Skip to main content

Molecular-Targeted Therapy for Cancer and Nanotechnology

  • Protocol
  • First Online:
Cancer Nanotechnology

Part of the book series: Methods in Molecular Biology ((MIMB,volume 624))

Abstract

In order to stop malignant tumor growth, >90% of a critical biochemical pathway needs to be blocked. Due to extraordinary advances in molecular biology, there is an increased understanding of rationale and relevant molecular targets in cancer. However, due to the heterogeneity of the molecular abnormalities in multiple tumor types, strategies designed to interfere with multiple molecular abnormalities will be necessary to impact survival. Nanoparticles have the potential to provide therapies not possible with other drug modalities. Researchers and clinicians must take advantage of these opportunities in order for nanotechnology to make an impact in the diagnosis and treatment of malignancy. A discussion of relevant targets either on the cell surface or the cytoplasm and strategies to achieve optimal drug targeting are the focus of this chapter.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Protocol
USD 49.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 84.99
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 169.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 109.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  1. Hanahan, D. and Weinberg, R. A. (2000) The hallmarks of cancer. Cell 100, 57–70.

    Article  CAS  PubMed  Google Scholar 

  2. Hahn, W. C. and Weinberg, R. A. (2002) Modeling the molecular circuitry of cancer. Nat Rev Cancer 2, 331–341.

    Article  CAS  PubMed  Google Scholar 

  3. Vogelstein, B., Lane, D., and Levine, A. J. (2000) Surfing the p53 network. Nature 408, 307–310.

    Article  CAS  PubMed  Google Scholar 

  4. el-Deiry, W. S. (1998) Regulation of p53 downstream genes. Semin Cancer Biol 8, 345–357.

    Article  CAS  PubMed  Google Scholar 

  5. Murphy, M., Ahn, J., Walker, K. K., Hoffman, W. H., Evans, R. M., Levine, A. J., et al. (1999) Transcriptional repression by wild-type p53 utilizes histone deacetylases, mediated by interaction with mSin3a. Genes Dev 13, 2490–2501.

    Article  CAS  PubMed  Google Scholar 

  6. Guimaraes, D. P. and Hainaut, P. (2002) TP53: a key gene in human cancer. Biochimie 84, 83–93.

    Article  CAS  PubMed  Google Scholar 

  7. Weinstein, I. B., Begemann, M., Zhou, P., Han, E. K., Sgambato, A., Doki, Y., 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 

  8. Lammers, T., Hennink, W. E., and Storm, G. (2008) Tumour-targeted nanomedicines: principles and practice. Br J Cancer 99, 392–397.

    Article  CAS  PubMed  Google Scholar 

  9. Cho, K., Wang, X., Nie, S., Chen, Z. G., and Shin, D. M. (2008) Therapeutic nanoparticles for drug delivery in cancer. Clin Cancer Res 14, 1310–1316.

    Article  CAS  PubMed  Google Scholar 

  10. Tolcher, A. W., Sugarman, S., Gelmon, K. A., Cohen, R., Saleh, M., Isaacs, C., et al. (1999) Randomized phase II study of BR96-doxorubicin conjugate in patients with metastatic breast cancer. J Clin Oncol 17, 478–484.

    CAS  PubMed  Google Scholar 

  11. Burgess, A. W. (2008) EGFR family: structure physiology signaling and therapeutic targets. Growth Factors 26, 263–274.

    Article  CAS  PubMed  Google Scholar 

  12. Bartlett, J. M., Langdon, S. P., Simpson, B. J., Stewart, M., Katsaros, D., Sismondi, P., et al. (1996) The prognostic value of epidermal growth factor receptor mRNA expression in primary ovarian cancer. Br J Cancer 73, 301–306.

    Article  CAS  PubMed  Google Scholar 

  13. Beckmann, M. W., Niederacher, D., Massenkeil, G., Tutschek, B., Beckmann, A., Schenko, G., et al. (1996) Expression analyses of epidermal growth factor receptor and HER-2/neu: no advantage of prediction of recurrence or survival in breast cancer patients. Oncology 53, 441–447.

    Article  CAS  PubMed  Google Scholar 

  14. Bucci, B., D’Agnano, I., Botti, C., Mottolese, M., Carico, E., Zupi, G., et al. (1997) EGF-R expression in ductal breast cancer: proliferation and prognostic implications. Anticancer Res 17, 769–774.

    CAS  PubMed  Google Scholar 

  15. Fischer-Colbrie, J., Witt, A., Heinzl, H., Speiser, P., Czerwenka, K., Sevelda, P., et al. (1997) EGFR and steroid receptors in ovarian carcinoma: comparison with prognostic parameters and outcome of patients. Anticancer Res 17, 613–619.

    CAS  PubMed  Google Scholar 

  16. Fontanini, G., De Laurentiis, M., Vignati, S., Chine, S., Lucchi, M., Silvestri, V., et al. (1998) Evaluation of epidermal growth factor-related growth factors and receptors and of neoangiogenesis in completely resected stage I-IIIA non-small-cell lung cancer: amphiregulin and microvessel count are independent prognostic indicators of survival. Clin Cancer Res 4, 241–249.

    CAS  PubMed  Google Scholar 

  17. Fujino, S., Enokibori, T., Tezuka, N., Asada, Y., Inoue, S., Kato, H., et al. (1996) A comparison of epidermal growth factor receptor levels and other prognostic parameters in non-small cell lung cancer. Eur J Cancer 32A, 2070–2074.

    Article  CAS  PubMed  Google Scholar 

  18. Messa, C., Russo, F., Caruso, M. G., and Di, L. A. (1998) EGF, TGF-alpha, and EGF-R in human colorectal adenocarcinoma. Acta Oncol 37, 285–289.

    Article  CAS  PubMed  Google Scholar 

  19. Rusch, V., Klimstra, D., Venkatraman, E., Pisters, P. W., Langenfeld, J., and Dmitrovsky, E. (1997) Overexpression of the epidermal growth factor receptor and its ligand transforming growth factor alpha is frequent in resectable non-small cell lung cancer but does not predict tumor progression. Clin Cancer Res 3, 515–522.

    CAS  PubMed  Google Scholar 

  20. Salomon, D. S., Brandt, R., Ciardiello, F., and Normanno, N. (1995) Epidermal growth factor-related peptides and their receptors in human malignancies. Crit Rev Oncol Hematol 19, 183–232.

    Article  CAS  PubMed  Google Scholar 

  21. Uegaki, K., Nio, Y., Inoue, Y., Minari, Y., Sato, Y., Song, M. M., et al. (1997) Clinicopathological significance of epidermal growth factor and its receptor in human pancreatic cancer. Anticancer Res 17, 3841–3847.

    CAS  PubMed  Google Scholar 

  22. Walker, R. A. and Dearing, S. J. (1999) Expression of epidermal growth factor receptor mRNA and protein in primary breast carcinomas. Breast Cancer Res Treat 53, 167–176.

    Article  CAS  PubMed  Google Scholar 

  23. Baselga, J., Tripathy, D., Mendelsohn, J., Baughman, S., Benz, C. C., Dantis, L., et al. (1996) Phase II study of weekly intravenous recombinant humanized anti-p185HER2 monoclonal antibody in patients with HER2/neu-overexpressing metastatic breast cancer. J Clin Oncol 14, 737–744.

    CAS  PubMed  Google Scholar 

  24. Cobleigh, M. A., Vogel, C. L., Tripathy, D., Robert, N. J., Scholl, S., Fehrenbacher, L., et al. (1999) Multinational study of the efficacy and safety of humanized anti-HER2 monoclonal antibody in women who have HER2-overexpressing metastatic breast cancer that has progressed after chemotherapy for metastatic disease. J Clin Oncol 17, 2639–2648.

    CAS  PubMed  Google Scholar 

  25. Nahta, R., Iglehart, J. D., Kempkes, B., and Schmidt, E. V. (2002) Rate-limiting effects of Cyclin D1 in transformation by ErbB2 predicts synergy between herceptin and flavopiridol. Cancer Res 62, 2267–2271.

    CAS  PubMed  Google Scholar 

  26. Xu, F., Lupu, R., Rodriguez, G. C., Whitaker, R. S., Boente, M. P., Berchuck, A., et al. (1993) Antibody-induced growth inhibition is mediated through immunochemically and functionally distinct epitopes on the extracellular domain of the c-erbB-2 (HER-2/neu) gene product p185. Int J Cancer 53, 401–408.

    Article  CAS  PubMed  Google Scholar 

  27. Vogel, C. L., Cobleigh, M. A., Tripathy, D., Gutheil, J. C., Harris, L. N., Fehrenbacher, L., 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.

