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:

Mechanisms of Disease: oncogene addiction—a rationale for molecular targeting in cancer therapy

Abstract

There has been considerable progress in the systemic treatment of cancer because of the rapid development and clinical application of molecular targeted agents. Although patients with a particular type and stage of cancer are often treated as a single group, more-specific therapy is being considered, as subsets of these patients who are more likely to benefit from treatment with particular agents are being identified. We previously introduced the concept of 'oncogene addiction' to explain how some cancers that contain multiple genetic, epigenetic, and chromosomal abnormalities are dependent on or 'addicted' to one or a few genes for both maintenance of the malignant phenotype and cell survival. Thus, reversal of only one or a few of these abnormalities can inhibit cancer cell growth and in some cases translate to improved survival rates. This review summarizes current experimental and clinical evidence for the concept of oncogene addiction and describes molecular mechanisms that may explain this phenomenon. In addition, we discuss how high-throughput screening methods, including gene-expression profiling and proteomics, and emerging methods for analyzing complex cellular networks can be used to identify the state of oncogene addiction, i.e. the 'Achilles' heel,' in specific cancers. Finally, we discuss the use of molecular targeted agents in combination with other anticancer agents as a strategy to optimize therapy and prevent disease recurrence.

Key Points

  • 'Oncogene addiction' describes the phenomenon by which some cancers that contain multiple genetic and epigenetic abnormalities remain dependent on (addicted to) one or a few genes for both maintenance of the malignant phenotype and cell survival

  • Evidence that supports the concept of oncogene addiction has now been obtained in three diverse systems: 1) genetically engineered mouse models of human cancer, 2) mechanistic studies in human cancer cell lines, and 3) clinical trials with specific molecular targeted agents

  • 'Network theory' and other techniques of systems biology may provide methods for analyzing the entire circuitry of cancer cells and thus facilitate identification of pathways of oncogene addiction in specific types of human cancer

  • These insights will guide the development and clinical application of novel molecular targeted agents

  • Treatment regimens that combine molecular targeted agents with other anticancer agents could provide the optimal strategy for treating and preventing cancer

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

Similar content being viewed by others

References

  1. Weinstein IB et al. (1997) Disorders in cell circuitry associated with multistage carcinogenesis: exploitable targets for cancer prevention and therapy. Clin Cancer Res 3: 2696–2702

    CAS  PubMed  Google Scholar 

  2. Weinstein IB (2000) Disorders in cell circuitry during multistage carcinogenesis: the role of homeostasis. Carcinogenesis 21: 857–864

    Article  CAS  Google Scholar 

  3. Weinstein IB (2002) Cancer. Addiction to oncogenes—the Achilles heal of cancer. Science 297: 63–64

    Article  CAS  Google Scholar 

  4. Sawyers C (2004) Targeted cancer therapy. Nature 432: 294–297

    Article  CAS  Google Scholar 

  5. Kaelin WG Jr (2005) The concept of synthetic lethality in the context of anticancer therapy. Nat Rev Cancer 5: 689–698

    Article  CAS  Google Scholar 

  6. Felsher DW and Bishop JM (1999) Reversible tumorigenesis by MYC in hematopoietic lineages. Mol Cell 4: 199–207

    Article  CAS  Google Scholar 

  7. Huettner CS et al. (2000) Reversibility of acute B-cell leukaemia induced by BCR-ABL1. Nat Genet 24: 57–60

    Article  CAS  Google Scholar 

  8. Chin L et al. (1999) Essential role for oncogenic Ras in tumour maintenance. Nature 400: 468–472

    Article  CAS  Google Scholar 

  9. Jackson EL et al. (2001) Analysis of lung tumor initiation and progression using conditional expression of oncogenic K-ras. Genes Dev 15: 3243–3248

    Article  CAS  Google Scholar 

  10. Pelengaris S et al. (2002) Suppression of Myc-induced apoptosis in beta cells exposes multiple oncogenic properties of Myc and triggers carcinogenic progression. Cell 109: 321–334

    Article  CAS  Google Scholar 

  11. Jain M et al. (2002) Sustained loss of a neoplastic phenotype by brief inactivation of MYC. Science 297: 102–104

    Article  CAS  Google Scholar 

  12. Moody SE et al. (2002) Conditional activation of Neu in the mammary epithelium of transgenic mice results in reversible pulmonary metastasis. Cancer Cell 2: 451–461

