Skip to main content
  • Mini Review
  • Published:

Molecularly targeting the PI3K-Akt-mTOR pathway can sensitize cancer cells to radiotherapy and chemotherapy

Abstract

Radiotherapy and chemotherapeutic agents that damage DNA are the current major non-surgical means of treating cancer. However, many patients develop resistances to chemotherapy drugs in their later lives. The PI3K and Ras signaling pathways are deregulated in most cancers, so molecularly targeting PI3K-Akt or Ras-MAPK signaling sensitizes many cancer types to radiotherapy and chemotherapy, but the underlying molecular mechanisms have yet to be determined. During the multi-step processes of tumorigenesis, cancer cells gain the capability to disrupt the cell cycle checkpoint and increase the activity of CDK4/6 by disrupting the PI3K, Ras, p53, and Rb signaling circuits. Recent advances have demonstrated that PI3K-Akt-mTOR signaling controls FANCD2 and ribonucleotide reductase (RNR). FANCD2 plays an important role in the resistance of cells to DNA damage agents and the activation of DNA damage checkpoints, while RNR is critical for the completion of DNA replication and repair in response to DNA damage and replication stress. Regulation of FANCD2 and RNR suggests that cancer cells depend on PI3K-Akt-mTOR signaling for survival in response to DNA damage, indicating that the PI3K-AktmTOR pathway promotes resistance to chemotherapy and radiotherapy by enhancing DNA damage repair.

Abbreviations

AMPK1:

AMP-activated protein kinase

ATM kinase:

ataxiatelangiectasia mutated kinase

ATR:

ataxia telangiectasia and Rad3-related

CDK4/6:

cyclin-dependent kinase 4/6

Chk1:

checkpoint kinase 1

Chk2:

checkpoint kinase 2

FA:

Fanconi anemia

FANCD2:

Fanconi anemia group D2

FANCI:

Fanconi anemia group I

HR:

homologous recombination

ICL:

DNA interstrand crosslinker

IGFBP-3:

insulin-like growth factor binding protein 3

IRS:

insulin receptor substrate

MAPK:

mitogen-activated protein kinase

mTOR:

mammalian target of rapamycin

NER:

nucleotide excision repair

PH:

pleckstrin homology

PI3K:

phosphoinositide 3-kinase

PIP2:

phosphatidylinositol 4,5-phosphate

PIP3:

phosphatidylinositol 3,4,5-trisphosphate

PTEN:

phosphatase/tensin homolog deleted on chromosome 10

Rb:

retinoblastoma

Rheb:

Ras-homolog enriched in brain

RNR:

ribonucleotide reductase

RTK:

receptor tyrosine kinase

TLS:

translesion DNA synthesis

TSC2:

tuberous sclerosis complex-2

References

  1. Wullschleger, S., Loewith, R. and Hall, M.N. TOR signaling in growth and metabolism. Cell 124 (2006) 471–484.

    Article  CAS  PubMed  Google Scholar 

  2. Zoncu, R., Efeyan, A. and Sabatini, D.M. mTOR: From growth signal integration to cancer, diabetes and ageing. Nat. Rev. Mol. Cell Biol. 12 (2011) 21–35.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  3. Laplante, M. and Sabatini, D.M. mTOR signaling in growth control and disease. Cell 149 (2012) 274–293.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  4. Cornu, M., Albert, V. and Hall, M.N. mTOR in aging, metabolism, and cancer. Curr. Opin. Genet. Dev. 23 (2013) 53–62.

    Article  CAS  PubMed  Google Scholar 

  5. Hennessy, B.T., Smith, D.L., Ram, P.T., Lu, Y. and Mills, G.B. Exploiting the PI3K/AKT pathway for cancer drug discovery. Nat. Rev. Drug Discov. 4 (2005) 988–1004.

    Article  CAS  PubMed  Google Scholar 

  6. Sarbassov, D.D., Guertin, D.A., Ali, S.M. and Sabatini, D.M. Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex. Science 307 (2005) 1098–1101.

    Article  CAS  PubMed  Google Scholar 

  7. Hung, C.M., Garcia-Haro, L., Sparks, C.A. and Guertin, D.A. mTORdependent cell survival mechanisms. Cold Spring Harb, Perspect. Biol. 4 (2012) DOI: 10.1101/cshperspect.a008771.

  8. Shaw, R.J. and Cantley, L.C. Ras, PI(3)K and mTOR signalling controls tumour cell growth. Nature 441 (2006) 424–430.

    Article  CAS  PubMed  Google Scholar 

  9. Liu, W., Zhou, Y., Reske, S.N. and Shen, C. PTEN mutation: many birds with one stone in tumorigenesis. Anticancer Res. 28 (2008) 3613–3620.

