Skip to main content
Log in

Temozolomide therapy for aggressive pituitary tumours – current understanding and future perspectives

  • Published:
Reviews in Endocrine and Metabolic Disorders Aims and scope Submit manuscript

Abstract

The use of temozolomide (TMZ) for the management of aggressive pituitary tumours (APT) has revolutionised clinical practice in this field with significantly improved clinical outcomes and long-term survival. Its use is now well established however a large number of patients do not respond to treatment and recurrence after cessation of TMZ is common. A number of challenges remain for clinicians such as appropriate patient selection, treatment duration and the role of combination therapy. This review will examine the use of TMZ to treat APT including mechanism of action, treatment regimen and duration; biomarkers predicting response to treatment and patient selection; and current evidence for administration of TMZ in combination with other agents.

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. Fadul CE, Kominsky AL, Meyer LP, Kingman LS, Kinlaw WB, Rhodes CH, et al. Long-term response of pituitary carcinoma to temozolomide. Report of two cases J Neurosurg. 2006;105:621–6.

    PubMed  Google Scholar 

  2. Lim S, Shahinian H, Maya MM, Yong W, Heaney AP. Temozolomide: a novel treatment for pituitary carcinoma. The Lancet Oncology. 2006;7:518–20.

    Article  PubMed  Google Scholar 

  3. Syro LV, Uribe H, Penagos LC, Ortiz LD, Fadul CE, Horvath E, et al. Antitumour effects of temozolomide in a man with a large, invasive prolactin-producing pituitary neoplasm. Clin Endocrinol. 2006;65:552–3.

    Article  Google Scholar 

  4. Kaltsas GA, Mukherjee JJ, Plowman PN, Monson JP, Grossman AB, Besser GM. The role of cytotoxic chemotherapy in the management of aggressive and malignant pituitary tumours. J Clin Endocrinol Metab. 1998;83:4233–8.

    Article  CAS  PubMed  Google Scholar 

  5. Ji Y, Vogel RI, Lou E. Temozolomide treatment of pituitary carcinomas and atypical adenomas: systematic review of case reports. Neurooncol Pract. 2016;3:188–95.

    PubMed  Google Scholar 

  6. Bengtsson D, Schrøder HD, Andersen M, Maiter D, Berinder K, Feldt Rasmussen U, et al. Long-term outcome and MGMT as a predictive marker in 24 patients with atypical pituitary adenomas and pituitary carcinomas given treatment with temozolomide. J Clin Endocrinol Metab. 2015;100:1689–98.

    Article  CAS  PubMed  Google Scholar 

  7. Elbelt U, Schlaffer SM, Buchfelder M, Knappe UJ, Vila G, Micko A, et al. Efficacy of temozolomide therapy in patients with aggressive pituitary adenomas and carcinomas – a German survey. J Clin Endocrinol Metab.

  8. Lasolle H, Cortet C, Castinetti F, Cloix L, Caron P, Delemer B, et al. Temozolomide treatment can improve overall survival in aggressive pituitary tumours and pituitary carcinomas. Eur J Endocrinol. 2017;176:769–77.

    Article  CAS  PubMed  Google Scholar 

  9. Losa M, Bogazzi F, Cannavo S, Ceccato F, Curtò L, De Marinis L, et al. Temozolomide therapy in patients with aggressive pituitary adenomas or carcinomas. J Neuro-Oncol. 2016;126:519–25.

    Article  CAS  Google Scholar 

  10. McCormack A, Dekkers OM, Petersenn S, Popovic V, Trouillas J, Raverot G, et al. Treatment of aggressive pituitary tumours and carcinomas: results of a European Society of Endocrinology (ESE) survey 2016. Eur J Endocrinol. 2018;178:265–76.

    Article  CAS  PubMed  Google Scholar 

  11. McCormack AI, Wass JAH, Grossman AB. Aggressive pituitary tumours: the role of temozolomide and the assessment of MGMT status. Eur J Clin Investig. 2011;41:1133–48.

    Article  CAS  Google Scholar 

  12. Raverot G, Burman P, McCormack A, Heaney A, Petersenn S, Popovic V, et al. European Society of Endocrinology European Society of endocrinology clinical practice guidelines for the management of aggressive pituitary tumours and carcinomas. Eur J Endocrinol. 2018;178:G1–G24.

    Article  CAS  PubMed  Google Scholar 

  13. Trouillas J, Burman P, McCormack A, Petersenn S, Popovic V, Dekkers O, et al. Aggressive pituitary tumours and carcinomas: two sides of the same coin? Eur J Endocrinol. 2018;178:C7–9.

    Article  CAS  PubMed  Google Scholar 

  14. Lloyd R, Osamura R, Kloppel G, Rosai J. WHO classification of tumours of endocrine organs. 4th ed. Lyon, 2017; Vol. 10; ISBN 978-92-832-4493-6.

  15. Kulke MH, Hornick JL, Frauenhoffer C, Hooshmand S, Ryan DP, Enzinger PC, et al. O6-methylguanine DNA methyltransferase deficiency and response to temozolomide-based therapy in patients with neuroendocrine tumours. Clin Cancer Res. 2009;15:338–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Marchesi F, Turriziani M, Tortorelli G, Avvisati G, Torino F, Vecchis L. Triazene compounds: mechanism of action and related DNA repair systems. Pharmacological research : the official journal of the Italian Pharmacological Society. 2007;56:275–87.

    Article  CAS  Google Scholar 

  17. Kaina B, Christmann M, Naumann S, Roos WP. MGMT: key node in the battle against genotoxicity, carcinogenicity and apoptosis induced by alkylating agents. DNA Repair (Amst). 2007;6:1079–99.

    Article  CAS  Google Scholar 

  18. De Sousa SM, McCormack AI. Aggressive pituitary tumours and pituitary carcinomas. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, Dungan K, Grossman A, Hershman JM, Kaltsas G, Koch C, Kopp P, Korbonits M, McLachlan R, Morley JE, New M, Perreault L, Purnell J, Rebar R, Singer F, Trence DL, Vinik A, Wilson DP, editors. Endotext. MDText.com, Inc.: South Dartmouth (MA), 2000.

  19. Zhang J, Stevens M, Bradshaw T. Temozolomide: mechanisms of action, repair and resistance. Curr Mol Pharmacol. 2011;5:102–14.

    Article  Google Scholar 

  20. Liu L, Gerson SL. Targeted modulation of MGMT: clinical implications. Clin Cancer Res. 2006;12:328–31.

    Article  CAS  PubMed  Google Scholar 

  21. Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJB, et al. Radiotherapy plus concomitant and adjuvant Temozolomide for Glioblastoma. N Engl J Med. 2005;352:987–96.

    Article  CAS  PubMed  Google Scholar 

  22. Bush ZM, Longtine JA, Cunningham T, Schiff D, Jane JA, Vance ML, et al. Temozolomide treatment for aggressive pituitary tumours: correlation of clinical outcome with O(6)-methylguanine methyltransferase (MGMT) promoter methylation and expression. J Clin Endocrinol Metab. 2010;95:E280–90.

  23. Gilbert MR, Wang M, Aldape KD, Stupp R, Hegi ME, Jaeckle KA, et al. Dose-dense temozolomide for newly diagnosed glioblastoma: a randomized phase III clinical trial. J Clin Oncol. 2013;31:4085–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Neyns B, Tosoni A, Hwu W-J, Reardon DA. Dose-dense temozolomide regimens: antitumour activity, toxicity, and immunomodulatory effects. Cancer. 2010;116:2868–77.

    Article  CAS  PubMed  Google Scholar 

  25. Mendola M, Passeri E, Ambrosi B, Corbetta S. Multiple cerebral hemorrhagic foci from metastases during Temozolomide treatment in a patient with Corticotroph pituitary carcinoma. J Clin Endocrinol Metab. 2014;99:2623–4.

    Article  CAS  PubMed  Google Scholar 

  26. Ekeblad S, Sundin A, Janson E, Welin S, Granberg D, Kindmark H, et al. Temozolomide as Monotherapy is effective in treatment of advanced malignant neuroendocrine Tumours. Clinical cancer research: an official journal of the American Association for Cancer Research. 2007;13:2986–91.

    Article  CAS  Google Scholar 

  27. Campderá M, Palacios N, Aller J, Magallón R, Martín P, Saucedo G, et al. Temozolomide for aggressive ACTH pituitary tumours: failure of a second course of treatment. Pituitary. 2015;19.

  28. Neyns B, Cordera S, Joosens E, Nader P. Non-Hodgkin’s lymphoma in patients with Glioma treated with Temozolomide. JCO. 2008;26:4518–9.

    Article  Google Scholar 

  29. George B, Eichinger J, Richard T. A rare case of aplastic Anemia caused by Temozolomide. South Med J. 2009;102:974–6.

    Article  PubMed  Google Scholar 

  30. Raverot G, Sturm N, de Fraipont F, Muller M, Salenave S, Caron P, et al. Temozolomide treatment in aggressive pituitary Tumours and pituitary carcinomas: a French multicenter experience. J Clin Endocrinol Metab. 2010;95:4592–9.

    Article  CAS  PubMed  Google Scholar 

  31. Philippon M, Morange I, Barrie M, Barlier A, Taieb D, Dufour H, et al. Long-term control of a MEN1 prolactin secreting pituitary carcinoma after temozolomide treatment. Ann Endocrinol (Paris). 2012;73:225–9.

    Article  CAS  Google Scholar 

  32. Asimakopoulou A, Tzanela M, Koletti A, Kontogeorgos G, Tsagarakis S. Long-term remission in an aggressive Crooke cell adenoma of the pituitary, 18 months after discontinuation of treatment with temozolomide. Clin Case Rep. 2014;2:1–3.

    Article  PubMed  Google Scholar 

  33. Barkhoudarian G, Palejwala SK, Ogunbameru R, Wei H, Eisenberg A, Kelly DF. Early recognition and initiation of Temozolomide chemotherapy for refractory, invasive pituitary Macroprolactinoma with long-term sustained remission. World Neurosurgery. 2018;118:118–24.

    Article  PubMed  Google Scholar 

  34. Bengtsson D, Schrøder HD, Berinder K, Maiter D, Hoybye C, Ragnarsson O, et al. Tumoural MGMT content predicts survival in patients with aggressive pituitary tumours and pituitary carcinomas given treatment with temozolomide. Endocrine. 2018;62:737–9.

    Article  CAS  PubMed  Google Scholar 

  35. Rotman LE, Vaughan TB, Hackney JR, Riley KO. Long-term survival after transformation of an adrenocorticotropic hormone-secreting pituitary macroadenoma to a silent Corticotroph pituitary carcinoma. World Neurosurgery. 2019;122:417–23.

    Article  PubMed  Google Scholar 

  36. Tanaka S, Yamamoto M, Morita M, Takeno A, Kanazawa I, Yamaguchi T, et al. Successful reduction of ACTH secretion in a case of intractable Cushing’s disease with pituitary Crooke’s cell adenoma by combined modality therapy including temozolomide. Endocr J. 2019;66:701–8.

    Article  CAS  PubMed  Google Scholar 

  37. Kim J-Y, Jackman JG, Woodring S, McSherry F, Herndon JE, Desjardins A, et al. Second primary cancers in long-term survivors of glioblastoma. Neurooncol Pract. 2019;6:386–91.

    PubMed  PubMed Central  Google Scholar 

  38. Momota H, Narita Y, Miyakita Y, Shibui S. Secondary hematological malignancies associated with temozolomide in patients with glioma. Neuro-Oncology. 2013;15:1445–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Alvino E, Castiglia D, Caporali S, Pepponi R, Caporaso P, Lacal PM, et al. A single cycle of treatment with temozolomide, alone or combined with O6-benzylguanine, induces strong chemoresistance in melanoma cell clones in vitro: role of O6-methylguanine-DNA methyltransferase and the mismatch repair system. Int J Oncol. 2006;29:785–97.

    CAS  PubMed  Google Scholar 

  40. Syro LV, Rotondo F, Camargo M, Ortiz LD, Serna CA, Kovacs K. Temozolomide and pituitary tumours: current understanding, unresolved issues, and future directions. Front Endocrinol (Lausanne) 2018, 9.

  41. Hegi ME, Diserens A-C, Gorlia T, Hamou M-F, de Tribolet N, Weller M, et al. MGMT gene silencing and benefit from Temozolomide in Glioblastoma. N Engl J Med. 2005;352:997–1003.

    Article  CAS  PubMed  Google Scholar 

  42. Cros J, Hentic O, Rebours V, Zappa M, Gille N, Theou-Anton N, et al. MGMT expression predicts response to temozolomide in pancreatic neuroendocrine tumours. Endocr Relat Cancer. 2016;23:625–33.

    Article  CAS  PubMed  Google Scholar 

  43. Kovacs K, Scheithauer BW, Lombardero M, McLendon RE, Syro LV, Uribe H, et al. MGMT immunoexpression predicts responsiveness of pituitary tumours to temozolomide therapy. Acta Neuropathol. 2008;115:261–2.

    Article  PubMed  Google Scholar 

  44. Hagen C, Schroeder HD, Hansen S, Hagen C, Andersen M. Temozolomide treatment of a pituitary carcinoma and two pituitary macroadenomas resistant to conventional therapy. Eur J Endocrinol. 2009;161:631–7.

    Article  CAS  PubMed  Google Scholar 

  45. McCormack AI, McDonald KL, Gill AJ, Clark SJ, Burt MG, Campbell KA, et al. Low O6-methylguanine-DNA methyltransferase (MGMT) expression and response to temozolomide in aggressive pituitary tumours. Clin Endocrinol. 2009;71:226–33.

    Article  CAS  Google Scholar 

  46. Whitelaw BC, Dworakowska D, Thomas NW, Barazi S, Riordan-Eva P, King AP, et al. Temozolomide in the management of dopamine agonist-resistant prolactinomas. Clin Endocrinol. 2012;76:877–86.

    Article  CAS  Google Scholar 

  47. Annamalai AK, Dean AF, Kandasamy N, Kovacs K, Burton H, Halsall DJ, et al. Temozolomide responsiveness in aggressive corticotroph tumours: a case report and review of the literature. Pituitary. 2012;15:276–87.

    Article  CAS  PubMed  Google Scholar 

  48. Bengtsson D, Schrøder HD, Berinder K, Maiter D, Hoybye C, Ragnarsson O, et al. Tumoural MGMT content predicts survival in patients with aggressive pituitary tumours and pituitary carcinomas given treatment with temozolomide. Endocrine. 2018;62:737–9.

    Article  CAS  PubMed  Google Scholar 

  49. Lau Q, Scheithauer B, Kovacs K, Horvath E, Syro LV, Lloyd R. MGMT immunoexpression in aggressive pituitary adenoma and carcinoma. Pituitary. 2010;13:367–79.

    Article  PubMed  Google Scholar 

  50. Salehi F, Scheithauer BW, Kros JM, Lau Q, Fealey M, Erickson D, et al. MGMT promoter methylation and immunoexpression in aggressive pituitary adenomas and carcinomas. J Neuro-Oncol. 2011;104:647–57.

    Article  CAS  Google Scholar 

  51. Hirohata T, Asano K, Ogawa Y, Takano S, Amano K, Isozaki O, et al. DNA mismatch repair protein (MSH6) correlated with the responses of atypical pituitary adenomas and pituitary carcinomas to temozolomide: the national cooperative study by the Japan Society for Hypothalamic and Pituitary Tumours. J Clin Endocrinol Metab. 2013;98:1130–6.

  52. Micko ASG, Höftberger R, Wöhrer A, Millesi M, Knosp E, Wolfsberger S. MGMT assessment in pituitary adenomas: comparison of different immunohistochemistry fixation chemicals. Pituitary. 2018;21:266–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Micko ASG, Wöhrer A, Höftberger R, Vila G, Marosi C, Knosp E, et al. MGMT and MSH6 immunoexpression for functioning pituitary macroadenomas. Pituitary. 2017;20:643–53.

  54. Spiro T, Liu L, Gerson S. New cytotoxic agents for the treatment of metastatic malignant melanoma: temozolomide and related alkylating agents in combination with guanine analogues to abrogate drug resistance. Forum (Genova). 2000;10:274–85.

    CAS  Google Scholar 

  55. Rabik CA, Njoku MC, Dolan ME. Inactivation of O6-alkylguanine DNA alkyltransferase as a means to enhance chemotherapy. Cancer Treat Rev. 2006;32:261–76.

    Article  CAS  PubMed  Google Scholar 

  56. Stojic L, Brun R, Jiricny J. Mismatch repair and DNA damage signalling. DNA Repair. 2004;3:1091–101.

    Article  CAS  PubMed  Google Scholar 

  57. Fu D, Calvo JA, Samson LD. Balancing repair and tolerance of DNA damage caused by alkylating agents. Nat Rev Cancer. 2012;12:104–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Richman S. Deficient mismatch repair: read all about it (review). Int J Oncol. 2015;47:1189–202.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Aquilina G, Bignami M. Mismatch repair in correction of replication errors and processing of DNA damage. J Cell Physiol. 2001;187:145–54.

    Article  CAS  PubMed  Google Scholar 

  60. Bengtsson D, Joost P, Aravidis C, Askmalm Stenmark M, Backman A-S, Melin B, et al. Corticotroph pituitary carcinoma in a patient with lynch syndrome (LS) and pituitary Tumours in a Nationwide LS cohort. J Clin Endocrinol Metab. 2017;102:3928–32.

  61. Nguyen SA, Stechishin ODM, Luchman HA, Lun XQ, Senger DL, Robbins SM, et al. Novel MSH6 mutations in treatment-naïve glioblastoma and anaplastic oligodendroglioma contribute to temozolomide resistance independently of MGMT promoter methylation. Clin Cancer Res. 2014;20:4894–903.

  62. Cahill DP, Levine KK, Betensky RA, Codd PJ, Romany CA, Reavie LB, et al. Loss of the mismatch repair protein MSH6 in human glioblastomas is associated with tumour progression during temozolomide treatment. Clin Cancer Res. 2007;2038–2045:13.

  63. Murakami, M.; Mizutani, A.; Asano, S.; Katakami, H.; Ozawa, Y.; Yamazaki, K.; Ishida, Y.; Takano, K.; Okinaga, H.; Matsuno, A. A Mechanism of Acquiring Temozolomide Resistance During Transformation of Atypical Prolactinoma Into Prolactin-Producing Pituitary Carcinoma: Case Report. Neurosurgery2011, 68, E1761–7; discussion E1767.

  64. Zacharia B, Gulati A, Bruce J, Carminucci A, Wardlaw S, Siegelin M, et al. High response rates and prolonged survival in patients with Corticotroph pituitary Tumours and refractory Cushing disease from Capecitabine and Temozolomide (CAPTEM): a case series. Neurosurgery. 2014;74:E447–55.

  65. Erasimus H, Gobin M, Niclou S, Van Dyck E. DNA repair mechanisms and their clinical impact in glioblastoma. Mutation Research/Reviews in Mutation Research. 2016;769:19–35.

    Article  CAS  PubMed  Google Scholar 

  66. Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD, et al. PD-1 blockade in Tumours with mismatch-repair deficiency. N Engl J Med. 2015;372:2509–20.

  67. Lin AL, Jonsson P, Tabar V, Yang TJ, Cuaron J, Beal K, et al. Marked response of a Hypermutated ACTH-secreting pituitary carcinoma to Ipilimumab and Nivolumab. J Clin Endocrinol Metab. 2018;103:3925–30.

  68. Gubin MM, Zhang X, Schuster H, Caron E, Ward JP, Noguchi T, et al. Checkpoint blockade cancer immunotherapy targets tumour-specific mutant antigens. Nature. 2014;515:577–81.

  69. Le DT, Durham JN, Smith KN, Wang H, Bartlett BR, Aulakh LK, et al. Mismatch repair deficiency predicts response of solid tumours to PD-1 blockade. Science. 2017;357:409–13.