    Article  CAS  Google Scholar 

  13. Moody SE et al. (2005) The transcriptional repressor Snail promotes mammary tumor recurrence. Cancer Cell 8: 197–209

    Article  CAS  Google Scholar 

  14. D'Cruz CM et al. (2001) c-MYC induces mammary tumorigenesis by means of a preferred pathway involving spontaneous Kras2 mutations. Nat Med 7: 235–239

    Article  CAS  Google Scholar 

  15. Gunther EJ et al. (2003) Impact of p53 loss on reversal and recurrence of conditional Wnt-induced tumorigenesis. Genes Dev 17: 488–501

    Article  CAS  Google Scholar 

  16. Colomer R et al. (1994) erbB-2 antisense oligonucleotides inhibit the proliferation of breast carcinoma cells with erbB-2 oncogene amplification. Br J Cancer 70: 819–825

    Article  CAS  Google Scholar 

  17. Zhou P et al. (1995) Antisense to cyclin D1 inhibits growth and reverses the transformed phenotype of human esophageal cancer cells. Oncogene 11: 571–580

    CAS  PubMed  Google Scholar 

  18. Arber N et al. (1997) Antisense to cyclin D1 inhibits the growth and tumorigenicity of human colon cancer cells. Cancer Res 57: 1569–1574

    CAS  PubMed  Google Scholar 

  19. Kornmann, M et al. (1999) Inhibition of cyclin D1 expression in human pancreatic cancer cells is associated with increased chemosensitivity and decreased expression of multiple chemoresistance genes. Cancer Res 59: 3505–3511

    CAS  PubMed  Google Scholar 

  20. Sauter ER et al. (1999) Antisense cyclin D1 induces apoptosis and tumor shrinkage in human squamous carcinomas. Cancer Res 59: 4876–4881

    CAS  PubMed  Google Scholar 

  21. Hui AB et al. (2005) Array-based comparative genomic hybridization analysis identified cyclin D1 as a target oncogene at 11q13.3 in nasopharyngeal carcinoma. Cancer Res 65: 8125–8133

    Article  CAS  Google Scholar 

  22. Aoki K et al. (1997) Suppression of Ki-ras p21 levels leading to growth inhibition of pancreatic cancer cell lines with Ki-ras mutation but not those without Ki-ras mutation. Mol Carcinog 20: 251–258

    Article  CAS  Google Scholar 

  23. Brummelkamp TR et al. (2002) Stable suppression of tumorigenicity by virus-mediated RNA interference. Cancer Cell 2: 243–247

    Article  CAS  Google Scholar 

  24. Verma UN et al. (2003) Small interfering RNAs directed against beta-catenin inhibit the in vitro and in vivo growth of colon cancer cells. Clin Cancer Res 9: 1291–1300

    CAS  PubMed  Google Scholar 

  25. Li K et al. (2003) Use of RNA interference to target cyclin E-overexpressing hepatocellular carcinoma. Cancer Res 63: 3593–3597

    CAS  PubMed  Google Scholar 

  26. Sharma A et al. (2005) Mutant V599EB-Raf regulates growth and vascular development of malignant melanoma tumors. Cancer Res 65: 2412–2421

    Article  CAS  Google Scholar 

  27. Miller AJ et al. (2004) Transcriptional regulation of the melanoma prognostic marker melastatin (TRPM1) by MITF in melanocytes and melanoma. Cancer Res 64: 509–516

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  30. Baselga J and Albanell J (2001) Mechanism of action of anti-HER2 monoclonal antibodies. Ann Oncol 12 (Suppl 1): S35–S41

    Article  Google Scholar 

  31. Hughes TP et al. (2003) Frequency of major molecular responses to imatinib or interferon alfa plus cytarabine in newly diagnosed chronic myeloid leukemia. N Engl J Med 349: 1423–1432

    Article  CAS  Google Scholar 

  32. Shepherd FA et al. (2005) Erlotinib in previously treated non-small-cell lung cancer. N Engl J Med 353: 123–132

    Article  CAS  Google Scholar 

  33. Demetri GD et al. (2002) Efficacy and safety of imatinib mesylate in advanced gastrointestinal stromal tumors. N Engl J Med 347: 472–480

    Article  CAS  Google Scholar 

  34. Moore MJ (2005) Brief communication: a new combination in the treatment of advanced pancreatic cancer. Semin Oncol 32: 5–6