    CAS  PubMed  Google Scholar 

  10. McCubrey, J.A., Steelman, L.S., Chappell, W.H., Abrams, S.L., Franklin, R.A., Montalto, G., Cervello, M., Libra, M., Candido, S., Malaponte, G., Mazzarino, M.C., Fagone, P., Nicoletti, F., Bäsecke, J., Mijatovic, S., Maksimovic-Ivanic, D., Milella, M., Tafuri, A., Chiarini, F., Evangelisti, C., Cocco, L. and Martelli, A.M. Ras/Raf/MEK/ERK and PI3K/PTEN/Akt/mTOR cascade inhibitors: how mutations can result in therapy resistance and how to overcome resistance. Oncotarget 3 (2012) 1068–1111.

    PubMed Central  PubMed  Google Scholar 

  11. Rodon, J., Dienstmann, R., Serra, V. and Tabernero, J. Development of PI3K inhibitors: lessons learned from early clinical trials. Nat. Rev. Clin. Oncol. 10 (2013) 143–153.

    Article  CAS  PubMed  Google Scholar 

  12. Bjornsti, M.A. and Houghton, P.J. The TOR pathway: a target for cancer therapy. Nat. Rev. Cancer 4 (2004) 335–348.

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  14. Luo, J., Solimini, N.L. and Elledge, S.J. Principles of cancer therapy: oncogene and non-oncogene addiction. Cell 136 (2009) 823–837.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  15. Dick, F.A. and Rubin, S.M. Molecular mechanisms underlying RB protein function. Nat. Rev. Mol. Cell Biol. 14 (2013) 297–306.

    Article  CAS  PubMed  Google Scholar 

  16. Chen, H.Z., Tsai, S.Y. and Leone, G. Emerging roles of E2Fs in cancer: an exit from cell cycle control. Nat. Rev. Cancer 9 (2009) 785–797.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  17. Manning, B.D. and Cantley, L.C. AKT/PKB signaling: navigating downstream. Cell 129 (2007) 1261–1274.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  18. Heitman, J., Movva, N.R. and Hall, M.N. Targets for cell cycle arrest by the immunosuppressant rapamycin in yeast. Science 253 (1991) 905–909.

    Article  CAS  PubMed  Google Scholar 

  19. Loewith, R., Jacinto, E., Wullschleger, S., Lorberg, A., Crespo, J.L., Bonenfant, D., Oppliger, W., Jenoe, P. and Hall, M.N. Two TOR complexes, only one of which is rapamycin sensitive, have distinct roles in cell growth control. Mol. Cell 10 (2002) 457–468.

    Article  CAS  PubMed  Google Scholar 

  20. Sarbassov, D.D., Ali, S.M. and Sabatini, D.M. Growing roles for the mTOR pathway. Curr. Opin. Cell Biol. 17 (2005) 596–603.

    Article  CAS  PubMed  Google Scholar 

  21. Soulard, A. and Hall, M.N. SnapShot: mTOR signaling. Cell 129 (2007) 434.

    Article  PubMed  Google Scholar 

  22. Polak, P. and Hall, M.N. mTOR and the control of whole body metabolism. Curr. Opin. Cell Biol. 21 (2009) 209–218.

    Article  CAS  PubMed  Google Scholar 

  23. Inoki, K., Ouyang, H., Zhu, T., Lindvall, C., Wang, Y., Zhang, X., Yang, Q., Bennett, C., Harada, Y., Stankunas, K., Wang, C.Y., He, X., MacDougald, O.A., You, M., Williams, B.O. and Guan, K.L. TSC2 integrates Wnt and energy signals via a coordinated phosphorylation by AMPK and GSK3 to regulate cell growth. Cell 126 (2006) 955–968.

    Article  CAS  PubMed  Google Scholar 

  24. Li, Y., Inoki, K., Vacratsis, P. and Guan, K.L. The p38 and MK2 kinase cascade phosphorylates tuberin, the tuberous sclerosis 2 gene product, and enhances its interaction with 14-3-3. J. Biol. Chem. 278 (2003) 13663–13671.

    Article  CAS  PubMed  Google Scholar 

  25. Lee, D.F., Kuo, H.P., Chen, C.T., Hsu, J.M., Chou, C.K., Wei, Y., Sun, H.L., Li, L.Y., Ping, B., Huang, W.C., He, X., Hung, J.Y., Lai, C.C., Ding, Q., Su, J.L., Yang, J.Y., Sahin, A.A., Hortobagyi, G.N., Tsai, F.J., Tsai, C.H. and Hung, M.C. IKK beta suppression of TSC1 links inflammation and tumor angiogenesis via the mTOR pathway. Cell 130 (2007) 440–455.