  70. Hombach-Klonisch S, Mehrpour M, Shojaei S, Harlos C, Pitz M, Hamai A, et al. Glioblastoma and chemoresistance to alkylating agents: involvement of apoptosis, autophagy, and unfolded protein response. Pharmacol Ther. 2018;184:13–41.

  71. Agnihotri S, Gajadhar AS, Ternamian C, Gorlia T, Diefes KL, Mischel PS, et al. Alkylpurine–DNA–N-glycosylase confers resistance to temozolomide in xenograft models of glioblastoma multiforme and is associated with poor survival in patients. J Clin Invest. 2012;122:253–66.

  72. Cho HR, Thakur N, Thakur N, Vu H, Kim H, Choi S. Decreased APE-1 by Nitroxoline enhances therapeutic effect in a Temozolomide-resistant Glioblastoma: correlation with diffusion weighted imaging. Sci Rep. 2019;9.

  73. Jiapaer S, Furuta T, Tanaka S, Kitabayashi T, Nakada M. Potential Strategies Overcoming the Temozolomide Resistance for Glioblastoma. Neurol. Med. Chir. (Tokyo). 2018;58:405–21.

    Article  Google Scholar 

  74. Ceccato, F.; Lombardi, G.; Albiger, N.; Mazzai, L.; Pambuku, A.; Rolma, G.; Zagonel, V.; Scaroni, C. Temozolomide cytoreductive treatment in a giant cabergoline-resistant prolactin-secreting pituitary neuroendocrine tumour Available online: https://www.ingentaconnect.com/content/wk/ancad/2019/00000030/00000005/art00013?crawler=true&mimetype=application/pdf (accessed on Jan 23, 2020).

  75. Whitelaw, B. How and when to use temozolomide to treat aggressive pituitary tumours. Endocr Relat Cancer 2019.

  76. Chakravarti A, Erkkinen MG, Nestler U, Stupp R, Mehta M, Aldape K, et al. Temozolomide-mediated radiation enhancement in glioblastoma: a report on underlying mechanisms. Clin Cancer Res. 2006;12:4738–46.

  77. Zhong C, Yin S, Zhou P, Jiang S. Pituitary atypical adenoma or carcinoma sensitive to temozolomide combined with radiation therapy: a case report of early identification and management. Turk Neurosurg. 2014;24:963–6.

    PubMed  Google Scholar 

  78. Kamiya-Matsuoka C, Cachia D, Waguespack SG, Crane CH, Mahajan A, Brown PD, et al. Radiotherapy with concurrent temozolomide for the management of extraneural metastases in pituitary carcinoma. Pituitary. 2016;19:415–21.

  79. Misir Krpan A, Dusek T, Rakusic Z, Solak M, Kraljevic I, Bisof V, et al. A rapid biochemical and radiological response to the concomitant therapy with Temozolomide and radiotherapy in an aggressive ACTH pituitary adenoma. Case Rep Endocrinol. 2017;2017.

  80. Ness, K.K.; Kirkland, J.L.; Gramatges, M.M.; Wang, Z.; Kundu, M.; McCastlain, K.; Li-Harms, X.; Zhang, J.; Tchkonia, T.; Pluijm, S.M.F.; et al. Premature Physiologic Aging as a Paradigm for Understanding Increased Risk of Adverse Health Across the Lifespan of Survivors of Childhood Cancer. Journal of Clinical Oncology 2018.

  81. Landman RE, Horwith M, Peterson RE, Khandji AG, Wardlaw SL. Long-term survival with ACTH-secreting carcinoma of the pituitary: a case report and review of the literature. J Clin Endocrinol Metab. 2002;87:3084–9.

    Article  CAS  PubMed  Google Scholar 

  82. van der Klaauw AA, Kienitz T, Strasburger CJ, Smit JWA, Romijn JA. Malignant pituitary corticotroph adenomas: report of two cases and a comprehensive review of the literature. Pituitary. 2009;12:57–69.

    Article  PubMed  Google Scholar 

  83. Weller M, Tabatabai G, Kästner B, Felsberg J, Steinbach JP, Wick A, et al. MGMT promoter methylation is a strong prognostic biomarker for benefit from dose-intensified Temozolomide Rechallenge in progressive Glioblastoma: the DIRECTOR trial. Clin Cancer Res. 2015;21:2057–64.

  84. Bilbao I, Egaña N, García C, Olaizola I. Failure of a second temozolomide cycle in a patient with a prolactin-secreting pituitary carcinoma. Endocrinol Diabetes Nutr. 2017;64:564–6.

    Article  PubMed  Google Scholar 

  85. Jordan JT, Miller JJ, Cushing T, Seijo M, Batchelor TT, Arrillaga-Romany IC, et al. Temozolomide therapy for aggressive functioning pituitary adenomas refractory to surgery and radiation: a case series. Neurooncol Pract. 2018;5:64–8.

  86. Goel HL, Mercurio AM. VEGF targets the tumour cell. Nat Rev Cancer. 2013;13:871–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Yang, J.; Yan, J.; Liu, B. Targeting VEGF/VEGFR to modulate antitumour immunity. Front Immunol 2018, 9.

  88. Ellis LM, Hicklin DJ. VEGF-targeted therapy: mechanisms of anti-tumour activity. Nat Rev Cancer. 2008;8:579–91.

    Article  CAS  PubMed  Google Scholar 

  89. Touma W, Hoostal S, Peterson R, Wiernik A, SantaCruz K, Lou E. Successful treatment of pituitary carcinoma with concurrent radiation, temozolomide, and bevacizumab after resection. J Clin Neurosci. 2017;41:75–7.

    Article  CAS  PubMed  Google Scholar 

  90. O’Riordan LM, Greally M, Coleman N, Breathnach OS, Hennessy B, Thompson CJ, et al. Metastatic ACTH-producing pituitary carcinoma managed with combination pasireotide and bevacizumab following failure of temozolamide therapy: a case report. JCO. 2013;31:e13022–2.

  91. Wang Y, He Q, Meng X, Zhou S, Zhu Y, Xu J, et al. Apatinib (YN968D1) and Temozolomide in recurrent invasive pituitary adenoma: case report and literature review | Elsevier enhanced reader. World Neurosurgery. 2019;124:319–22.

  92. Mei Y, Bi WL, Greenwald NF, Du Z, Agar NYR, Kaiser UB, et al. Increased expression of programmed death ligand 1 (PD-L1) in human pituitary tumours. Oncotarget. 2016;7:76565–76.

  93. Wang P, Wang T, Yang Y, Yao K, Li Z, Li YM, et al. The expression profile of PD-L1 and CD8+ lymphocyte in pituitary adenomas indicating for immunotherapy. J Neuro-Oncol. 2018;139:89–95.

  94. Wang P, Wang T, Yang Y, Yu C, Liu N, Yan C. Detection of programmed death ligand 1 protein and CD8+ lymphocyte infiltration in plurihormonal pituitary adenomas: a case report and review of the literatures. Medicine. 2017;96.

  95. Caccese M, Barbot M, Ceccato F, Padovan M, Gardiman MP, Fassan M, et al. Rapid disease progression in patient with mismatch-repair deficiency pituitary ACTH-secreting adenoma treated with checkpoint inhibitor pembrolizumab. Anti-Cancer Drugs. 2020;31:199–204.

  96. Heynckes S, Daka K, Franco P, Gaebelein A, Frenking JH, Doria-Medina R, et al. Crosslink between Temozolomide and PD-L1 immune-checkpoint inhibition in glioblastoma multiforme. BMC Cancer. 2019;19:117.

  97. Karachi A, Yang C, Dastmalchi F, Sayour JE, Huang J, Azari H, et al. Modulation of temozolomide dose differentially affects T-cell response to immune checkpoint inhibition. Neuro-Oncology. 2019;21:730–41.