    Article  Google Scholar 

  35. Taron M et al. (2005) Activating mutations in the tyrosine kinase domain of the epidermal growth factor receptor are associated with improved survival in gefitinib-treated chemorefractory lung adenocarcinomas. Clin Cancer Res 11: 5878–5885

    Article  CAS  Google Scholar 

  36. 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

    Article  CAS  Google Scholar 

  37. Pao W et al. (2004) EGF receptor gene mutations are common in lung cancers from “never smokers” and are associated with sensitivity of tumors to gefitinib and erlotinib. Proc Natl Acad Sci USA 101: 13306–13311

    Article  CAS  Google Scholar 

  38. Mellinghoff IK et al. (2005) Molecular determinants of the response of glioblastomas to EGFR kinase inhibitors. N Engl J Med 353: 2012–2024

    Article  CAS  Google Scholar 

  39. Baselga J et al. (2005) Phase II multicenter study of the antiepidermal growth factor receptor monoclonal antibody cetuximab in combination with platinum-based chemotherapy in patients with platinum-refractory metastatic and/or recurrent squamous cell carcinoma of the head and neck. J Clin Oncol 23: 5568–5577

    Article  CAS  Google Scholar 

  40. Cunningham D et al. (2004) Cetuximab monotherapy and cetuximab plus irinotecan in irinotecan-refractory metastatic colorectal cancer. N Engl J Med 351: 337–345

    Article  CAS  Google Scholar 

  41. Miller KD et al. (2005) Randomized phase III trial of capecitabine compared with bevacizumab plus capecitabine in patients with previously treated metastatic breast cancer. J Clin Oncol 23: 792–799

    Article  CAS  Google Scholar 

  42. Hurwitz H et al. (2004) Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med 350: 2335–2342

    Article  CAS  Google Scholar 

  43. Yang JC et al. (2003) A randomized trial of bevacizumab, an anti-vascular endothelial growth factor antibody, for metastatic renal cancer. N Engl J Med 349: 427–434

    Article  CAS  Google Scholar 

  44. Gorre ME et al. (2001) Clinical resistance to STI-571 cancer therapy caused by BCR-ABL gene mutation or amplification. Science 293: 876–880

    Article  CAS  Google Scholar 

  45. Kobayashi S et al. (2005) EGFR mutation and resistance of non-small-cell lung cancer to gefitinib. N Engl J Med 352: 786–792

    Article  CAS  Google Scholar 

  46. La Rosee P et al. (2002) Activity of the Bcr-Abl kinase inhibitor PD180970 against clinically relevant Bcr-Abl isoforms that cause resistance to imatinib mesylate (Gleevec®, STI571). Cancer Res 62: 7149–7153

    CAS  PubMed  Google Scholar 

  47. Kerr JF et al. (1994) Apoptosis. Its significance in cancer and cancer therapy. Cancer 73: 2013–2026

    Article  CAS  Google Scholar 

  48. Mills GB et al. (2001) Linking molecular therapeutics to molecular diagnostics: inhibition of the FRAP/RAFT/TOR component of the PI3K pathway preferentially blocks PTEN mutant cells in vitro and in vivo. Proc Natl Acad Sci USA 98: 10031–10033

    Article  CAS  Google Scholar 

  49. Sordella R et al. (2004) Gefitinib-sensitizing EGFR mutations in lung cancer activate anti-apoptotic pathways. Science 305: 1163–1167

    Article  CAS  Google Scholar 

  50. Cappuzzo F et al. (2005) Epidermal growth factor receptor gene and protein and gefitinib sensitivity in non-small-cell lung cancer. J Natl Cancer Inst 97: 643–655

    Article  CAS  Google Scholar 

  51. Dowell JE and Minna JD (2004) The impact of epidermal-growth-factor-receptor mutations in response to lung-cancer therapy. Nat Clin Pract Oncol 1: 2–3

    Article  Google Scholar 

  52. Paddison PJ et al. (2004) A resource for large-scale RNA-interference-based screens in mammals. Nature 428: 427–431

    Article  CAS  Google Scholar 

  53. Zhang SZ et al. (2005) Knockdown of c-Met by adenovirus-delivered small interfering RNA inhibits hepatocellular carcinoma growth in vitro and in vivo. Mol Cancer Ther 4: 1577–1584

    Article  CAS  Google Scholar 

  54. Varga AC and Wrana JL (2005) The disparate role of BMP in stem cell biology. Oncogene 24: 5713–5721

    Article  CAS  Google Scholar 

  55. Stadler WM (2005) Targeted agents for the treatment of advanced renal cell carcinoma. Cancer 104: 2323–2333

    Article  CAS  Google Scholar 

  56. Garraway LA et al. (2005) Integrative genomic analyses identify MITF as a lineage survival oncogene amplified in malignant melanoma. Nature 436: 117–122

    Article  CAS  Google Scholar 

  57. Berman, DM et al. (2002) Medulloblastoma growth inhibition by hedgehog pathway blockade. Science 297: 1559–1561

    Article  CAS  Google Scholar 

  58. Radtke F and Clevers H (2005) Self-renewal and cancer of the gut: two sides of a coin. Science 307: 1904–1909

    Article  CAS  Google Scholar 

  59. Ottino JM (2004) Engineering complex systems. Nature 427: 399

    Article  CAS  Google Scholar 

  60. Bray D (2003) Molecular networks: the top-down view. Science 301: 1864–1865

    Article  CAS  Google Scholar 

  61. Alon U (2003) Biological networks: the tinkerer as an engineer. Science 301: 1866–1867

    Article  CAS  Google Scholar 

  62. Huang S (1999) Gene expression profiling, genetic networks, and cellular states: an integrating concept for tumorigenesis and drug discovery. J Mol Med 77: 469–480

    Article  CAS  Google Scholar 

  63. Baak JP et al. (2005) Genomics and proteomics—the way forward. Ann Oncol 16 (Suppl 2): ii30–44

    Article  Google Scholar 

  64. Wittekind C and Neid M (2005) Cancer invasion and metastasis. Oncology 69 (Suppl 1): 14–16

    Article  Google Scholar 

  65. Cheng JQ et al. (2005) The Akt/PKB pathway: molecular target for cancer drug discovery. Oncogene 24: 7482–7492

    Article  CAS  Google Scholar 

  66. Bergers G and Benjamin LE (2003) Tumorigenesis and the angiogenic switch. Nat Rev Cancer 3: 401–410

    Article  CAS  Google Scholar 

  67. Jonkers J and Berns A (2004) Oncogene addiction: sometimes a temporary slavery. Cancer Cell 6: 535–538

    CAS  PubMed  Google Scholar 

  68. Fan QW et al. (2003) Combinatorial efficacy achieved through two-point blockade within a signaling pathway—a chemical genetic approach. Cancer Res 63: 8930–8938

    CAS  PubMed  Google Scholar 

  69. Frantz S (2005) Drug discovery: playing dirty. Nature 437: 942–943

    Article  CAS  Google Scholar 

  70. Whitesell L and Lindquist SL (2005) HSP90 and the chaperoning of cancer. Nat Rev Cancer 5: 761–772

    Article  CAS  Google Scholar 

  71. Adams J (2004) The development of proteasome inhibitors as anticancer drugs. Cancer Cell 5: 417–421

    Article  CAS  Google Scholar 

  72. McCormick F (2001) Cancer gene therapy: fringe or cutting edge? Nat Rev Cancer 1: 130–141

    Article  CAS  Google Scholar 

  73. Egger G et al. (2004) Epigenetics in human disease and prospects for epigenetic therapy. Nature 429: 457–463

    Article  CAS  Google Scholar 

  74. Balch C et al. (2005) Antimitogenic and chemosensitizing effects of the methylation inhibitor zebularine in ovarian cancer. Mol Cancer Ther 4: 1505–1514

    Article  CAS  Google Scholar 

  75. Sandor V et al. (2002) Phase I trial of the histone deacetylase inhibitor, depsipeptide (FR901228, NSC 630176), in patients with refractory neoplasms. Clin Cancer Res 8: 718–728

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by awards to IBW from the National Foundation for Cancer Research, the Entertainment Industry–National Colorectal Research Alliance, and the TJ Martell Foundation, and awards to AKJ from the National Cancer Institute (5K23CA101669) and the National Institutes of Health (MO1-RR00645).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to I Bernard Weinstein.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Weinstein, I., Joe, A. Mechanisms of Disease: oncogene addiction—a rationale for molecular targeting in cancer therapy. Nat Rev Clin Oncol 3, 448–457 (2006). https://doi.org/10.1038/ncponc0558

Download citation

  • Received:

  • Accepted:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncponc0558

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