    Article  CAS  PubMed  Google Scholar 

  26. Astrinidis, A., Senapedis, W., Coleman, T.R. and Henske, E.P. Cell cycleregulated phosphorylation of hamartin, the product of the tuberous sclerosis complex 1 gene, by cyclin-dependent kinase 1/cyclin B. J. Biol. Chem. 278 (2003) 51372–51379.

    Article  CAS  PubMed  Google Scholar 

  27. Kim, E., Goraksha-Hicks, P., Li, L., Neufeld, T.P. and Guan, K.L. Regulation of TORC1 by Rag GTPases in nutrient response. Nat. Cell Biol. 10 (2008) 935–945.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  28. Meric-Bernstam, F. and Gonzalez-Angulo, A.M. Targeting the mTOR signaling network for cancer therapy. J. Clin. Oncol. 27 (2009) 2278–2287.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  29. Yap, T.A., Garrett, M.D., Walton, M.I., Raynaud, F., de Bono, J.S. and Workman, P. Targeting the PI3K-AKT-mTOR pathway: progress, pitfalls, and promises. Curr. Opin. Pharmacol. 8 (2008) 393–412.

    Article  CAS  PubMed  Google Scholar 

  30. Moldovan, G.L. and D’Andrea, A.D. How the fanconi anemia pathway guards the genome. Annu. Rev. Genet. 43 (2009) 223–249.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  31. Kitao, H. and Takata, M. Fanconi anemia: a disorder defective in the DNA damage response. Int. J. Hematol. 93 (2011) 417–424.

    Article  CAS  PubMed  Google Scholar 

  32. Kim, H. and D’Andrea, A.D. Regulation of DNA cross-link repair by the Fanconi anemia/BRCA pathway. Genes Dev. 26 (2012) 1393–1408.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  33. Kee, Y. and D’Andrea, A.D. Expanded roles of the Fanconi anemia pathway in preserving genomic stability. Genes Dev. 24 (2010) 1680–1694.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  34. Knipscheer, P., Raschle, M., Smogorzewska. A., Enoiu, M., Ho, T.V., Scharer, O.D., Elledge, S.J. and Walter, J.C. The Fanconi anemia pathway promotes replication-dependent DNA interstrand cross-link repair. Science 326 (2009) 1698–1701.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  35. Joo, W., Xu, G., Persky, N.S., Smogorzewska, A., Rudge, D.G., Buzovetsky, O., Elledge, S.J. and Pavletich, N.P. Structure of the FANCIFANCD2 complex: insights into the Fanconi anemia DNA repair pathway. Science 333 (2011) 312–316.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  36. Shen, C., Oswald, D., Phelps, D., Cam, H., Pelloski, C.E., Pang, Q. and Houghton, P.J. Regulation of FANCD2 by the mTOR pathway contributes to the resistance of cancer cells to DNA double strand breaks. Cancer Res. 73 (2013) 3393–3401.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  37. Kastan, M.B. and Bartek, J. Cell-cycle checkpoints and cancer. Nature 432 (2004) 316–323.

    Article  CAS  PubMed  Google Scholar 

  38. Guo, F., Li, J., Du, W., Zhang, S., O’Connor, M., Thomas, G., Kozma, S., Zingarelli, B., Pang, Q. and Zheng, Y. mTOR regulates DNA damage response through NF-κB-mediated FANCD2 pathway in hematopoietic cells. Leukemia 27 (2013) 2040–2046.

    Article  CAS  PubMed  Google Scholar 

  39. Guo, F., Li, J., Zhang, S., Du, W., Amarachintha, S., Sipple, J., Phelan, J., Grimes, H.L., Zheng, Y. and Pang, Q. mTOR kinase inhibitor sensitizes T-cell lymphoblastic leukemia for chemotherapy-induced DNA damage via suppressing FANCD2 expression. Leukemia 28 (2014) 203–206.

    Article  CAS  PubMed  Google Scholar 

  40. Huang, M., Zhou, Z. and Elledge, S.J. The DNA replication and damage checkpoint pathways induce transcription by inhibition of the Crt1 repressor. Cell 94 (1998) 595–605.

    Article  CAS  PubMed  Google Scholar 

  41. Zhao, X. and Rothstein, R. The Dun1 checkpoint kinase phosphorylates and regulates the ribonucleotide reductase inhibitor Sml1. Proc. Natl. Acad. Sci. USA 99 (2002) 3746–3751.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  42. Kolberg, M., Strand, K.R., Graff, P. and Andersson, K.K. Structure, function, and mechanism of ribonucleotide reductases. Biochim. Biophys. Acta 1699 (2004) 1–34.