    Article  CAS  PubMed  Google Scholar 

  28. Braconi, C., Bracci, R., and Cellerino, R. (2008) Molecular targets in gastrointestinal stromal tumors (GIST) therapy. Curr Cancer Drug Targets 8, 359–366.

    Article  CAS  PubMed  Google Scholar 

  29. Ellis, L. M. and Hicklin, D. J. (2008) VEGF-targeted therapy: mechanisms of anti-tumour activity. Nat Rev Cancer 8, 579–591.

    Article  CAS  PubMed  Google Scholar 

  30. Hurwitz, H., Fehrenbacher, L., Novotny, W., Cartwright, T., Hainsworth, J., Heim, W., et al. (2004) Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med 350, 2335–2342.

    Article  CAS  PubMed  Google Scholar 

  31. Bergmann, U., Funatomi, H., Yokoyama, M., Beger, H. G., and Korc, M. (1995) Insulin-like growth factor I overexpression in human pancreatic cancer: evidence for autocrine and paracrine roles. Cancer Res 55, 2007–2011.

    CAS  PubMed  Google Scholar 

  32. Bergmann, U., Funatomi, H., Kornmann, M., Beger, H. G., and Korc, M. (1996) Increased expression of insulin receptor substrate-1 in human pancreatic cancer. Biochem Biophys Res Commun 220, 886–890.

    Article  CAS  PubMed  Google Scholar 

  33. Ishiwata, T., Bergmann, U., Kornmann, M., Lopez, M., Beger, H. G., and Korc, M. (1997) Altered expression of insulin-like growth factor II receptor in human pancreatic cancer. Pancreas 15, 367–373.

    Article  CAS  PubMed  Google Scholar 

  34. Rodon, J., DeSantos, V., Ferry, R. J., Jr., and Kurzrock, R. (2008) Early drug development of inhibitors of the insulin-like growth factor-I receptor pathway: lessons from the first clinical trials. Mol Cancer Ther 7, 2575–2588.

    Article  CAS  PubMed  Google Scholar 

  35. Gomis, R. R., Alarcon, C., Nadal, C., Van, P. C., and Massague, J. (2006) C/EBPbeta at the core of the TGFbeta cytostatic response and its evasion in metastatic breast cancer cells. Cancer Cell 10, 203–214.

    Article  CAS  PubMed  Google Scholar 

  36. Massague, J. and Gomis, R. R. (2006) The logic of TGFbeta signaling. FEBS Lett 580, 2811–2820.

    Article  CAS  PubMed  Google Scholar 

  37. Weiner, T. M., Liu, E. T., Craven, R. J., and Cance, W. G. (1993) Expression of focal adhesion kinase gene and invasive cancer. Lancet 342, 1024–1025.

    Article  CAS  PubMed  Google Scholar 

  38. Cance, W. G., Harris, J. E., Iacocca, M. V., Roche, E., Yang, X., Chang, J., et al. (2000) Immunohistochemical analyses of focal adhesion kinase expression in benign and malignant human breast and colon tissues: correlation with preinvasive and invasive phenotypes. Clin Cancer Res 6, 2417–2423.

    CAS  PubMed  Google Scholar 

  39. Owens, L. V., Xu, L., Craven, R. J., Dent, G. A., Weiner, T. M., Kornberg, L., et al. (1995) Overexpression of the focal adhesion kinase (p125FAK) in invasive human tumors. Cancer Res 55, 2752–2755.

    CAS  PubMed  Google Scholar 

  40. Owens, L. V., Xu, L., Dent, G. A., Yang, X., Sturge, G. C., Craven, R. J., et al. (1996) Focal adhesion kinase as a marker of invasive potential in differentiated human thyroid cancer. Ann Surg Oncol 3, 100–105.

    Article  CAS  PubMed  Google Scholar 

  41. Kurenova, E., Xu, L. H., Yang, X., Baldwin, A. S., Jr., Craven, R. J., Hanks, S. K., et al. (2004) Focal adhesion kinase suppresses apoptosis by binding to the death domain of receptor-interacting protein. Mol Cell Biol 24, 4361–4371.

    Article  CAS  PubMed  Google Scholar 

  42. Golubovskaya, V. M., Finch, R., and Cance, W. G. (2005) Direct interaction of the N-terminal domain of focal adhesion kinase with the N-terminal transactivation domain of p53. J Biol Chem 280, 25008–25021.

