Skip to main content
Log in

MK-2206 Causes Growth Suppression and Reduces Neuroendocrine Tumor Marker Production in Medullary Thyroid Cancer Through Akt Inhibition

  • Endocrine Tumors
  • Published:
Annals of Surgical Oncology Aims and scope Submit manuscript

Abstract

Background

Development of targeted therapies for medullary thyroid cancer (MTC) has focused on inhibition of the rearranged during transfection (RET) proto-oncogene. Akt has been demonstrated to be a downstream target of RET via the key mediator phosphoinositide-3-kinase. MK-2206 is an orally administered allosteric Akt inhibitor that has exhibited minimal toxicity in phase I trials. We explored the antitumor effects of this compound in MTC.

Methods

Human MTC-TT cells were treated with MK-2206 (0–20 μM) for 8 days. Assays for cell viability were performed at multiple time points with MTT (3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide). The mechanism of action, mechanism of growth inhibition, and production of neuroendocrine tumor markers were assessed with Western blot analysis.

Results

MK-2206 suppressed MTC cell proliferation in a dose-dependent manner (p ≤ 0.001). Levels of Akt phosphorylated at serine 473 declined with increasing doses of MK-2206, indicating successful Akt inhibition. The apoptotic proteins cleaved poly (ADP-ribose) polymerase and cleaved caspase-3 increased in a dose-dependent manner with MK-2206, while the apoptosis inhibitor survivin was markedly reduced. Importantly, the antitumor effects of MK-2206 were independent of RET inhibition, as the levels of RET protein were not blocked.

Conclusions

MK-2206 significantly suppresses MTC proliferation without RET inhibition. Given its high oral bioavailability and low toxicity profile, phase II studies with this drug alone or in combination with RET inhibitors are warranted.

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

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
FIG. 3
Fig. 4
Fig. 5

Similar content being viewed by others

References

  1. Sippel RS, Kunnimalaiyaan M, Chen H. Current management of medullary thyroid cancer. Oncologist. 2008;13:539–47.

    Article  PubMed  Google Scholar 

  2. Pitt SC, Moley JF. Medullary, anaplastic, and metastatic cancers of the thyroid. Semin Oncol. 2010;37:567–79.

    Article  PubMed  Google Scholar 

  3. Chen H, Sippel RS, O’Dorisio MS, Vinik AI, Lloyd RV, Pacak K; North American Neuroendocrine Tumor Society. The North American Neuroendocrine Tumor Society consensus guideline for the diagnosis and management of neuroendocrine tumors: pheochromocytoma, paraganglioma, and medullary thyroid cancer. Pancreas. 2010;39:775–83.

    Google Scholar 

  4. Pitt SC, Chen H. The phosphatidylinositol 3-kinase/Akt signaling pathway in medullary thyroid cancer. Surgery. 2008;144:721–4.

    Article  PubMed  Google Scholar 

  5. Pitt SC, Chen H, Kunnimalaiyaan M. Inhibition of phosphatidylinositol 3-kinase/Akt signaling suppresses tumor cell proliferation and neuroendocrine marker expression in GI carcinoid tumors. Ann Surg Oncol. 2009;16:2936–42.

    Article  PubMed  Google Scholar 

  6. Pitt SC, Chen H, Kunnimalaiyaan M. Phosphatidylinositol 3-kinase-Akt signaling in pulmonary carcinoid cells. J Am Coll Surg. 2009;209:82–8.

    Article  PubMed  Google Scholar 

  7. Adler JT, Hottinger DG, Kunnimalaiyaan M, Chen H. Inhibition of the PI3K pathway suppresses hormonal secretion and limits growth in pheochromocytoma cells. World J Surg. 2009;33:2452–7.

    Article  PubMed  Google Scholar 

  8. Jaskula-Sztul R, Pisarnturakit P, Landowski M, Chen H, Kunnimalaiyaan M. Expression of the active Notch1 decreases MTC tumor growth in vivo. J Surg Res. 2011;171:23–27.

    Article  PubMed  CAS  Google Scholar 

  9. Kunnimalaiyaan M, Ndiaye M, Chen H. Neuroendocrine tumor cell growth inhibition by ZM336372 through alterations in multiple signaling pathways. Surgery. 2007;142:959–64.

    Article  PubMed  Google Scholar 

  10. Sippel RS, Carpenter JE, Kunnimalaiyaan M, Lagerholm S, Chen H. Raf-1 activation suppresses neuroendocrine marker and hormone levels in human gastrointestinal carcinoid cells. Am J Physiol Gastrointest Liver Physiol. 2003;285:G245–54.

    PubMed  CAS  Google Scholar 

  11. Sippel RS, Carpenter JE, Kunnimalaiyaan M, Chen H. The role of human achaete-scute homolog-1 in medullary thyroid cancer cells. Surgery. 2003;134:866–71.

    Article  PubMed  Google Scholar 

  12. Kunnimalaiyaan M, Ndiaye M, Chen H. Apoptosis-mediated medullary thyroid cancer growth suppression by the PI3K inhibitor LY294002. Surgery. 2006;140:1009–14.

    Article  PubMed  Google Scholar 

  13. Kunnimalaiyaan M, Vaccaro AM, Ndiaye MA, Chen H. Overexpression of the NOTCH1 intracellular domain inhibits cell proliferation and alters the neuroendocrine phenotype of medullary thyroid cancer cells. J Biol Chem. 2006;281:39819–30.

    Article  PubMed  CAS  Google Scholar 

  14. Segouffin-Cariou C, Billaud M. Transforming ability of MEN2A-RET requires activation of the phosphatidylinositol 3-kinase/AKT signaling pathway. J Biol Chem. 2000;275:3568–76.

    Article  PubMed  CAS  Google Scholar 

  15. Mandal M, Kim S, Younes MN, et al. The Akt inhibitor KP372-1 suppresses Akt activity and cell proliferation and induces apoptosis in thyroid cancer cells. Br J Cancer. 2005;92:1899–905.

    Article  PubMed  CAS  Google Scholar 

  16. Rodriguez-Viciana P, Warne PH, Dhand R, et al. Phosphatidylinositol-3-OH kinase as a direct target of Ras. Nature. 1994;370:527–32.

    Article  PubMed  CAS  Google Scholar 

  17. Wallin JJ, Edgar KA, Guan J, et al. GDC-0980 is a novel class I PI3K/mTOR kinase inhibitor with robust activity in cancer models driven by the PI3K pathway. Mol Cancer Ther. 2011;10:2426–36.

    Article  PubMed  CAS  Google Scholar 

  18. Hennessy BT, Smith DL, Ram PT, Lu Y, Mills GB. Exploiting the PI3K/AKT pathway for cancer drug discovery. Nat Rev Drug Discov. 2005;4:988–1004.

    Article  PubMed  CAS  Google Scholar 

  19. Hirai H, Sootome H, Nakatsuru Y, et al. MK-2206, an allosteric Akt inhibitor, enhances antitumor efficacy by standard chemotherapeutic agents or molecular targeted drugs in vitro and in vivo. Mol Cancer Ther. 2010;9:1956–67.

    Article  PubMed  CAS  Google Scholar 

  20. Liu R, Liu D, Trink E, Bojdani E, Ning G, Xing M. The Akt-specific inhibitor MK2206 selectively inhibits thyroid cancer cells harboring mutations that can activate the PI3K/Akt pathway. J Clin Endocrinol Metab. 2011;96:E577–85.

    Article  PubMed  CAS  Google Scholar 

  21. Knowles JA, Golden B, Yan L, Carroll WR, Helman EE, Rosenthal EL. Disruption of the AKT pathway inhibits metastasis in an orthotopic model of head and neck squamous cell carcinoma. Laryngoscope. 2011;121:2359–65.

    Article  PubMed  CAS  Google Scholar 

  22. Yap TA, Yan L, Patnaik A, et al. First-in-man clinical trial of the oral pan-AKT inhibitor MK-2206 in patients with advanced solid tumors. J Clin Oncol. 2011;29:4688–95.

    Article  PubMed  CAS  Google Scholar 

  23. Kunnimalaiyaan M, Traeger K, Chen H. Conservation of the Notch1 signaling pathway in gastrointestinal carcinoid cells. Am J Physiol Gastrointest Liver Physiol. 2005;289:G636–42.

    PubMed  CAS  Google Scholar 

  24. Chen H, Udelsman R, Zeiger M, Ball D. Human achaete-scute homolog-1 is highly expressed in a subset of neuroendocrine tumors. Oncol Rep. 1997;4:775–8.

    PubMed  CAS  Google Scholar 

  25. Chen H, Kunnimalaiyaan M, Van Gompel JJ. Medullary thyroid cancer: the functions of Raf-1 and human achaete-scute homologue-1. Thyroid. 2005;15:511–21.

    Article  PubMed  CAS  Google Scholar 

  26. Zarebczan B, Pinchot SN, Kunnimalaiyaan M, Chen H. Hesperetin, a potential therapy for carcinoid cancer. Am J Surg. 2011;201:329–32.

    Article  PubMed  CAS  Google Scholar 

  27. Lindsley CW, Barnett SF, Layton ME, Bilodeau MT. The PI3K/Akt pathway: recent progress in the development of ATP-competitive and allosteric Akt kinase inhibitors. Curr Cancer Drug Targets. 2008;8:7–18.

    Article  PubMed  CAS  Google Scholar 

  28. Chu XY, Chen LB, Wang JH, et al. Overexpression of survivin is correlated with increased invasion and metastasis of colorectal cancer. J Surg Oncol. 2012;105:520–8.

    Article  PubMed  CAS  Google Scholar 

  29. Ekeblad S, Lejonklou MH, Stålberg P, Skogseid B. Prognostic relevance of survivin in pancreatic endocrine tumors. World J Surg. 2012;36:1411–8.

    Article  PubMed  Google Scholar 

  30. Stamatakos M, Palla V, Karaiskos I, et al. Cell cyclins: triggering elements of cancer or not? World J Surg Oncol. 2010;8:111.

    Article  PubMed  Google Scholar 

  31. Fu M, Wang C, Li Z, Sakamaki T, Pestell RG. Cyclin D1: normal and abnormal functions [minireview]. Endocrinology. 2004;145:5439–47.

    Article  PubMed  CAS  Google Scholar 

  32. Mologni L, Sala E, Riva B, et al. Expression, purification, and inhibition of human RET tyrosine kinase. Protein Expr Purif. 2005;41:177–85.

    Article  PubMed  CAS  Google Scholar 

  33. Kato M, Takeda K, Kawamoto Y, et al. Repair by Src kinase of function-impaired RET with multiple endocrine neoplasia type 2A mutation with substitutions of tyrosines in the COOH-terminal kinase domain for phenylalanine. Cancer Res. 2002;62:2414–22.

    PubMed  CAS  Google Scholar 

  34. Prazeres H, Torres J, Rodrigues F, et al. How to treat a signal? Current basis for RET-genotype-oriented choice of kinase inhibitors for the treatment of medullary thyroid cancer. J Thyroid Res. 2011;2011:678357.

    Article  PubMed  Google Scholar 

  35. Kunnimalaiyaan M, Vaccaro AM, Ndiaye MA, Chen H. Inactivation of glycogen synthase kinase-3beta, a downstream target of the raf-1 pathway, is associated with growth suppression in medullary thyroid cancer cells. Mol Cancer Ther. 2007;6:1151–8.

    Article  PubMed  CAS  Google Scholar 

  36. Bhattacharyya N. A population-based analysis of survival factors in differentiated and medullary thyroid carcinoma. Otolaryngol Head Neck Surg. 2003;128:115–23.

    Article  PubMed  Google Scholar 

  37. Wells SA, Robinson BG, Gagel RF, et al. Vandetanib in patients with locally advanced or metastatic medullary thyroid cancer: a randomized, double-blind phase III trial. J Clin Oncol. 2012;30:134–41.

    Article  PubMed  CAS  Google Scholar 

  38. Schneider T, Abdulrahman R, Corssmit EP, Morreau H, Smit JW, Kapiteijn E. Long term analysis of the efficacy and tolerability of sorafenib in advanced radio-iodine refractory differentiated thyroid carcinoma: final results of a phase II trial. Eur J Endocrinol. 2012;167:643–50.

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgment

This work was supported in part by Grants from the American College of Surgeons Resident Research Fellowship (J. Burke), NIH Grant RO1 CA121115 (H. Chen), NIH T32 Research Grant in Surgical Oncology CA090217 (H. Chen), the American Cancer Society Research Scholars Grant (H. Chen), and the American Cancer Society MEN2 Professorship (H. Chen). We thank the Merck Corporation for supplying the MK-2206 used in this study and Maria Georgen for creating the Fig. 5 artwork.

Disclosure

The authors declare no conflict of interest.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Herbert Chen MD, FACS.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Burke, J.F., Schlosser, L., Harrison, A.D. et al. MK-2206 Causes Growth Suppression and Reduces Neuroendocrine Tumor Marker Production in Medullary Thyroid Cancer Through Akt Inhibition. Ann Surg Oncol 20, 3862–3868 (2013). https://doi.org/10.1245/s10434-013-3168-2

Download citation

  • Received:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1245/s10434-013-3168-2

Keywords

Navigation