    Article  CAS  PubMed  Google Scholar 

  43. Shen, C., Lancaster, C.S., Shi, B., Guo, H., Thimmaiah, P. and Bjornsti, M.A. TOR signaling is a determinant of cell survival in response to DNA damage. Mol. Cell. Biol. 27 (2007) 7007–7017.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  44. Tanaka, H., Arakawa, H., Yamaguchi, T., Shiraishi, K., Fukuda, S., Matsui, K., Takei, Y. and Nakamura, Y. A ribonucleotide reductase gene involved in a p53-dependent cell-cycle checkpoint for DNA damage. Nature 404 (2000) 42–49.

    Article  CAS  PubMed  Google Scholar 

  45. D’Angiolella, V., Donato, V., Forrester, F.M., Jeong. Y.T., Pellacani, C., Kudo, Y., Saraf, A., Florens, L., Washburn, M.P. and Pagano, M. Cyclin F-mediated degradation of ribonucleotide reductase M2 controls genome integrity and DNA repair. Cell 149 (2012) 1023–1034.

    Article  PubMed Central  PubMed  Google Scholar 

  46. Imataka, H., Gradi, A. and Sonenberg, N. A newly identified N-terminal amino acid sequence of human eIF4G binds poly(A)-binding protein and functions in poly(A)-dependent translation. EMBO J. 17 (1998) 7480–7489.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  47. Chow, L.M. and Baker, S.J. PTEN function in normal and neoplastic growth. Cancer Lett. 241 (2006) 184–196.

    Article  CAS  PubMed  Google Scholar 

  48. Graat, H.C., Carette, J.E., Schagen, F.H., Vassilev, L.T., Gerritsen, W.R., Kaspers, G.J., Wuisman, P.I. and van Beusechem, V.W. Enhanced tumor cell kill by combined treatment with a small-molecule antagonist of mouse double minute 2 and adenoviruses encoding p53. Mol. Cancer Ther. 6 (2007) 1552–1561.

    Article  CAS  PubMed  Google Scholar 

  49. Wang, W. and El-Deiry, W.S. Restoration of p53 to limit tumor growth. Curr. Opin. Oncol. 20 (2008) 90–96.

    Article  PubMed  Google Scholar 

  50. Shepard, H.M., Jin, P., Slamon, D.J., Pirot, Z. and Maneval, D.C. Herceptin. Handb. Exp. Pharmacol. 181 (2008) 183–219.

    Article  CAS  PubMed  Google Scholar 

  51. Rivera, F., Vega-Villegas, M.E., Lopez-Brea, M.F. and Marquez, R. Current situation of Panitumumab, Matuzumab, Nimotuzumab and Zalutumumab. Acta Oncol. 47 (2008) 9–19.

    Article  CAS  PubMed  Google Scholar 

  52. Chresta, C.M., Davies, B.R., Hickson, I., Harding, T., Cosulich, S., Critchlow, S.E., Vincent, J.P., Ellston, R., Jones, D., Sini, P., James, D., Howard, Z., Dudley, P., Hughes, G., Smith, L., Maguire, S., Hummersone, M., Malagu, K., Menear, K., Jenkins, R., Jacobsen, M., Smith, G.C., Guichard, S. and Pass, M. AZD8055 is a potent, selective, and orally bioavailable ATPcompetitive mammalian target of rapamycin kinase inhibitor with in vitro and in vivo antitumor activity. Cancer Res. 70 (2010) 288–298.

    Article  CAS  PubMed  Google Scholar 

  53. Sangai, T., Akcakanat, A., Chen, H., Tarco, E., Wu, Y., Do, K.A., Miller, T.W., Arteaga, C.L., Mills, G.B., Gonzalez-Angulo, A.M. and Meric-Bernstam, F. Biomarkers of response to Akt inhibitor MK-2206 in breast cancer. Clin. Cancer Res. 18 (2012) 5816–5828.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  54. Vousden, K.H. and Lane, D.P. p53 in health and disease. Nat. Rev. Mol. Cell Biol. 8 (2007) 275–283.

    Article  CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Ziwen Wang or Jiqiang Zhang.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Wang, Z., Huang, Y. & Zhang, J. Molecularly targeting the PI3K-Akt-mTOR pathway can sensitize cancer cells to radiotherapy and chemotherapy. Cell Mol Biol Lett 19, 233–242 (2014). https://doi.org/10.2478/s11658-014-0191-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.2478/s11658-014-0191-7

Keywords