    Article  CAS  PubMed  Google Scholar 

  43. Golubovskaya, V. M. and Cance, W. G. (2007) Focal adhesion kinase and p53 signaling in cancer cells. Int Rev Cytol 263, 103–153.

    Article  CAS  PubMed  Google Scholar 

  44. Garces, C. A., Kurenova, E. V., Golubovskaya, V. M., and Cance, W. G. (2006) Vascular endothelial growth factor receptor-3 and focal adhesion kinase bind and suppress apoptosis in breast cancer cells. Cancer Res 66, 1446–1454.

    Article  CAS  PubMed  Google Scholar 

  45. Liu, W., Bloom, D. A., Cance, W. G., Kurenova, E. V., Golubovskaya, V. M., and Hochwald, S. N. (2008) FAK and IGF-IR interact to provide survival signals in human pancreatic adenocarcinoma cells. Carcinogenesis 29, 1096–1107.

    Article  PubMed  CAS  Google Scholar 

  46. Beierle, E. A., Trujillo, A., Nagaram, A., Kurenova, E. V., Finch, R., Ma, X., et al. (2007) N-MYC regulates focal adhesion kinase expression in human neuroblastoma. J Biol Chem 282, 12503–12516.

    Article  CAS  PubMed  Google Scholar 

  47. Choi, H. S., Wang, Z., Richmond, W., He, X., Yang, K., Jiang, T., et al. (2006) Design and synthesis of 7H-pyrrolo[2,3-d] pyrimidines as focal adhesion kinase inhibitors. Part 2. Bioorg Med Chem Lett 16, 2689–2692.

    Article  CAS  PubMed  Google Scholar 

  48. Shi, Q., Hjelmeland, A. B., Keir, S. T., Song, L., Wickman, S., Jackson, D., et al. (2007) A novel low-molecular weight inhibitor of focal adhesion kinase, TAE226, inhibits glioma growth. Mol Carcinog 46, 488–496.

    Article  CAS  PubMed  Google Scholar 

  49. Slack-Davis, J. K., Martin, K. H., Tilghman, R. W., Iwanicki, M., Ung, E. J., Autry, C., et al. (2007) Cellular characterization of a novel focal adhesion kinase inhibitor. J Biol Chem 282, 14845–14852.

    Article  CAS  PubMed  Google Scholar 

  50. Roberts, W. G., Ung, E., Whalen, P., Cooper, B., Hulford, C., Autry, C., et al. (2008) Antitumor activity and pharmacology of a selective focal adhesion kinase inhibitor, PF-562,271. Cancer Res 68, 1935–1944.

    Article  CAS  PubMed  Google Scholar 

  51. Bagi, C. M., Roberts, G. W., and Andresen, C. J. (2008) Dual focal adhesion kinase/Pyk2 inhibitor has positive effects on bone tumors: implications for bone metastases. Cancer 112, 2313–2321.

    Article  CAS  PubMed  Google Scholar 

  52. Summy, J. M., Trevino, J. G., Lesslie, D. P., Baker, C. H., Shakespeare, W. C., Wang, Y., et al. (2005) AP23846, a novel and highly potent Src family kinase inhibitor, reduces vascular endothelial growth factor and interleukin-8 expression in human solid tumor cell lines and abrogates downstream angiogenic processes. Mol Cancer Ther 4, 1900–1911.

    Article  CAS  PubMed  Google Scholar 

  53. Rucci, N., Susa, M., and Teti, A. (2008) Inhibition of protein kinase c-Src as a therapeutic approach for cancer and bone metastases. Anticancer Agents Med Chem 8, 342–349.

    Article  CAS  PubMed  Google Scholar 

  54. Heath, J. R. and Davis, M. E. (2008) Nanotechnology and cancer. Annu Rev Med 59, 251–265.

    Article  CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2010 Springer Science+Business Media, LLC

About this protocol

Cite this protocol

Hochwald, S.N. (2010). Molecular-Targeted Therapy for Cancer and Nanotechnology. In: Grobmyer, S., Moudgil, B. (eds) Cancer Nanotechnology. Methods in Molecular Biology, vol 624. Humana Press. https://doi.org/10.1007/978-1-60761-609-2_2

Download citation

  • DOI: https://doi.org/10.1007/978-1-60761-609-2_2

  • Published:

  • Publisher Name: Humana Press

  • Print ISBN: 978-1-60761-608-5

  • Online ISBN: 978-1-60761-609-2

  • eBook Packages: Springer Protocols

Publish with us

Policies and ethics