhibition of mutated, activated BRAF in metastatic melanoma. N. Engl. J. Med. 363, 809–819 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Nazarian, R. et al. Melanomas acquire resistance to B-RAF(V600E) inhibition by RTK or N-RAS upregulation. Nature 468, 973–977 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Shi, H., Kong, X., Ribas, A. & Lo, R.S. Combinatorial treatments that overcome PDGFRβ-driven resistance of melanoma cells to V600E B-RAF inhibition. Cancer Res. 71, 5067–5074 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Mullenders, J. & Bernards, R. Loss-of-function genetic screens as a tool to improve the diagnosis and treatment of cancer. Oncogene 28, 4409–4420 (2009).

    Article  CAS  PubMed  Google Scholar 

  97. Cully, M., You, H., Levine, A. J. & Mak, T. W. Beyond PTEN mutations: the PI3K pathway as an integrator of multiple inputs during tumorigenesis. Nat. Rev. Cancer 6, 184–192 (2006).

    Article  CAS  PubMed  Google Scholar 

  98. Iorns, E. et al. Identification of CDK10 as an important determinant of resistance to endocrine therapy for breast cancer. Cancer Cell 13, 91–104 (2008).

    Article  CAS  PubMed  Google Scholar 

  99. Fotheringham, S. et al. Genome-wide loss-of-function screen reveals an important role for the proteasome in HDAC inhibitor-induced apoptosis. Cancer Cell 15, 57–66 (2009).

    Article  CAS  PubMed  Google Scholar 

  100. Khan, O. et al. HR23B is a biomarker for tumor sensitivity to HDAC inhibitor-based therapy. Proc. Natl Acad. Sci. USA 107, 6532–6537 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Stimson, L., Wood, V., Khan, O., Fotheringham, S. & La Thangue, N. B. HDAC inhibitor-based therapies and haematological malignancy. Ann. Oncol. 20, 1293–1302 (2009).

    Article  CAS  PubMed  Google Scholar 

  102. McConkey, D. Proteasome and HDAC: who's zooming who? Blood 116, 308–309 (2010).

    Article  CAS  PubMed  Google Scholar 

  103. Whitehurst, A. W. et al. Synthetic lethal screen identification of chemosensitizer loci in cancer cells. Nature 446, 815–819 (2007).

    Article  CAS  PubMed  Google Scholar 

  104. Shapiro, C. L. et al. Phase I trial of bortezomib (Velcade) in combination with paclitaxel in advanced solid tumor patients (pts) [abstract]. J. Clin. Oncol. 23 (Suppl.), a3104 (2005).

    Article  Google Scholar 

  105. Young, R. C. Cancer clinical trials–a chronic but curable crisis. N. Engl. J. Med. 363, 306–309 (2010).

    Article  CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Contributions

Both authors researched data for inclusion in the article. N. B. La Thangue contributed to the writing, editing and reviewing the manuscript before submission and during the reviewing process.

Corresponding author

Correspondence to Nicholas B. La Thangue.

Ethics declarations

Competing interests

N. B. La Thangue is a consultant, stock holder and patent holder for Celleron Therapeutics. D. J. Kerr is a consultant for Celleron Therapeutics.

Rights and permissions

Reprints and permissions

About this article

Cite this article

La Thangue, N., Kerr, D. Predictive biomarkers: a paradigm shift towards personalized cancer medicine. Nat Rev Clin Oncol 8, 587–596 (2011). https://doi.org/10.1038/nrclinonc.2011.121

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrclinonc.2011.121

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer