Skip to main content
Top
Published in: Endocrine 2/2014

01-06-2014 | Review

Molecular basis of pharmacological therapy in Cushing’s disease

Authors: Diego Ferone, Claudia Pivonello, Giovanni Vitale, Maria Chiara Zatelli, Annamaria Colao, Rosario Pivonello

Published in: Endocrine | Issue 2/2014

Login to get access

Abstract

Cushing’s disease (CD) is a severe endocrine condition caused by an adrenocorticotropin (ACTH)-producing pituitary adenoma that chronically stimulates adrenocortical cortisol production and with potentially serious complications if not or inadequately treated. Active CD may produce a fourfold increase in mortality and is associated with significant morbidities. Moreover, excess mortality risk may persist even after CD treatment. Although predictors of risk in treated CD are not fully understood, the importance of early recognition and adequate treatment is well established. Surgery with resection of a pituitary adenoma is still the first line therapy, being successful in about 60–70 % of patients; however, recurrence within 2–4 years may often occur. When surgery fails, medical treatment can reduce cortisol production and ameliorate clinical manifestations while more definitive therapy becomes effective. Compounds that target hypothalamic–pituitary axis, glucocorticoid synthesis or adrenocortical function are currently used to control the deleterious effects of chronic glucocorticoid excess. In this review we describe and analyze the molecular basis of the drugs targeting the disease at central level, suppressing ACTH secretion, as well as at peripheral level, acting as adrenal inhibitors, or glucocorticoid receptor antagonists. Understanding of the underlying molecular mechanisms in CD and of glucocorticoid biology should promote the development of new targeted and more successful therapies in the future. Indeed, most of the drugs discussed have been tested in limited clinical trials, but there is potential therapeutic benefit in compounds with better specificity for the class of receptors expressed by ACTH-secreting tumors. However, long-term follow-up with management of persistent comorbidities is needed even after successful treatment of CD.
Appendix
Available only for authorised users
Literature
1.
go back to reference R. Pivonello, M.C. De Martino, M. De Leo, G. Lombardi, A. Colao, Cushing’s syndrome. Endocrinol. Metab. Clin. N. Am. 37, 135–149 (2008) R. Pivonello, M.C. De Martino, M. De Leo, G. Lombardi, A. Colao, Cushing’s syndrome. Endocrinol. Metab. Clin. N. Am. 37, 135–149 (2008)
2.
go back to reference B.M. Biller, A.B. Grossman, P.M. Stewart, S. Melmed, X. Bertagna, J. Bertherat, M. Buchfelder, A. Colao, A.R. Hermus, L.J. Hofland, A. Klibanski, A. Lacroix, J.R. Lindsay, J. Newell-Price, L.K. Nieman, S. Petersenn, N. Sonino, G.K. Stalla, B. Swearingen, M.L. Vance, J.A. Wass, M. Boscaro, Treatment of adrenocorticotropin-dependent Cushing’s syndrome: a consensus statement. J. Clin. Endocrinol. Metab. 93, 2454–2462 (2008)PubMedCentralPubMed B.M. Biller, A.B. Grossman, P.M. Stewart, S. Melmed, X. Bertagna, J. Bertherat, M. Buchfelder, A. Colao, A.R. Hermus, L.J. Hofland, A. Klibanski, A. Lacroix, J.R. Lindsay, J. Newell-Price, L.K. Nieman, S. Petersenn, N. Sonino, G.K. Stalla, B. Swearingen, M.L. Vance, J.A. Wass, M. Boscaro, Treatment of adrenocorticotropin-dependent Cushing’s syndrome: a consensus statement. J. Clin. Endocrinol. Metab. 93, 2454–2462 (2008)PubMedCentralPubMed
3.
go back to reference J.S. Lim, S.K. Lee, S.H. Kim, E.J. Lee, S.H. Kim, Intraoperative multiple-staged resection and tumor tissue identification using frozen sections provide the best result for the accurate localization and complete resection of tumors in Cushing’s disease. Endocrine 40, 452–461 (2011)PubMed J.S. Lim, S.K. Lee, S.H. Kim, E.J. Lee, S.H. Kim, Intraoperative multiple-staged resection and tumor tissue identification using frozen sections provide the best result for the accurate localization and complete resection of tumors in Cushing’s disease. Endocrine 40, 452–461 (2011)PubMed
4.
go back to reference A.E. Calogero, Neurotransmitter regulation of the hypothalamic corticotropin-releasing hormone neuron. Ann. N. Y. Acad. Sci. 771, 31–40 (1995)PubMed A.E. Calogero, Neurotransmitter regulation of the hypothalamic corticotropin-releasing hormone neuron. Ann. N. Y. Acad. Sci. 771, 31–40 (1995)PubMed
5.
go back to reference R. Giordano, M. Pellegrino, A. Picu, L. Bonelli, M. Balbo, R. Berardelli, F. Lanfranco, E. Ghigo, E. Arvat, Neuroregulation of the hypothalamus–pituitary–adrenal (HPA) axis in humans: effects of GABA-, mineralocorticoid-, and GH-secretagogue-receptor modulation. Sci. World J. 6, 1–11 (2006) R. Giordano, M. Pellegrino, A. Picu, L. Bonelli, M. Balbo, R. Berardelli, F. Lanfranco, E. Ghigo, E. Arvat, Neuroregulation of the hypothalamus–pituitary–adrenal (HPA) axis in humans: effects of GABA-, mineralocorticoid-, and GH-secretagogue-receptor modulation. Sci. World J. 6, 1–11 (2006)
6.
go back to reference R.W. Fuller, The involvement of serotonin in regulation of pituitary–adrenocortical function. Front. Neuroendocrinol. 13, 250–270 (1992)PubMed R.W. Fuller, The involvement of serotonin in regulation of pituitary–adrenocortical function. Front. Neuroendocrinol. 13, 250–270 (1992)PubMed
7.
go back to reference B. Ambrosi, M. Gaggini, F. Secchi, G. Faglia, Lack of effect of antiserotoninergic and/or dopaminergic treatment in patients with pituitary-dependent Cushing’s syndrome. Horm. Metab. Res. 11, 318–319 (1979)PubMed B. Ambrosi, M. Gaggini, F. Secchi, G. Faglia, Lack of effect of antiserotoninergic and/or dopaminergic treatment in patients with pituitary-dependent Cushing’s syndrome. Horm. Metab. Res. 11, 318–319 (1979)PubMed
8.
go back to reference F. Cavagnini, U. Raggi, P. Micossi, A. Di Landro, C. Invitti, Effect of an antiserotoninergic drug, metergoline, on the ACTH and cortisol response to insulin hypoglycemia and lysine-vasopressin in man. J. Clin. Endocrinol. Metab. 43, 306–312 (1976)PubMed F. Cavagnini, U. Raggi, P. Micossi, A. Di Landro, C. Invitti, Effect of an antiserotoninergic drug, metergoline, on the ACTH and cortisol response to insulin hypoglycemia and lysine-vasopressin in man. J. Clin. Endocrinol. Metab. 43, 306–312 (1976)PubMed
9.
go back to reference N. Sonino, G.A. Fava, F. Fallo, A. Franceschetto, P. Belluardo, M. Boscaro, Effect of the serotonin antagonists ritanserin and ketanserin in Cushing’s disease. Pituitary 3, 55–59 (2000)PubMed N. Sonino, G.A. Fava, F. Fallo, A. Franceschetto, P. Belluardo, M. Boscaro, Effect of the serotonin antagonists ritanserin and ketanserin in Cushing’s disease. Pituitary 3, 55–59 (2000)PubMed
10.
go back to reference R. Tanakol, F. Alagöl, H. Azizlerli, O. Sandalci, T. Terzioğlu, F. Berker, Cyproheptadine treatment in Cushing’s disease. J. Endocrinol. Invest. 19, 242–247 (1996)PubMed R. Tanakol, F. Alagöl, H. Azizlerli, O. Sandalci, T. Terzioğlu, F. Berker, Cyproheptadine treatment in Cushing’s disease. J. Endocrinol. Invest. 19, 242–247 (1996)PubMed
11.
go back to reference K.I. Alexandraki, A.B. Grossman, Pituitary-targeted medical therapy of Cushing’s disease. Expert Opin. Investig. Drugs 17, 669–677 (2008)PubMed K.I. Alexandraki, A.B. Grossman, Pituitary-targeted medical therapy of Cushing’s disease. Expert Opin. Investig. Drugs 17, 669–677 (2008)PubMed
