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:

Double-edged swords as cancer therapeutics: simultaneously targeting p53 and NF-κB pathways

Key Points

  • p53 is the most extensively studied tumor suppressor deregulated in most human cancers.

  • It is stimulated by various activating stimuli such as DNA damage, oncogene activation, stress and hypoxia.

  • Much effort has focused on studying activation of p53 for cancer therapy and various small-molecule activators have been identified so far. Some approaches for p53 activation for therapy include MDM2 inhibition and CDK inhibition.

  • Hyperactivation of the NF-κB pathway is seen in many cancers and inflammatory disorders.

  • The NF-κB pathway is activated by more 200 stimuli including viruses, cytokines, stress, bacterial or fungal products.

  • Efforts have focused on identifying inhibitors of the NF-κB pathway with over 785 inhibitors identified so far and counting.

  • There is a lot of interest in understanding the crosstalk between these two critical signalling pathways, which have been implicated in various human ailments.

  • With combination therapy emerging as the way forward in cancer therapy, small molecules that simultaneously target both these pathways by activating p53 and inhibiting NF-κB hold great promise for cancer therapy.

  • This Review focuses on candidate small molecules that have these dual desirable properties and suggests mechanism of action and prospects for future development.

Abstract

The p53 and nuclear factor-κB (NF-κB) pathways play crucial roles in human cancer, in which inactivation of p53 and hyperactivation of NF-κB is a common occurrence. Activation of p53 and inhibition of NF-κB promotes apoptosis. Although drugs are being designed to selectively activate p53 or inhibit NF-κB, there is no concerted effort yet to deliberately make drugs that can simultaneously do both. Recent results suggest that a surprising selection of small molecules have this desirable dual activity. In this Review we describe the principles behind such dual activities, describe the current candidate molecules and suggest mechanisms and approaches to their further development.

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: The p53 pathway.
Figure 2: The NF-κB pathway.
Figure 3: Proposed models for bifunctional activity of small molecules.
Figure 4: Classes of compounds that simultaneously activate p53 and inhibit NF-κB.

Similar content being viewed by others

References

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

    Article  CAS  PubMed  Google Scholar 

  2. Vousden, K. H. & Lane, D. P. p53 in health and disease. Nature Rev. Mol. Cell Biol. 8, 275–283 (2007). This is a comprehensive overview of the various emerging roles of p53 and its relevance in not just protecting against cancer, but also its broader role in various diseases and overall survival of the organism.

    CAS  Google Scholar 

  3. Haupt, S. & Haupt, Y. Improving cancer therapy through p53 management. Cell Cycle 3, 912–916 (2004).

    CAS  PubMed  Google Scholar 

  4. Vassilev, L. T. MDM2 inhibitors for cancer therapy. Trends Mol. Med. 13, 23–31 (2007).

    CAS  PubMed  Google Scholar 

  5. Pahl, H. L. Activators and target genes of Rel/NF-κB transcription factors. Oncogene 18, 6853–6866 (1999).

    CAS  PubMed  Google Scholar 

  6. Gilmore, T. D. & Herscovitch, M. Inhibitors of NF-κB signaling: 785 and counting. Oncogene 25, 6887–6899 (2006). This is a complete review of the various inhibitors of the NF-κB pathway: the nodes they target and their implications. An excellent read for identifying and characterizing molecules that can be used to simultaneously target multiple pathways.

    CAS  PubMed  Google Scholar 

  7. Chen, L. F. & Greene, W. C. Shaping the nuclear action of NF-κB. Nature Rev. Mol. Cell Biol. 5, 392–401 (2004).

    CAS  Google Scholar 

  8. Perkins, N. D. Integrating cell-signalling pathways with NF-κB and IKK function. Nature Rev. Mol. Cell Biol. 8, 49–62 (2007). This is a comprehensive review discussing the crosstalk and integration of the NF-κB pathway with other signalling pathways and how these may contribute to cell fate.

