Skip to main content

Advertisement

Log in

New targeted therapies in pituitary carcinoma resistant to temozolomide

  • Published:
Pituitary Aims and scope Submit manuscript

Abstract

To evaluate the antitumoral efficacy of everolimus in pituitary carcinoma resistant to temozolomide, the correlation with mammalian target of rapamycin (mTOR) signaling in the tumor and to present recent advances and future treatments of pituitary carcinomas. Pituitary carcinomas are rare and largely unresponsive to current treatment options. Recent reports on the antitumoral efficacy of temozolomide in some such patients are encouraging, yet most patients appear to show resistance to its actions. As a potential alternative, the mTOR inhibitor, everolimus, has been shown to potently inhibit pituitary cell proliferation highlighting mTOR inhibition as a promising therapeutic approach for pituitary carcinomas. We described the tumoral effects of a combination therapy with everolimus (5 mg/day) and octreotide (30 mg/month) and the mTOR signalling expression in a patient with pituitary ACTH carcinoma, compared to 17 other ACTH adenomas. Clinical and biochemical evaluation were performed every month, and imaging after 3 month of treatment. mTOR signaling was assessed by microarray expression analysis of each of the 18 adenoma tissues. Combined therapy failed to control pituitary tumor growth and ACTH secretion. Slight activation of mTOR signaling was found in all ACTH tumors alongside important variations between tumors. Low antitumor efficacy shown by everolimus might be explained by the weak activation of mTOR pathway in ACTH tumors. Everolimus treatment was inefficient at controlling secretion and tumor growth of one ACTH pituitary carcinoma. More clinical cases, with mTOR signalling expression analysis of the tumor, must be published before any conclusions can be drawn.

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

Similar content being viewed by others

References

  1. Kaltsas GA, Nomikos P, Kontogeorgos G, Buchfelder M, Grossman AB (2005) Diagnosis and management of pituitary carcinomas. J Clin Endocrinol Metab 90(5):3089–3099. doi:10.1210/jc.2004-2231

    Google Scholar 

  2. Kaltsas GA, Mukherjee JJ, Plowman PN, Monson JP, Grossman AB, Besser GM (1998) The role of cytotoxic chemotherapy in the management of aggressive and malignant pituitary tumors. J Clin Endocrinol Metab 83(12):4233–4238

    Article  PubMed  CAS  Google Scholar 

  3. Lim S, Shahinian H, Maya MM, Yong W, Heaney AP (2006) Temozolomide: a novel treatment for pituitary carcinoma. Lancet Oncol 7(6):518–520

    Article  PubMed  Google Scholar 

  4. Raverot G, Sturm N, de Fraipont F, Muller M, Salenave S, Caron P, Chabre O, Chanson P, Cortet-Rudelli C, Assaker R, Dufour H, Gaillard S, Francois P, Jouanneau E, Passagia J-G, Bernier M, Cornelius A, Figarella-Branger D, Trouillas J, Borson-Chazot F, Brue T (2010) Temozolomide treatment in aggressive pituitary tumors and pituitary carcinomas: a French multicenter experience. J Clin Endocrinol Metab 95(10):4592–4599

    Article  PubMed  CAS  Google Scholar 

  5. Bush ZM, Longtine JA, Cunningham T, Schiff D, Jane JA, Vance ML, Thorner MO, Laws ER, Lopes MBS (2010) Temozolomide treatment for aggressive pituitary tumors: correlation of clinical outcome with O6-methylguanine methyltransferase (MGMT) promoter methylation and expression. J Clin Endocrinol Metab 95(11):E280–E290. doi:10.1210/jc.2010-0441

    Article  PubMed  Google Scholar 

  6. Losa M, Mazza E, Terreni MR, McCormack A, Gill AJ, Motta M, Cangi MG, Talarico A, Mortini P, Reni M (2010) Salvage therapy with temozolomide in patients with aggressive or metastatic pituitary adenomas: experience in six cases. Eur J Endocrinol 163(6):843–851. doi:10.1530/eje-10-0629

    Article  PubMed  CAS  Google Scholar 

  7. Dworakowska D, Wlodek E, Leontiou CA, Igreja S, Cakir M, Teng M, Prodromou N, Goth MI, Grozinsky-Glasberg S, Gueorguiev M, Kola B, Korbonits M, Grossman AB (2009) Activation of RAF/MEK/ERK and PI3K/AKT/mTOR pathways in pituitary adenomas and their effects on downstream effectors. Endocr Relat Cancer 16(4):1329–1338. doi:ERC-09-0101[pii]10.1677/ERC-09-0101

    Article  PubMed  CAS  Google Scholar 

  8. Yao JC, Shah MH, Ito T, Bohas CL, Wolin EM, Van Cutsem E, Hobday TJ, Okusaka T, Capdevila J, de Vries EG, Tomassetti P, Pavel ME, Hoosen S, Haas T, Lincy J, Lebwohl D, Oberg K (2011) Everolimus for advanced pancreatic neuroendocrine tumors. N Engl J Med 364(6):514–523. doi:10.1056/NEJMoa1009290

    Article  PubMed  CAS  Google Scholar 

  9. Gorshtein A, Rubinfeld H, Kendler E, Theodoropoulou M, Cerovac V, Stalla GK, Cohen ZR, Hadani M, Shimon I (2009) Mammalian target of rapamycin inhibitors rapamycin and RAD001 (everolimus) induce anti-proliferative effects in GH-secreting pituitary tumor cells in vitro. Endocr Relat Cancer 16(3):1017–1027. doi:ERC-08-0269[pii]10.1677/ERC-08-0269