12.
go back to reference T. Suda, F. Tozawa, T. Mouri, A. Sasaki, T. Shibasaki, H. Demura, K. Shizume, Effects of cyproheptadine, reserpine, and synthetic corticotropin-releasing factor on pituitary glands from patients with Cushing’s disease. J. Clin. Endocrinol. Metab. 56, 1094–1099 (1983)PubMed T. Suda, F. Tozawa, T. Mouri, A. Sasaki, T. Shibasaki, H. Demura, K. Shizume, Effects of cyproheptadine, reserpine, and synthetic corticotropin-releasing factor on pituitary glands from patients with Cushing’s disease. J. Clin. Endocrinol. Metab. 56, 1094–1099 (1983)PubMed
13.
go back to reference D.T. Krieger, L. Amorosa, F. Linick, Cyproheptadine-induced remission of Cushing’s disease. N. Engl. J. Med. 293, 893–896 (1975)PubMed D.T. Krieger, L. Amorosa, F. Linick, Cyproheptadine-induced remission of Cushing’s disease. N. Engl. J. Med. 293, 893–896 (1975)PubMed
14.
go back to reference N.G. Bowery, D.R. Hill, A.L. Hudson, G.W. Price, W.J. Turnbull, G.P. Wilson, Heterogeneity of mammalian GABA receptors, in Actions and interactions of GABA and benzodiazepine, ed. by N.G. Bowery (Raven Press, New York, 1984), pp. 81–108 N.G. Bowery, D.R. Hill, A.L. Hudson, G.W. Price, W.J. Turnbull, G.P. Wilson, Heterogeneity of mammalian GABA receptors, in Actions and interactions of GABA and benzodiazepine, ed. by N.G. Bowery (Raven Press, New York, 1984), pp. 81–108
15.
go back to reference J. Takahara, S. Yumoki, W. Yakushji, J. Yamauchi, H. Hosogi, T. Ofuji, Stimulatory effects of gamma-aminohydroxybutyric acid (GABOB) on growth hormone, prolactin and cortisol release in man. Horm. Metab. Res. 12, 31–34 (1980)PubMed J. Takahara, S. Yumoki, W. Yakushji, J. Yamauchi, H. Hosogi, T. Ofuji, Stimulatory effects of gamma-aminohydroxybutyric acid (GABOB) on growth hormone, prolactin and cortisol release in man. Horm. Metab. Res. 12, 31–34 (1980)PubMed
16.
go back to reference P.M. Plotsky, S. Otto, S. Sutton, Neurotransmitter modulation of corticotropin releasing factor secretion into the hypophysial-portal circulation. Life Sci. 41, 1311–1317 (1987)PubMed P.M. Plotsky, S. Otto, S. Sutton, Neurotransmitter modulation of corticotropin releasing factor secretion into the hypophysial-portal circulation. Life Sci. 41, 1311–1317 (1987)PubMed
17.
go back to reference H.P. Koppeschaar, R.J. Croughs, J.H. Thijssen, F. Schwarz, Sodium valproate and cyproheptadine may independently induce a remission in the same patient with Cushing’s disease. Acta Endocrinol. (Copenh). 104, 160–163 (1983)PubMed H.P. Koppeschaar, R.J. Croughs, J.H. Thijssen, F. Schwarz, Sodium valproate and cyproheptadine may independently induce a remission in the same patient with Cushing’s disease. Acta Endocrinol. (Copenh). 104, 160–163 (1983)PubMed
18.
go back to reference A. Beckers, A. Stevenaert, G. Pirens, P. Flandroy, J. Sulon, G. Hennen, Cyclical Cushing’s disease and its successful control under sodium valproate. J. Endocrinol. Invest. 13, 923–929 (1990)PubMed A. Beckers, A. Stevenaert, G. Pirens, P. Flandroy, J. Sulon, G. Hennen, Cyclical Cushing’s disease and its successful control under sodium valproate. J. Endocrinol. Invest. 13, 923–929 (1990)PubMed
19.
go back to reference A. Colao, R. Pivonello, F.S. Tripodi, F. Orio Jr, D. Ferone, G. Cerbone, C. Di Somma, B. Merola, G. Lombardi, Failure of long-term therapy with sodium valproate in Cushing’s disease. J. Endocrinol. Invest. 20, 387–392 (1997)PubMed A. Colao, R. Pivonello, F.S. Tripodi, F. Orio Jr, D. Ferone, G. Cerbone, C. Di Somma, B. Merola, G. Lombardi, Failure of long-term therapy with sodium valproate in Cushing’s disease. J. Endocrinol. Invest. 20, 387–392 (1997)PubMed