    CAS  Google Scholar 

  9. Aggarwal, B. B., Shishodia, S., Sandur, S. K., Pandey, M. K. & Sethi, G. Inflammation and cancer: how hot is the link? Biochem. Pharmacol. 72, 1605–1621 (2006). This review discusses in detail the links between inflammation and cancer: the various critical players and their therapeutic implications.

    CAS  PubMed  Google Scholar 

  10. Naugler, W. E. & Karin, M. NF-κB and cancer-identifying targets and mechanisms. Curr. Opin. Genet. Dev. 18, 19–26 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Pikarsky, E. et al. NF-κB functions as a tumour promoter in inflammation-associated cancer. Nature 431, 461–466 (2004).

    CAS  PubMed  Google Scholar 

  12. Webster, G. A. & Perkins, N. D. Transcriptional cross talk between NF-κB and p53. Mol. Cell. Biol. 19, 3485–3495 (1999). The first study detailing the molecular mechanism of the crosstalk between the p53 and NF-κB pathways.

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Tergaonkar, V. & Perkins, N. D. p53 and NF-κB crosstalk: IKKα tips the balance. Mol. Cell 26, 158–159 (2007).

    CAS  PubMed  Google Scholar 

  14. Huang, W. C., Ju, T. K., Hung., M. C. & Chen, C. C. Phosphorylation of CBP by IKKα promotes cell growth by switching the binding preference of CBP from p53 to NF-κB. Mol. Cell 26, 75–87 (2007).

    PubMed  PubMed Central  Google Scholar 

  15. Tergaonkar, V., Pando, M., Vafa, O., Wahl, G. & Verma, I. p53 stabilization is decreased upon NFκB activation: a role for NFκB in acquisition of resistance to chemotherapy. Cancer Cell 1, 493–503 (2002).

    CAS  PubMed  Google Scholar 

  16. Roth, B. L., Sheffler, D. J. & Kroeze, W. K. Magic shotguns versus magic bullets: selectively non-selective drugs for mood disorders and schizophrenia. Nature Rev. Drug Discov. 3, 353–359 (2004).

    CAS  Google Scholar 

  17. Hopkins, A. L., Mason, J. S. & Overington, J. P. Can we rationally design promiscuous drugs? Curr. Opin. Struct. Biol. 16, 127–136 (2006). This review discusses how rational design of promiscuous drugs with polypharmacology might be the way into the future.

    CAS  PubMed  Google Scholar 

  18. Overington, J. P., Al Lazikani, B. & Hopkins, A. L. How many drug targets are there? Nature Rev. Drug Discov. 5, 993–996 (2006). This is another interesting review that discusses the concept of polypharmacology. It addresses some critical issues and concerns on complexities of finding drug targets and their mechanism of action, such as the efficacy of drugs and the rate of target innovation.

    CAS  Google Scholar 

  19. Shapiro, G. I. Cyclin-dependent kinase pathways as targets for cancer treatment. J. Clin. Oncol. 24, 1770–1783 (2006).

    CAS  PubMed  Google Scholar 

  20. Senderowicz, A. M. Small-molecule cyclin-dependent kinase modulators. Oncogene 22, 6609–6620 (2003).

    CAS  PubMed  Google Scholar 

  21. Maccallum, D. E. et al. Seliciclib (CYC202, R-Roscovitine) induces cell death in multiple myeloma cells by inhibition of RNA polymerase II-dependent transcription and down-regulation of Mcl-1. Cancer Res. 65, 5399–5407 (2005).

    CAS  PubMed  Google Scholar 

  22. Lu, W., Chen, L., Peng, Y. & Chen, J. Activation of p53 by roscovitine-mediated suppression of MDM2 expression. Oncogene 20, 3206–3216 (2001).

    CAS  PubMed  Google Scholar 

  23. Rubbi, C. P. & Milner, J. Disruption of the nucleolus mediates stabilization of p53 in response to DNA damage and other stresses. EMBO J. 22, 6068–6077 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. McClue, S. J. et al. In vitro and in vivo antitumor properties of the cyclin dependent kinase inhibitor CYC202 (R-roscovitine). Int. J. Cancer 102, 463–468 (2002).

