touchNEUROLOGY touchNEUROLOGY
Neurological Oncology
Read Time: 2 mins

Treatment of Aggressive Pituitary Adenomas and Carcinomas – An Overview

Copy Link
Published Online: Jun 27th 2012 European Neurological Review, 2012;7(3):178-180 DOI: http://doi.org/10.17925/ENR.2012.07.03.178
Authors: Luis V Syro, Leon D Ortiz, Fabio Rotondo, Humberto Uribe, Luis C Penagos, Eva Horvath, Kalman Kovacs
Quick Links:
Abstract
Article
Article Information
Abstract:
Overview

Most pituitary tumours are non-invasive, benign adenomas that remain confined to the sella turcica. Some of them recur, have a rapid growth rate, and invade surrounding tissues. These adenomas, considered aggressive pituitary tumours, are difficult to manage and present problems due to incomplete resection. A pituitary carcinoma is diagnosed when craniospinal and/or systemic metastases are documented. Treatment options for pituitary adenomas are surgery, radiation and drugs. Recent publications report the efficacy of temozolomide in the treatment of aggressive pituitary adenomas and carcinomas. Indications for, results with, and side effects of temozolomide therapy in aggressive pituitary tumours and pituitary carcinomas are reviewed here. Alternative treatment options for resistant or recurrent pituitary tumours are also discussed.

Keywords

Pituitary adenoma, pituitary carcinoma, O6-methylguanine-DNA methyltransferase (MGMT), temozolomide, everolimus, bevacizumab

Article:

Most pituitary tumours are non-invasive, benign adenomas that remain confined to the sella turcica. Although there is, at present, no accepted definition of aggressive pituitary adenomas, one would suggest that these have a tendency to recur after initial surgery. They have a rapid growth rate and invade surrounding structures such as the sphenoid and cavernous sinus as well as the skull base bone. They are clinically difficult to manage and present major problems due to incomplete resection.1

Pituitary carcinomas are rare – 0.2 % of all pituitary tumours. They present major diagnostic and therapeutic challenges. They may initially appear as benign pituitary adenomas subsequently transforming into an aggressive neoplasm, or they may be aggressive tumours from the beginning.2–4 A pituitary carcinoma is diagnosed when craniospinal and/or systemic metastases are documented.5 Predicting pituitary tumour behaviour remains a real challenge. Studies suggest that increased mitotic activity, high Ki-67, nuclear labelling index and P53 expression might be associated with tumour progression.3,5

Multiple treatment approaches – including surgery, external beam radiotherapy, gamma knife, drugs and various chemotherapeutic agents – have been used. Until recently, the treatment of pituitary carcinomas was mainly palliative and did not seem to increase overall survival. Progression of disease after a diagnosis of pituitary carcinoma was variable; approximately 75 % of patients with systemic metastasis died of the disease within one year.4 Recent publications report efficacy of temozolomide, an alkylating agent used to treat gliomas, in the management of aggressive pituitary adenomas and carcinomas.6–36 As in gliomas, the outcome of treatment might depend on the expression of O6-methylguanine-DNA methyltransferase (MGMT), a DNA repair enzyme that counteracts the action of temozolomide.6,13,37

Temozolomide
Temozolomide is an alkylating chemotherapeutic agent related to a series of imidazotetrazines. Orally administered, it readily crosses the blood–brain barrier. It exerts its cytotoxic effect through methylation of DNA at the O6 position of guanine,38 which then mispairs with thymine during the next cycle of DNA replication. Temozolomide is accepted as an effective drug in the treatment of glioblastoma multiforme and other tumours of the central nervous system.39 Recent reports point out its efficacy in malignant neuroendocrine neoplasms,40 melanomas41,42 and colorectal carcinomas.43

To view the full article in PDF or eBook formats, please click on the icons above.

References

  1. Buchfelder M, Management of aggressive pituitary adenomas: current treatment strategies, Pituitary, 2009;12(3):256–60.
  2. Kaltsas GA, Nomikos P, Kontogeorgos G, et al., Clinical review: Diagnosis and management of pituitary carcinomas, J Clin Endocrinol Metab, 2005;90(5):3089–99.
  3. Colao A, Ochoa AS, Auriemma RS, et al., Pituitary carcinomas, Front Horm Res, 2010;38:94–108.
  4. Pernicone PJ, Scheithauer BW, Sebo TJ, et al., Pituitary carcinoma: a clinicopathologic study of 15 cases, Cancer, 1997;79(4):804–12.
  5. Heaney AP, Clinical review: Pituitary carcinoma: difficult diagnosis and treatment, J Clin Endocrinol Metab, 2011;96(12):3649–60. Erratum in: J Clin Endocrinol Metab, 2012;97(3):1064.
  6. Raverot G, Castinetti F, Jouanneau E, et al., Pituitary carcinomas and aggressive pituitary tumours: merits and pitfalls of temozolomide treatment, Clin Endocrinol (Oxf), 2012;76(6):769–75.
  7. Ortiz LD, Syro LV, Scheithauer BW, et al., Temozolomide in aggressive pituitary adenomas and carcinomas, Clinics (Sao Paulo), 2012;67(Suppl. 1):119–23.
  8. Lim S, Shahinian H, Maya MM, et al., Temozolomide: a novel treatment for pituitary carcinoma, Lancet Oncol, 2006;7(6):518–20.
  9. Fadul CE, Kominsky AL, Meyer LP, et al., Long-term response of pituitary carcinoma to temozolomide. Report of two cases, J Neurosurg, 2006;105(4):621–6.
  10. Syro LV, Uribe H, Penagos LC, et al., Antitumour effects of temozolomide in a man with a large, invasive prolactinproducing pituitary neoplasm, Clin Endocrinol (Oxf), 2006;65(4):552–3.
  11. Kovacs K, Horvath E, Syro LV, et al., Temozolomide therapy in a man with an aggressive prolactin-secreting pituitary neoplasm: Morphological findings, Hum Pathol, 2007;38(1):185–9.
  12. Neff LM, Weil M, Cole A, et al., Temozolomide in the treatment of an invasive prolactinoma resistant to dopamine agonists, Pituitary, 2007;10(1):81–6.
  13. Kovacs K, Scheithauer BW, Lombardero M, et al., MGMT immunoexpression predicts responsiveness of pituitary tumors to temozolomide therapy, Acta Neuropathol, 2008;115(2):261–2.
  14. Debono M, Bridgewater C, Ross R, Newell Price J, Treating an aggressive prolactinoma in a patient with MEN 1: beneficial response to temozolomide, Presented at: Society for Endocrinology BES 2008, Harrogate, UK, April 7–10, 2008; Endocrine Abstracts, 2008;15:188.
  15. Moyes VJ, Alusi G, Sabin HI, et al., Treatment of Nelson’s syndrome with temozolomide, Eur J Endocrinol, 2009;160(1):115–9.
  16. McCormack AI, McDonald KL, Gill AJ, et al., Low O6- methylguanine-DNA methyltransferase (MGMT) expression and response to temozolomide in aggressive pituitary tumours, Clin Endocrinol (Oxf), 2009;71(2):226–33.
  17. Mohammed S, Kovacs K, Mason W, et al., Use of temozolomide in aggressive pituitary tumors: case report, Neurosurgery, 2009;64(4):E773–4; discussion E774.
  18. Takeshita A, Inoshita N, Taguchi M, et al., High incidence of low O(6)-methylguanine DNA methyltransferase expression in invasive macroadenomas of Cushing’s disease, Eur J Endocrinol, 2009;161(4):553–9.
  19. Hagen C, Schroeder HD, Hansen S, et al., Temozolomide treatment of a pituitary carcinoma and two pituitary macroadenomas resistant to conventional therapy, Eur J Endocrinol, 2009;161(4):631–7.
  20. Byrne S, Karapetis C, Vrodos N, A novel use of temozolomide in a patient with malignant prolactinoma, J Clin Neurosci, 2009;16(12):1694–6.
  21. Thearle MS, Freda PU, Bruce JN, et al., Temozolomide (Temodar®) and capecitabine (Xeloda®) treatment of an aggressive corticotroph pituitary tumor, Pituitary, 2011;14(4):418–24.
  22. Syro LV, Scheithauer BW, Ortiz LD, et al., Effect of temozolomide in a patient with recurring oncocytic gonadotrophic pituitary adenoma, Hormones (Athens), 2009;8(4):303–6.
  23. Morin E, Berthelet F, Weisnagel J, et al., Failure of temozolomide and conventional doses of pegvisomant to attain biochemical control in a severe case of acromegaly, Pituitary, 2012;15(1):97–100.
  24. Bode H, Seiz M, Lammert A, et al., SOM230 (pasireotide) and temozolomide achieve sustained control of tumour progression and ACTH secretion in pituitary carcinoma with widespread metastases, Exp Clin Endocrinol Diabetes, 2010;118(10):760–3.
  25. Raverot G, Sturm N, de Fraipont F, et al., Temozolomide treatment in aggressive pituitary tumors and pituitary carcinomas: a French multicenter experience, J Clin Endocrinol Metab, 2010;95(10):4592–9.
  26. Bush ZM, Longtine JA, Cunningham T, et al., Temozolomide treatment for aggressive pituitary tumors: correlation of clinical outcome with O(6)-methylguanine methyltransferase (MGMT) promoter methylation and expression, J Clin Endocrinol Metab, 2010;95(11):E280–90.
  27. Syro LV, Ortiz LD, Scheithauer BW, et al., Treatment of pituitary neoplasms with temozolomide: a review, Cancer, 2011;117(3):454–62.
  28. Losa M, Mazza E, Terreni MR, et al., Salvage therapy with temozolomide in patients with aggressive or metastatic pituitary adenomas: experience in six cases, Eur J Endocrinol, 2010;163(6):843–51.
  29. Dillard TH, Gultekin SH, Delashaw JB Jr, et al., Temozolomide for corticotroph pituitary adenomas refractory to standard therapy, Pituitary, 2011;14(1):80–91.
  30. Curtò L, Torre ML, Ferraù F, et al., Temozolomide-induced shrinkage of a pituitary carcinoma causing Cushing’s disease – report of a case and literature review, ScientificWorldJournal, 2010;10:2132–8.
  31. Murakami M, Mizutani A, Asano S, et al., A mechanism of acquiring temozolomide resistance during transformation of atypical prolactinoma into prolactin-producing pituitary carcinoma: case report, Neurosurgery, 2011;68(6):E1761–7.
  32. Moshkin O, Syro LV, Scheithauer BW, et al., Aggressive silent corticotroph adenoma progressing to pituitary carcinoma: the role of temozolomide therapy, Hormones (Athens), 2011;10(2):162–7.
  33. Arnold PM, Ratnasingam D, O’Neil MF, Johnson PL, Pituitary carcinoma recurrent to the lumbar intradural extramedullary space: case report, J Spinal Cord Med, 2012;35(2):118–21.
  34. Jouanneau E, Wierinckx A, Ducray F, et al., New targeted therapies in pituitary carcinoma resistant to temozolomide, Pituitary, 2012;15(1):37–43.
  35. Philippon M, Morange I, Barrie M, et al., Long-term control of a MEN1 prolactin secreting pituitary carcinoma after temozolomide treatment, Ann Endocrinol (Paris), 2012; [Epub ahead of print].
  36. Whitelaw BC, Dworakowska D, Thomas NW, et al., Temozolomide in the management of dopamine agonistresistant prolactinomas, Clin Endocrinol (Oxf), 2012;76(6):877–86.
  37. McCormack AI, Wass JA, Grossman AB, Aggressive pituitary tumours: the role of temozolomide and the assessment of MGMT status, Eur J Clin Invest, 2011;41(10):1133–48.
  38. Stevens MF, Hickman JA, Langdon SP, et al., Antitumor activity and pharmacokinetics in mice of 8-carbamoyl-3- methyl-imidazo[5,1-d]-1,2,3,5-tetrazin-4(3H)-one (CCRG 81045; M & B 39831), a novel drug with potential as an alternative to dacarbazine, Cancer Res, 1987;47(22):5846–52.