20.
go back to reference S.S. Nussey, P. Price, J.S. Jenkins, A.R. Altaher, B. Gillham, M.T. Jones, The combined use of sodium valproate and metyrapone in the treatment of Cushing’s syndrome. Clin. Endocrinol. (Oxf). 28, 373–380 (1988)PubMed S.S. Nussey, P. Price, J.S. Jenkins, A.R. Altaher, B. Gillham, M.T. Jones, The combined use of sodium valproate and metyrapone in the treatment of Cushing’s syndrome. Clin. Endocrinol. (Oxf). 28, 373–380 (1988)PubMed
21.
go back to reference M. Mannelli, G. Cantini, G. Poli, M. Mangoni, G. Nesi, L. Canu, E. Rapizzi, E. Borgogni, T. Ercolino, V. Piccini, M. Luconi, Role of the PPAR-γ system in normal and tumoral pituitary corticotropic cells and adrenal cells. Neuroendocrinology 92(Suppl 1), 23–27 (2010)PubMed M. Mannelli, G. Cantini, G. Poli, M. Mangoni, G. Nesi, L. Canu, E. Rapizzi, E. Borgogni, T. Ercolino, V. Piccini, M. Luconi, Role of the PPAR-γ system in normal and tumoral pituitary corticotropic cells and adrenal cells. Neuroendocrinology 92(Suppl 1), 23–27 (2010)PubMed
22.
go back to reference A.P. Heaney, M. Fernando, W.H. Yong, S. Melmed, Functional PPAR-gamma receptor is a novel therapeutic target for ACTH-secreting pituitary adenomas. Nat. Med. 8, 1281–1287 (2002)PubMed A.P. Heaney, M. Fernando, W.H. Yong, S. Melmed, Functional PPAR-gamma receptor is a novel therapeutic target for ACTH-secreting pituitary adenomas. Nat. Med. 8, 1281–1287 (2002)PubMed
23.
go back to reference K. Winczyk, J. Kunert-Radek, A. Gruszka, M. Radek, H. Ławnicka, M. Pawlikowski, Effects of rosiglitazone-peroxisome proliferators-activated receptor gamma (PPARgamma) agonist on cell viability of human pituitary adenomas in vitro. Neuro. Endocrinol. Lett. 30, 107–110 (2009)PubMed K. Winczyk, J. Kunert-Radek, A. Gruszka, M. Radek, H. Ławnicka, M. Pawlikowski, Effects of rosiglitazone-peroxisome proliferators-activated receptor gamma (PPARgamma) agonist on cell viability of human pituitary adenomas in vitro. Neuro. Endocrinol. Lett. 30, 107–110 (2009)PubMed
24.
go back to reference L. Kreutzer, I. Jeske, B. Hofmann, I. Blumcke, R. Fahlbusch, M. Buchfelder, R. Buslei, No effect of the PPAR-gamma agonist rosiglitazone on ACTH or cortisol secretion in Nelson’s syndrome and Cushing’s disease in vitro and in vivo. Clin. Neuropathol. 28, 430–439 (2009)PubMed L. Kreutzer, I. Jeske, B. Hofmann, I. Blumcke, R. Fahlbusch, M. Buchfelder, R. Buslei, No effect of the PPAR-gamma agonist rosiglitazone on ACTH or cortisol secretion in Nelson’s syndrome and Cushing’s disease in vitro and in vivo. Clin. Neuropathol. 28, 430–439 (2009)PubMed
25.
go back to reference M. Manning, A. Misicka, A. Olma, K. Bankowski, S. Stoev, B. Chini, T. Durroux, B. Mouillac, M. Corbani, G. Guillon, Oxytocin and vasopressin agonists and antagonists as research tools and potential therapeutics. J. Neuroendocrinol. 24, 609–628 (2012)PubMedCentralPubMed M. Manning, A. Misicka, A. Olma, K. Bankowski, S. Stoev, B. Chini, T. Durroux, B. Mouillac, M. Corbani, G. Guillon, Oxytocin and vasopressin agonists and antagonists as research tools and potential therapeutics. J. Neuroendocrinol. 24, 609–628 (2012)PubMedCentralPubMed
26.
go back to reference G. Narayen, S.N. Mandal, Vasopressin receptor antagonists and their role in clinical medicine. Indian J. Endocrinol. Metab. 16, 183–191 (2012)PubMedCentralPubMed G. Narayen, S.N. Mandal, Vasopressin receptor antagonists and their role in clinical medicine. Indian J. Endocrinol. Metab. 16, 183–191 (2012)PubMedCentralPubMed
27.
go back to reference D.E. Schteingart, Drugs in the medical treatment of Cushing’s syndrome. Expert Opin. Emerg. Drugs 14, 661–671 (2009)PubMed D.E. Schteingart, