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.

  • Original Article
  • Published:

Tumor microenvironment confers mTOR inhibitor resistance in invasive intestinal adenocarcinoma

Abstract

Mechanistic target of rapamycin (mTOR) complex 1 (mTORC1) is frequently activated in cancers and can be counteracted with the clinical mTORC1 inhibitors everolimus and temsirolimus. Although mTORC1 and dual mTORC1/2 inhibitors are currently under development to treat various malignancies, the emergence of drug resistance has proven to be a major complication. Using the cis-Apc/Smad4 mouse model of locally invasive intestinal adenocarcinoma, we show that administration of everolimus or the dual mTORC1/2 inhibitor AZD8055 significantly reduces the growth of intestinal tumors. In contrast, although everolimus treatment at earlier phase of tumor progression delayed invasion of the tumors, both inhibitors exhibited little effect on blocking invasion of the tumors when administered later in their progression. Biochemical and immunohistochemical analyses revealed that treatment of cis-Apc/Smad4 mice with everolimus or AZD8055 induced marked increases in epidermal growth factor receptor (EGFR) and MEK/ERK signaling in tumor epithelial and stromal cells, respectively. Notably, co-administration of AZD8055 and the EGFR inhibitor erlotinib or the MEK inhibitor trametinib was sufficient to suppress tumor invasion in cis-Apc/Smad4 mice. These data indicate that mTOR inhibitor resistance in invasive intestinal tumors involves feedback signaling from both cancer epithelial and stromal cells, highlighting the role of tumor microenvironment in drug resistance, and support that simultaneous inhibition of mTOR and EGFR or MEK may be more effective in treating colon 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

Figure 1
Figure 2
Figure 3
Figure 4
Figure 5

Similar content being viewed by others

References

  1. Guertin DA, Sabatini DM . Defining the role of mTOR in cancer. Cancer Cell 2007; 12: 9–22.

    Article  CAS  Google Scholar 

  2. Laplante M, Sabatini DM . mTOR signaling in growth control and disease. Cell 2012; 149: 274–283.

    Article  CAS  Google Scholar 

  3. Hudes G, Carducci M, Tomczak P, Dutcher J, Figlin R, Kapoor A et al. Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma. N Engl J Med 2007; 356: 2271–2281.

    Article  CAS  Google Scholar 

  4. Motzer RJ, Escudier B, Oudard S, Hutson TE, Porta C, Bracarda S et al. Efficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomised, placebo-controlled phase III trial. Lancet 2008; 372: 449–456.

    Article  CAS  Google Scholar 

  5. Mohamed A, Krajewski K, Cakar B, Ma CX . Targeted therapy for breast cancer. Am J Pathol 2013; 183: 1096–1112.

    Article  CAS  Google Scholar 

  6. Easton JB, Houghton PJ . mTOR and cancer therapy. Oncogene 2006; 25: 6436–6446.

    Article  CAS  Google Scholar 

  7. Wander SA, Hennessy BT, Slingerland JM . Next-generation mTOR inhibitors in clinical oncology: how pathway complexity informs therapeutic strategy. J Clin Invest 2011; 121: 1231–1241.

    Article  CAS  Google Scholar 

  8. Fujishita T, Aoki K, Lane HA, Aoki M, Taketo MM . Inhibition of the mTORC1 pathway suppresses intestinal polyp formation and reduces mortality in ApcΔ716 mice. Proc Natl Acad Sci USA 2008; 105: 13544–13549.

    Article  CAS  Google Scholar 

  9. Metcalfe C, Ibrahim AE, Graeb M, de la Roche M, Schwarz-Romond T, Fiedler M et al. Dvl2 promotes intestinal length and neoplasia in the ApcMin mouse model for colorectal cancer. Cancer Res 2010; 70: 6629–6638.

    Article  CAS  Google Scholar 

  10. Fujishita T, Aoki M, Taketo MM . JNK signaling promotes intestinal tumorigenesis through activation of mTOR complex 1 in ApcΔ716 mice. Gastroenterology 2011; 140: 1556–1563.

    Article  CAS  Google Scholar 

  11. Takaku K, Oshima M, Miyoshi H, Matsui M, Seldin MF, Taketo MM . Intestinal tumorigenesis in compound mutant mice of both Dpc4 (Smad4) and Apc genes. Cell 1998; 92: 645–656.

    Article  CAS  Google Scholar 

  12. Kitamura T, Kometani K, Hashida H, Matsunaga A, Miyoshi H, Hosogi H et al. SMAD4-deficient intestinal tumors recruit CCR1+ myeloid cells that promote invasion. Nat Genet 2007; 39: 467–475.

    Article  CAS  Google Scholar 

  13. Choo AY, Yoon SO, Kim SG, Roux PP, Blenis J . Rapamycin differentially inhibits S6Ks and 4E-BP1 to mediate cell-type-specific repression of mRNA translation. Proc Natl Acad Sci USA 2008; 105: 17414–17419.

    Article  CAS  Google Scholar 

  14. Hsu PP, Kang SA, Rameseder J, Zhang Y, Ottina KA, Lim D et al. The mTOR-regulated phosphoproteome reveals a mechanism of mTORC1-mediated inhibition of growth factor signaling. Science 2011; 332: 1317–1322.

    Article  CAS  Google Scholar 

  15. Yu Y, Yoon SO, Poulogiannis G, Yang Q, Ma XM, Villén J et al. Phosphoproteomic analysis identifies Grb10 as an mTORC1 substrate that negatively regulates insulin signaling. Science 2011; 332: 1322–1326.