  39. Stupp R, Mason WP, van den Bent MJ, et al., Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma, N Engl J Med, 2005;352(10):987–96.
  40. Kulke MH, Stuart K, Enzinger PC, et al., Phase II study of temozolomide and thalidomide in patients with metastatic neuroendocrine tumors, J Clin Oncol, 2006;24(3):401–6.
  41. Agarwala SS, Kirkwood JM, Temozolomide, a novel alkylating agent with activity in the central nervous system, may improve the treatment of advanced metastatic melanoma, Oncologist, 2000;5(2):144–51.
  42. Quirt I, Verma S, Petrella T, et al., Temozolomide for the treatment of metastatic melanoma: a systematic review, Oncologist, 2007;12(9):1114–23.
  43. Shacham-Shmueli E, Beny A, Geva R, et al., Response to temozolomide in patients with metastatic colorectal cancer with loss of MGMT expression: a new approach in the era of personalized medicine? J Clin Oncol, 2011;29(10):e262–5.
  44. Clarke JL, Iwamoto FM, Sul J, et al., Randomized phase II trial of chemoradiotherapy followed by either dose-dense or metronomic temozolomide for newly diagnosed glioblastoma, J Clin Oncol, 2009;27(23):3861–7.
  45. Mrugala MM, Chamberlain MC, Mechanisms of disease: temozolomide and glioblastoma – look to the future, Nat Clin Pract Oncol, 2008;5(8):476–86.
  46. Liu L, Gerson SL, Targeted modulation of MGMT: clinical implications, Clin Cancer Res, 2006;12(2):328–31.
  47. Rodriguez FJ, Thibodeau SN, Jenkins RB, et al., MGMT immunohistochemical expression and promoter methylation in human glioblastoma, Appl Immunohistochem Mol Morphol, 2008;16(1):59–65.
  48. Gerson SL, Clinical relevance of MGMT in the treatment of cancer, J Clin Oncol, 2002;20(9):2388–99.
  49. Esteller M, Hamilton SR, Burger PC, et al., Inactivation of the DNA repair gene O6-methylguanine-DNA methyltransferase by promoter hypermethylation is a common event in primary human neoplasia, Cancer Res, 1999;59(4):793–7.
  50. Sharma S, Salehi F, Scheithauer BW, et al., Role of MGMT in tumor development, progression, diagnosis, treatment and prognosis, Anticancer Res, 2009;29(10):3759–68.
  51. Esteller M, Garcia-Foncillas J, Andion E, et al., Inactivation of the DNA-repair gene MGMT and the clinical response of gliomas to alkylating agents, N Engl J Med, 2000;343(19):1350–4.
  52. Kaltsas GA, Mukherjee JJ, Plowman PN, et al., The role of cytotoxic chemotherapy in the management of aggressive and malignant pituitary tumors, J Clin Endocrinol Metab, 1998;83(12):4233–8.
  53. Melmed S, Casanueva FF, Hoffman AR, et al., Diagnosis and treatment of hyperprolactinemia: an Endocrine Society clinical practice guideline, J Clin Endocrinol Metab, 2011;96(2):273–88.
  54. Jouanneau E, Wierinckx A, Ducray F, et al., New targeted therapies in pituitary carcinoma resistant to temozolomide, Pituitary, 2012;15(1):37–43.
  55. Ortiz LD, Syro LV, Scheithauer BW, et al., Anti-VEGF therapy in pituitary carcinoma, Pituitary, 2011; [Epub ahead of print].
  56. Gerlinger M, Rowan AJ, Horswell S, et al., Intratumor heterogeneity and branched evolution revealed by multiregion sequencing, N Engl J Med, 2012;366(10):883–92.
  57. Longo DL, Tumor heterogeneity and personalized medicine, N Engl J Med, 2012;366(10):956–7.
  58. Yap TA, Gerlinger M, Futreal PA, et al., Intratumor heterogeneity: seeing the wood for the trees, Sci Transl Med, 2012;4(127):127ps10.