    CAS  PubMed  Google Scholar 

  25. Raynaud, F. I. et al. In vitro and in vivo pharmacokinetic–pharmacodynamic relationships for the trisubstituted aminopurine cyclin-dependent kinase inhibitors olomoucine, bohemine and CYC202. Clin. Cancer Res. 11, 4875–4887 (2005).

    CAS  PubMed  Google Scholar 

  26. Whittaker, S. R., Walton, M. I., Garrett, M. D. & Workman, P. The Cyclin-dependent kinase inhibitor CYC202 (R-roscovitine) inhibits retinoblastoma protein phosphorylation, causes loss of Cyclin D1, and activates the mitogen-activated protein kinase pathway. Cancer Res. 64, 262–272 (2004).

    CAS  PubMed  Google Scholar 

  27. Raje, N. et al. Seliciclib (CYC202 or R-roscovitine), a small-molecule cyclin-dependent kinase inhibitor, mediates activity via down-regulation of Mcl-1 in multiple myeloma. Blood 106, 1042–1047 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Alvi, A. J. et al. A novel CDK inhibitor, CYC202 (R-roscovitine), overcomes the defect in p53-dependent apoptosis in B-CLL by down-regulation of genes involved in transcription regulation and survival. Blood 105, 4484–4491 (2005).

    CAS  PubMed  Google Scholar 

  29. Monaco, E. A. III, Beaman-Hall, C. M., Mathur, A. & Vallano, M. L. Roscovitine, olomoucine, purvalanol: inducers of apoptosis in maturing cerebellar granule neurons. Biochem. Pharmacol. 67, 1947–1964 (2004).

    CAS  PubMed  Google Scholar 

  30. Crescenzi, E., Palumbo, G. & Brady, H. J. Roscovitine modulates DNA repair and senescence: implications for combination chemotherapy. Clin. Cancer Res. 11, 8158–8171 (2005).

    CAS  PubMed  Google Scholar 

  31. Dey, A., Wong, E. T., Cheok, C. F., Tergaonkar, V. & Lane, D. P. R-Roscovitine simultaneously targets both the p53 and NF-κB pathways and causes potentiation of apoptosis: implications in cancer therapy. Cell Death. Differ. 15, 263–273 (2008). An example of a compound, R -roscovitine, a CDK inhibitor that has the desirable property of targeting both the p53 and NF-κB pathways. This paper also describes the mechanism of action of R -roscovitine in detail.

    CAS  PubMed  Google Scholar 

  32. Rossi, A. G. et al. Cyclin-dependent kinase inhibitors enhance the resolution of inflammation by promoting inflammatory cell apoptosis. Nature Med. 12, 1056–1064 (2006).

    CAS  PubMed  Google Scholar 

  33. Bukanov, N. O., Smith, L. A., Klinger, K. W., Ledbetter, S. R. & Ibraghimov-Beskrovnaya, O. Long-lasting arrest of murine polycystic kidney disease with CDK inhibitor roscovitine. Nature 444, 949–952 (2006).

    CAS  PubMed  Google Scholar 

  34. Demidenko, Z. N. & Blagosklonny, M. V. Flavopiridol induces p53 via initial inhibition of Mdm2 and p21 and, independently of p53, sensitizes apoptosis-reluctant cells to tumor necrosis factor. Cancer Res. 64, 3653–3660 (2004).

    CAS  PubMed  Google Scholar 

  35. Kim, D. M. et al. Rapid induction of apoptosis by combination of flavopiridol and tumor necrosis factor (TNF)-α or TNF-related apoptosis-inducing ligand in human cancer cell lines. Cancer Res. 63, 621–626 (2003).

    CAS  PubMed  Google Scholar 

  36. Takada, Y. & Aggarwal, B. B. Flavopiridol inhibits NF-κB activation induced by various carcinogens and inflammatory agents through inhibition of IκBα kinase and p65 phosphorylation: abrogation of cyclin D1, cyclooxygenase-2, and matrix metalloprotease-9. J. Biol. Chem. 279, 4750–4759 (2004).

    CAS  PubMed  Google Scholar 

  37. Gurova, K. V. et al. Small molecules that reactivate p53 in renal cell carcinoma reveal a NF-κB-dependent mechanism of p53 suppression in tumors. Proc. Natl Acad. Sci. USA 102, 17448–17453 (2005). Another example of a small molecule that has the bifunctional property. This is a particularly interesting example of antimalarial compounds like quinacrine and their mechanism of action. The main advantage of these compounds is that they are already known to be well tolerated in humans.

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Gurova, K. V., Hill, J. E., Razorenova, O. V., Chumakov, P. M. & Gudkov, A. V. p53 pathway in renal cell carcinoma is repressed by a dominant mechanism. Cancer Res. 64, 1951–1958 (2004).

    CAS  PubMed  Google Scholar 

  39. Gorbachev, A. V., Gasparian, A. V., Gurova, K. V., Gudkov, A. V. & Fairchild, R. L. Quinacrine inhibits the epidermal dendritic cell migration initiating T cell-mediated skin inflammation. Eur. J. Immunol. 37, 2257–2267 (2007).

    CAS  PubMed  Google Scholar 

  40. Wallace, D. J. The use of quinacrine (Atabrine) in rheumatic diseases: a reexamination. Semin. Arthritis Rheum. 18, 282–296 (1989).

    CAS  PubMed  Google Scholar 

  41. Jung, K. J. et al. Small molecule inhibitor which reactivates p53 in HTLV-1 transformed cells. J. Virol. 82, 8537–8547 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Wang, W. et al. Acridine derivatives activate p53 and induce tumor cell death through Bax. Cancer Biol. Ther. 4, 893–898 (2005).

    CAS  PubMed  Google Scholar 

  43. Tovar, C. et al. Small-molecule MDM2 antagonists reveal aberrant p53 signaling in cancer: implications for therapy. Proc. Natl Acad. Sci. USA 103, 1888–1893 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Vassilev, L. T. et al. In vivo activation of the p53 pathway by small-molecule antagonists of MDM2. Science 303, 844–848 (2004).

    CAS  PubMed  Google Scholar 

  45. Dey, A., Wong, E. T., Bist, P., Tergaonkar, V. & Lane, D. P. Nutlin-3 inhibits the NFκB pathway in a p53-dependent manner: implications in lung cancer therapy. Cell Cycle 6, 2178–2185 (2007). Nutlins have been amongst the most extensively studied compounds activating p53. This study shows the potential of nutlin-3 to inhibit the NF-κB pathway as well. This is another example of bifunctional drugs, although in this case its effects are dependent on the p53 status of cells.

    CAS  PubMed  Google Scholar 

  46. Dey, A., Verma, C. S. & Lane, D. P. Updates on p53: modulation of p53 degradation as a therapeutic approach. Br. J. Cancer 98, 4–8 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Yang, Y. et al. Inhibitors of ubiquitin-activating enzyme (E1), a new class of potential cancer therapeutics. Cancer Res. 67, 9472–9481 (2007).

    CAS  PubMed  Google Scholar 

  48. Tergaonkar, V., Bottero, V., Ikawa, M., Li, Q. & Verma, I. M. IκB kinase-independent IκBα degradation pathway: functional NF-κB activity and implications for cancer therapy. Mol. Cell. Biol. 23, 8070–8083 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Campbell, K. J., O'Shea, J. M. & Perkins, N. D. Differential regulation of NF-κB activation and function by topoisomerase II inhibitors. BMC Cancer 6, 101 (2006).

    PubMed  PubMed Central  Google Scholar 

  50. Campbell, K. J., Rocha, S. & Perkins, N. D. Active repression of antiapoptotic gene expression by RelA(p65) NF-κB. Mol. Cell 13, 853–865 (2004).