    Article  PubMed  CAS  Google Scholar 

  10. Zatelli MC, Minoia M, Filieri C, Tagliati F, Buratto M, Ambrosio MR, Lapparelli M, Scanarini M, degli Uberti EC (2010) Effect of everolimus on cell viability in nonfunctioning pituitary adenomas. J Clin Endocrinol Metab 95(2):968–976. doi:10.1210/jc.2009-1641

    Article  PubMed  CAS  Google Scholar 

  11. Cerovac V, Monteserin-Garcia J, Rubinfeld H, Buchfelder M, Losa M, Florio T, Paez-Pereda M, Stalla GK, Theodoropoulou M (2010) The somatostatin analogue octreotide confers sensitivity to rapamycin treatment on pituitary tumor cells. Cancer Res 70(2):666–674. doi:0008-5472.CAN-09-2951[pii]10.1158/0008-5472.CAN-09-2951

    Article  PubMed  CAS  Google Scholar 

  12. Jouanneau E, Ducluzeau PH, Tilikete C, Borson-Chazot F, Trouillas J, Perrin G (2001) Should silent corticotroph-cell adenoma be classified as a non-functional pituitary adenoma? Neurochirurgie 47(2–3 Pt 1):128–132

    Google Scholar 

  13. Raverot G, Wierinckx A, Jouanneau E, Auger C, Borson-Chazot F, Lachuer J, Pugeat M, Trouillas J (2010) Clinical, hormonal and molecular characterization of pituitary ACTH adenomas without (silent corticotroph adenomas) and with Cushing’s disease. Eur J Endocrinol doi:10.1530/EJE-10-0076

  14. McCabe CJ, Boelaert K, Tannahill LA, Heaney AP, Stratford AL, Khaira JS, Hussain S, Sheppard MC, Franklyn JA, Gittoes NJ (2002) Vascular endothelial growth factor, its receptor KDR/Flk-1, and pituitary tumor transforming gene in pituitary tumors. J Clin Endocrinol Metab 87(9):4238–4244

    Article  PubMed  CAS  Google Scholar 

  15. Zatelli MC, Piccin D, Vignali C, Tagliati F, Ambrosio MR, Bondanelli M, Cimino V, Bianchi A, Schmid HA, Scanarini M, Pontecorvi A, De Marinis L, Maira G, degli Uberti EC (2007) Pasireotide, a multiple somatostatin receptor subtypes ligand, reduces cell viability in non-functioning pituitary adenomas by inhibiting vascular endothelial growth factor secretion. Endocr Relat Cancer 14(1):91–102. doi:14/1/91[pii]10.1677/ERC-06-0026

    Article  PubMed  CAS  Google Scholar 

  16. Luque GM, Perez-Millan MI, Ornstein AM, Cristina C, Becu-Villalobos D (2011) Inhibitory effects of antivascular endothelial growth factor strategies in experimental dopamine-resistant prolactinomas. J Pharmacol Exp Ther 337(3):766–774. doi:10.1124/jpet.110.177790

    Article  PubMed  CAS  Google Scholar 

  17. Fukuoka H, Cooper O, Mizutani J, Tong Y, Ren SG, Bannykh S, Melmed S (2011) HER2/ErbB2 receptor signaling in rat and human prolactinoma cells: strategy for targeted prolactinoma therapy. Mol Endocrinol doi:me.2010-0353 [pii]10.1210/me.2010-0353

  18. Liu NA, Jiang H, Ben-Shlomo A, Wawrowsky K, Fan XM, Lin S, Melmed S (2011) Targeting zebrafish and murine pituitary corticotroph tumors with a cyclin-dependent kinase (CDK) inhibitor. Proc Natl Acad Sci USA 108(20):8414–8419. doi:1018091108[pii]10.1073/pnas.1018091108

    Article  PubMed  CAS  Google Scholar 

  19. Raverot G, Wierinckx A, Dantony E, Auger C, Chapas G, Villeneuve L, Brue T, Figarella-Branger D, Roy P, Jouanneau E, Jan M, Lachuer J, Trouillas J (2010) Prognostic factors in prolactin pituitary tumors: clinical, histological, and molecular data from a series of 94 patients with a long postoperative follow-up. J Clin Endocrinol Metab 95(4):1708–1716. doi:jc.2009-1191[pii]10.1210/jc.2009-1191

    Article  PubMed  CAS  Google Scholar 

  20. Wierinckx A, Raverot G, Nazaret N, Jouanneau E, Auger C, Lachuer J, Trouillas J (2010) Proliferation markers of human pituitary tumors: contribution of a genome-wide transcriptome approach. Mol Cell Endocrinol doi:10.1016/j.mce.2010.02.043

Download references

Acknowledgments

The authors thank Angloscribe for English language editing. This work was supported by a grant from the Ministère de la Santé (Programme Hospitalier de Recherche National no. 27-43).

Conflict of interest

The authors have nothing to disclose.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Gérald Raverot.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Jouanneau, E., Wierinckx, A., Ducray, F. et al. New targeted therapies in pituitary carcinoma resistant to temozolomide. Pituitary 15, 37–43 (2012). https://doi.org/10.1007/s11102-011-0341-0

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11102-011-0341-0

Keywords

Navigation