Further Resources

Share this Article
Related Content In Neurological Oncology
  • Copied to clipboard!
    accredited arrow-down-editablearrow-downarrow_leftarrow-right-bluearrow-right-dark-bluearrow-right-greenarrow-right-greyarrow-right-orangearrow-right-whitearrow-right-bluearrow-up-orangeavatarcalendarchevron-down consultant-pathologist-nurseconsultant-pathologistcrosscrossdownloademailexclaimationfeedbackfiltergraph-arrowinterviewslinkmdt_iconmenumore_dots nurse-consultantpadlock patient-advocate-pathologistpatient-consultantpatientperson pharmacist-nurseplay_buttonplay-colour-tmcplay-colourAsset 1podcastprinter scenerysearch share single-doctor social_facebooksocial_googleplussocial_instagramsocial_linkedin_altsocial_linkedin_altsocial_pinterestlogo-twitter-glyph-32social_youtubeshape-star (1)tick-bluetick-orangetick-red tick-whiteticktimetranscriptup-arrowwebinar Sponsored Department Location NEW TMM Corporate Services Icons-07NEW TMM Corporate Services Icons-08NEW TMM Corporate Services Icons-09NEW TMM Corporate Services Icons-10NEW TMM Corporate Services Icons-11NEW TMM Corporate Services Icons-12Salary £ TMM-Corp-Site-Icons-01TMM-Corp-Site-Icons-02TMM-Corp-Site-Icons-03TMM-Corp-Site-Icons-04TMM-Corp-Site-Icons-05TMM-Corp-Site-Icons-06TMM-Corp-Site-Icons-07TMM-Corp-Site-Icons-08TMM-Corp-Site-Icons-09TMM-Corp-Site-Icons-10TMM-Corp-Site-Icons-11TMM-Corp-Site-Icons-12TMM-Corp-Site-Icons-13TMM-Corp-Site-Icons-14TMM-Corp-Site-Icons-15TMM-Corp-Site-Icons-16TMM-Corp-Site-Icons-17TMM-Corp-Site-Icons-18TMM-Corp-Site-Icons-19TMM-Corp-Site-Icons-20TMM-Corp-Site-Icons-21TMM-Corp-Site-Icons-22TMM-Corp-Site-Icons-23TMM-Corp-Site-Icons-24TMM-Corp-Site-Icons-25TMM-Corp-Site-Icons-26TMM-Corp-Site-Icons-27TMM-Corp-Site-Icons-28TMM-Corp-Site-Icons-29TMM-Corp-Site-Icons-30TMM-Corp-Site-Icons-31TMM-Corp-Site-Icons-32TMM-Corp-Site-Icons-33TMM-Corp-Site-Icons-34TMM-Corp-Site-Icons-35TMM-Corp-Site-Icons-36TMM-Corp-Site-Icons-37TMM-Corp-Site-Icons-38TMM-Corp-Site-Icons-39TMM-Corp-Site-Icons-40TMM-Corp-Site-Icons-41TMM-Corp-Site-Icons-42TMM-Corp-Site-Icons-43TMM-Corp-Site-Icons-44TMM-Corp-Site-Icons-45TMM-Corp-Site-Icons-46TMM-Corp-Site-Icons-47TMM-Corp-Site-Icons-48TMM-Corp-Site-Icons-49TMM-Corp-Site-Icons-50TMM-Corp-Site-Icons-51TMM-Corp-Site-Icons-52TMM-Corp-Site-Icons-53TMM-Corp-Site-Icons-54TMM-Corp-Site-Icons-55TMM-Corp-Site-Icons-56TMM-Corp-Site-Icons-57TMM-Corp-Site-Icons-58TMM-Corp-Site-Icons-59TMM-Corp-Site-Icons-60TMM-Corp-Site-Icons-61TMM-Corp-Site-Icons-62TMM-Corp-Site-Icons-63TMM-Corp-Site-Icons-64TMM-Corp-Site-Icons-65TMM-Corp-Site-Icons-66TMM-Corp-Site-Icons-67TMM-Corp-Site-Icons-68TMM-Corp-Site-Icons-69TMM-Corp-Site-Icons-70TMM-Corp-Site-Icons-71TMM-Corp-Site-Icons-72