    Article  CAS  Google Scholar 

  16. Rodrik-Outmezguine VS, Chandarlapaty S, Pagano NC, Poulikakos PI, Scaltriti M, Moskatel E et al. mTOR kinase inhibition causes feedback-dependent biphasic regulation of AKT signaling. Cancer Discov 2011; 1: 248–259.

    Article  CAS  Google Scholar 

  17. Chen SM, Guo CL, Shi JJ, Xu YC, Chen Y, Shen YY et al. HSP90 inhibitor AUY922 abrogates up-regulation of RTKs by mTOR inhibitor AZD8055 and potentiates its antiproliferative activity in human breast cancer. Int J Cancer 2014; 135: 2462–2474.

    Article  CAS  Google Scholar 

  18. He W, Rose DW, Olefsky JM, Gustafson TA . Grb10 interacts differentially with the insulin receptor, insulin-like growth factor I receptor, and epidermal growth factor receptor via the Grb10 Src homology 2 (SH2) domain and a second novel domain located between the pleckstrin homology and SH2 domains. J Biol Chem 1998; 273: 6860–6867.

    Article  CAS  Google Scholar 

  19. Fujishita T, Kajino-Sakamoto R, Kojima Y, Taketo MM, Aoki M . Antitumor activity of the MEK inhibitor trametinib on intestinal polyp formation in ApcΔ716 mice involves stromal COX-2. Cancer Sci 2015; 106: 692–699.

    Article  CAS  Google Scholar 

  20. Oshima H, Oshima M . The inflammatory network in the gastrointestinal tumor microenvironment: lessons from mouse models. J Gastroenterol 2012; 47: 97–106.

    Article  CAS  Google Scholar 

  21. Chresta CM, Davies BR, Hickson I, Harding T, Cosulich S, Critchlow SE et al. AZD8055 is a potent, selective, and orally bioavailable ATP-competitive mammalian target of rapamycin kinase inhibitor with in vitro and in vivo antitumor activity. Cancer Res 2010; 70: 288–298.

    Article  CAS  Google Scholar 

  22. Wang Q, Wei F, Li C, Lv G, Wang G, Liu T et al. Combination of mTOR and EGFR kinase inhibitors blocks mTORC1 and mTORC2 kinase activity and suppresses the progression of colorectal carcinoma. PLoS One 2013; 8: e73175.

    Article  CAS  Google Scholar 

  23. Holohan C, Van Schaeybroeck S, Longley DB, Johnston PG . Cancer drug resistance: an evolving paradigm. Nat Rev Cancer 2013; 13: 714–726.

    Article  CAS  Google Scholar 

  24. Herszényi L, Hritz I, Lakatos G, Varga MZ, Tulassay Z . The behavior of matrix metalloproteinases and their inhibitors in colorectal cancer. Int J Mol Sci 2012; 13: 13240–13263.

    Article  Google Scholar 

  25. Holt SV, Logie A, Davies BR, Alferez D, Runswick S, Fenton S et al. Enhanced apoptosis and tumor growth suppression elicited by combination of MEK (selumetinib) and mTOR kinase inhibitors (AZD8055). Cancer Res 2011; 72: 1804–1813.

    Article  Google Scholar 

  26. Jiang Q, Weiss JM, Back T, Chan T, Ortaldo JR, Guichard S et al. mTOR kinase inhibitor AZD8055 enhances the immunotherapeutic activity of an agonist CD40 antibody in cancer treatment. Cancer Res 2011; 71: 4074–4084.

    Article  CAS  Google Scholar 

  27. Oshima M, Oshima H, Kitagawa K, Kobayashi M, Itakura C, Taketo M . Loss of Apc heterozygosity and abnormal tissue building in nascent intestinal polyps in mice carrying a truncated Apc gene. Proc Natl Acad Sci USA 1995; 92: 4482–4486.

    Article  CAS  Google Scholar 

  28. Kanda Y . Investigation of the freely-available easy-to-use software ‘EZR’ (Easy R) for medical statistics. Bone Marrow Transplant 2013; 48: 452–458.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The authors thank PM McSheehy for everolimus; M Oshima, T Kitamura, M Sonoshita, S Arimura, F Kakizaki, A Deguchi, K Sakuma and K Aoki for discussions; and R Mitsuya, M Tsuda, A Kojima, K Kobori and Y Goto for technical assistance. This work was supported by JSPS KAKENHI Grant Number 24790382 (to TF), Third Term Comprehensive Control Research for Cancer from the Ministry of Health, Labour and Welfare (to MA), The Yasuda Medical Foundation, Suzuken Memorial Foundation, Nagono Medical Foundation, The Shimabara Science Promotion Foundation (to TF), Takeda Science Foundation (to TF and MA), and Princess Takamatsu Cancer Research Fund (to MA).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to M Aoki.

Ethics declarations

Competing interests

The authors declare no conflict of interest.

Additional information

Supplementary Information accompanies this paper on the Oncogene website

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Fujishita, T., Kojima, Y., Kajino-Sakamoto, R. et al. Tumor microenvironment confers mTOR inhibitor resistance in invasive intestinal adenocarcinoma. Oncogene 36, 6480–6489 (2017). https://doi.org/10.1038/onc.2017.242

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/onc.2017.242

This article is cited by

Search

Quick links