    CAS  PubMed  Google Scholar 

  51. Campbell, K. J., Witty, J. M., Rocha, S. & Perkins, N. D. Cisplatin mimics ARF tumor suppressor regulation of RelA (p65) nuclear factor-κB transactivation. Cancer Res. 66, 929–935 (2006).

    CAS  PubMed  Google Scholar 

  52. Tergaonkar, V., Correa, R. G., Ikawa, M. & Verma, I. M. Distinct roles of IκB proteins in regulating constitutive NF-κB activity. Nature Cell Biol. 7, 921–923 (2005).

    CAS  PubMed  Google Scholar 

  53. Kau, T. R. & Silver, P. A. Nuclear transport as a target for cell growth. Drug Discov. Today 8, 78–85 (2003).

    CAS  PubMed  Google Scholar 

  54. Rodriguez, M. S., Thompson, J., Hay, R. T. & Dargemont, C. Nuclear retention of IκBα protects it from signal-induced degradation and inhibits nuclear factor κB transcriptional activation. J. Biol. Chem. 274, 9108–9115 (1999).

    CAS  PubMed  Google Scholar 

  55. Poiesz, B. J. et al. Detection and isolation of type C retrovirus particles from fresh and cultured lymphocytes of a patient with cutaneous T-cell lymphoma. Proc. Natl Acad. Sci. USA 77, 7415–7419 (1980).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Yoshida, M., Miyoshi, I. & Hinuma, Y. Isolation and characterization of retrovirus from cell lines of human adult T-cell leukemia and its implication in the disease. Proc. Natl Acad. Sci. USA 79, 2031–2035 (1982).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Gessain, A. et al. Antibodies to human T-lymphotropic virus type-I in patients with tropical spastic paraparesis. Lancet 2, 407–410 (1985).

    CAS  PubMed  Google Scholar 

  58. Nakamura, F. et al. HTLV-1 associated myelopathy in an HTLV-1 and HBV double carrier family: report of a case and the mode of vertical transmission of both viruses. J. Gastroenterol. Hepatol. 4, 387–390 (1989).

    CAS  PubMed  Google Scholar 

  59. Osame, M. et al. HTLV-I associated myelopathy, a new clinical entity. Lancet 1, 1031–1032 (1986).

    CAS  PubMed  Google Scholar 

  60. Jeong, S. J., Radonovich, M., Brady, J. N. & Pise-Masison, C. A. HTLV-I Tax induces a novel interaction between p65/RelA and p53 that results in inhibition of p53 transcriptional activity. Blood 104, 1490–1497 (2004).

    CAS  PubMed  Google Scholar 

  61. Pise-Masison, C. A. et al. Inhibition of p53 transactivation function by the human T-cell lymphotropic virus type 1 Tax protein. J. Virol. 72, 1165–1170 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Pise-Masison, C. A., Radonovich, M., Sakaguchi, K., Appella, E. & Brady, J. N. Phosphorylation of p53: a novel pathway for p53 inactivation in human T-cell lymphotropic virus type 1-transformed cells. J. Virol. 72, 6348–6355 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Pise-Masison, C. A. et al. Inactivation of p53 by human T-cell lymphotropic virus type 1 Tax requires activation of the NF-κB pathway and is dependent on p53 phosphorylation. Mol. Cell. Biol. 20, 3377–3386 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Dasgupta, A., Jung, K. J., Jeong, S. J. & Brady, J. N. Inhibition of methyltransferases results in induction of G2/M checkpoint and programmed cell death in human T-lymphotropic virus type 1-transformed cells. J. Virol. 82, 49–59 (2008).

    CAS  PubMed  Google Scholar 

  65. Schwerk, C. & Schulze-Osthoff, K. Methyltransferase inhibition induces p53-dependent apoptosis and a novel form of cell death. Oncogene 24, 7002–7011 (2005).

    CAS  PubMed  Google Scholar 

  66. Jeong, S. J., Pise-Masison, C. A., Radonovich, M. F., Park, H. U. & Brady, J. N. Activated AKT regulates NF-κB activation, p53 inhibition and cell survival in HTLV-1-transformed cells. Oncogene 24, 6719–6728 (2005).