  98. Mathios, D.; Kim, J.E.; Mangraviti, A.; Phallen, J.; Park, C.-K.; Jackson, C.M.; Garzon-Muvdi, T.; Kim, E.; Theodros, D.; Polanczyk, M.; et al. Anti–PD-1 antitumour immunity is enhanced by local and abrogated by systemic chemotherapy in GBM. Science Translational Medicine 2016, 8, 370ra180-370ra180.

  99. Schvartsman G, Peng SA, Bis G, Lee JJ, Benveniste MFK, Zhang J, et al. Response rates to single-agent chemotherapy after exposure to immune checkpoint inhibitors in advanced non-small cell lung cancer. Lung Cancer. 2017;112:90–5.

  100. Park SE, Lee SH, Ahn JS, Ahn M-J, Park K, Sun J-M. Increased response rates to salvage chemotherapy administered after PD-1/PD-L1 inhibitors in patients with non–small cell lung Cancer. J Thorac Oncol. 2018;13:106–11.

    Article  CAS  PubMed  Google Scholar 

  101. Brahmer JR, Rodriguez-Abreu D, Robinson AG, Hui R, Csõszi T, Fülöp A, et al. Progression after the next line of therapy (PFS2) and updated OS among patients (pts) with advanced NSCLC and PD-L1 tumour proportion score (TPS) ≥50% enrolled in KEYNOTE-024. JCO. 2017;35:9000–0.

  102. Dwary AD, Master S, Patel A, Cole C, Mansour R, Mills G, et al. Excellent response to chemotherapy post immunotherapy. Oncotarget. 2017;8:91795–802.

  103. Alsuwaigh R, Lee J, Chan G, Chee CE, Choo SP. Response to targeted therapy or chemotherapy following immunotherapy in patients with gastrointestinal cancers - a case series. Journal for ImmunoTherapy of Cancer. 2019;7:162.

    Article  PubMed  Google Scholar 

  104. Simon A, Kourie HR, Kerger J. Is there still a role for cytotoxic chemotherapy after targeted therapy and immunotherapy in metastatic melanoma? A case report and literature review Chinese Journal of Cancer. 2017;36:10.

    PubMed  Google Scholar 

  105. Wang J, Cazzato E, Ladewig E, Frattini V, Rosenbloom DIS, Zairis S, et al. Clonal evolution of glioblastoma under therapy. Nat Genet. 2016;48:768–76.

  106. Hunter C, Smith R, Cahill DP, Stephens P, Stevens C, Teague J, et al. A Hypermutation phenotype and somatic MSH6 mutations in recurrent human malignant Gliomas after Alkylator chemotherapy. Cancer Res. 2006;66:3987–91.

  107. Yip S, Miao J, Cahill DP, Iafrate AJ, Aldape K, Nutt CL, et al. MSH6 mutations arise in glioblastomas during temozolomide therapy and mediate temozolomide resistance. Clin Cancer Res. 2009;15:4622–9.

  108. Steuer CE, Ramalingam SS. Tumour mutation burden: leading immunotherapy to the era of precision medicine? JCO. 2018;36:631–2.

    Article  CAS  Google Scholar 

  109. Chan TA, Yarchoan M, Jaffee E, Swanton C, Quezada SA, Stenzinger A, et al. Development of tumour mutation burden as an immunotherapy biomarker: utility for the oncology clinic. Ann Oncol. 2019;30:44–56.

  110. Dutta P, Reddy KS, Rai A, Madugundu AK, Solanki HS, Bhansali A, et al. Surgery, Octreotide, Temozolomide, Bevacizumab, radiotherapy, and Pegvisomant treatment of an AIP mutation–positive child. J Clin Endocrinol Metab. 2019;104:3539–44.

Download references

Availability of data and material

Not applicable.

Code availability

Not applicable.

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed to and agreed to the published version of the manuscript.

Corresponding author

Correspondence to Ann McCormack.

Ethics declarations

Conflict of interest

No conflicts of interest to declare.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Burman, P., Lamb, L. & McCormack, A. Temozolomide therapy for aggressive pituitary tumours – current understanding and future perspectives. Rev Endocr Metab Disord 21, 263–276 (2020). https://doi.org/10.1007/s11154-020-09551-y

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11154-020-09551-y

Keywords

Navigation