    CAS  PubMed  Google Scholar 

  67. de Moraes, E., Dar, N. A., Moura Gallo, C. V. & Hainaut, P. Cross-talks between cyclooxygenase-2 and tumor suppressor protein p53: Balancing life and death during inflammatory stress and carcinogenesis. Int. J. Cancer 121, 929–937 (2007).

    CAS  PubMed  Google Scholar 

  68. Swamy, M. V., Herzog, C. R. & Rao, C. V. Inhibition of COX-2 in colon cancer cell lines by celecoxib increases the nuclear localization of active p53. Cancer Res. 63, 5239–5242 (2003).

    CAS  PubMed  Google Scholar 

  69. Schonthal, A. H. Direct non-cyclooxygenase-2 targets of celecoxib and their potential relevance for cancer therapy. Br. J. Cancer 97, 1465–1468 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Li, H. L. et al. Changes of NF-κB, p53, Bcl-2 and caspase in apoptosis induced by JTE-522 in human gastric adenocarcinoma cell line AGS cells: role of reactive oxygen species. World J. Gastroenterol. 8, 431–435 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Aggarwal, B. B. & Shishodia, S. Molecular targets of dietary agents for prevention and therapy of cancer. Biochem. Pharmacol. 71, 1397–1421 (2006).

    CAS  PubMed  Google Scholar 

  72. Guzman, M. L. et al. An orally bioavailable parthenolide analog selectively eradicates acute myelogenous leukemia stem and progenitor cells. Blood 110, 4427–4435 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Oka, D. et al. Sesquiterpene lactone parthenolide suppresses tumor growth in a xenograft model of renal cell carcinoma by inhibiting the activation of NF-κB. Int. J. Cancer 120, 2576–2581 (2007).

    CAS  PubMed  Google Scholar 

  74. Folkman, J. Angiogenesis: an organizing principle for drug discovery? Nature Rev. Drug Discov. 6, 273–286 (2007).

    CAS  Google Scholar 

  75. Teodoro, J. G., Evans, S. K. & Green, M. R. Inhibition of tumor angiogenesis by p53: a new role for the guardian of the genome. J. Mol. Med. 85, 1175–1186 (2007).

    CAS  PubMed  Google Scholar 

  76. Naugler, W. E. & Karin, M. NF-κB and cancer-identifying targets and mechanisms. Curr. Opin. Genet. Dev. 18, 19–26 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Mantovani, A., Allavena, P., Sica, A. & Balkwill, F. Cancer-related inflammation. Nature 454, 436–444 (2008).

    CAS  PubMed  Google Scholar 

  78. Ingber, D. et al. Synthetic analogues of fumagillin that inhibit angiogenesis and suppress tumour growth. Nature 348, 555–557 (1990).

    CAS  PubMed  Google Scholar 

  79. Yeh, J. R., Mohan, R. & Crews, C. M. The antiangiogenic agent TNP-470 requires p53 and p21CIP/WAF for endothelial cell growth arrest. Proc. Natl Acad. Sci. USA 97, 12782–12787 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Mauriz, J. L. et al. TNP-470 inhibits oxidative stress, nitric oxide production and nuclear factor κB activation in a rat model of hepatocellular carcinoma. Free Radic. Res. 37, 841–848 (2003).

    CAS  PubMed  Google Scholar 

  81. Ozes, O. N. et al. NF-κB activation by tumour necrosis factor requires the Akt serine-threonine kinase. Nature 401, 82–85 (1999).

    CAS  PubMed  Google Scholar 

  82. Sizemore, N., Leung, S. & Stark, G. R. Activation of phosphatidylinositol 3-kinase in response to interleukin-1 leads to phosphorylation and activation of the NF-κB p65/RelA subunit. Mol. Cell. Biol. 19, 4798–4805 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Vivanco, I. & Sawyers, C. L. The phosphatidylinositol 3-kinase AKT pathway in human cancer. Nature Rev. Cancer 2, 489–501 (2002).

    CAS  Google Scholar 

  84. Lindsley, C. W. et al. Allosteric Akt (PKB) inhibitors: discovery and SAR of isozyme selective inhibitors. Bioorg. Med. Chem. Lett. 15, 761–764 (2005).

    CAS  PubMed  Google Scholar 

  85. Rocha, S., Campbell, K. J. & Perkins, N. D. p53- and Mdm2-independent repression of NF-κB transactivation by the ARF tumor suppressor. Mol. Cell 12, 15–25 (2003).

    CAS  PubMed  Google Scholar 

  86. Ryan, K. M., Ernst, M. K., Rice, N. R. & Vousden, K. H. Role of NF-κB in p53-mediated programmed cell death. Nature 404, 892–897 (2000).

    CAS  PubMed  Google Scholar 

  87. Schumm, K., Rocha, S., Caamano, J. & Perkins, N. D. Regulation of p53 tumour suppressor target gene expression by the p52 NF-κB subunit. EMBO J. 25, 4820–4832 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  88. Gudkov, A. V. & Komarova, E. A. Prospective therapeutic applications of p53 inhibitors. Biochem. Biophys. Res. Commun. 331, 726–736 (2005).

    CAS  PubMed  Google Scholar 

  89. Komarov, P. G. et al. A chemical inhibitor of p53 that protects mice from the side effects of cancer therapy. Science 285, 1733–1737 (1999).

    CAS  PubMed  Google Scholar 

  90. Bernardi, R. & Pandolfi, P. P. Structure, dynamics and functions of promyelocytic leukaemia nuclear bodies. Nature Rev. Mol. Cell Biol. 8, 1006–1016 (2007).

    CAS  Google Scholar 

  91. de Stanchina, E. et al. PML is a direct p53 target that modulates p53 effector functions. Mol. Cell 13, 523–535 (2004).

    CAS  PubMed  Google Scholar 

  92. Wu, W. S. et al. Promyelocytic leukemia protein sensitizes tumor necrosis factor α-induced apoptosis by inhibiting the NF-κB survival pathway. J. Biol. Chem. 278, 12294–12304 (2003).

    CAS  PubMed  Google Scholar 

  93. Weisz, L. et al. Mutant p53 enhances nuclear factor κB activation by tumor necrosis factor α in cancer cells. Cancer Res. 67, 2396–2401 (2007).

    CAS  PubMed  Google Scholar 

  94. Farooqui, A. A., Ong, W. Y. & Horrocks, L. A. Inhibitors of brain phospholipase A2 activity: their neuropharmacological effects and therapeutic importance for the treatment of neurologic disorders. Pharmacol. Rev. 58, 591–620 (2006).

    CAS  PubMed  Google Scholar 

  95. Lee, K. C. et al. Detection of the p53 response in zebrafish embryos using new monoclonal antibodies. Oncogene 27, 629–640 (2008).

    CAS  PubMed  Google Scholar 

  96. Correa, R. G. et al. Zebrafish IκB kinase 1 negatively regulates NF-κB activity. Curr. Biol. 15, 1291–1295 (2005).

    CAS  PubMed  Google Scholar 

  97. Kashatus, D., Cogswell, P. & Baldwin, A. S. Expression of the Bcl-3 proto-oncogene suppresses p53 activation. Genes Dev. 20, 225–235 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors apologize to all those whose primary work could not be cited owing to space limitations. This work is supported by the Agency for Science, Technology and Research (A*STAR), Singapore. D.P.L. is a Gibb Fellow of Cancer Research UK.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Vinay Tergaonkar or David P. Lane.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Related links

Related links

FURTHER INFORMATION

Cyclacel press release

Institute of Molecular and Cell Biology, Proteos

Rights and permissions

Reprints and permissions

About this article

Cite this article

Dey, A., Tergaonkar, V. & Lane, D. Double-edged swords as cancer therapeutics: simultaneously targeting p53 and NF-κB pathways. Nat Rev Drug Discov 7, 1031–1040 (2008). https://doi.org/10.1038/nrd2759

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrd2759

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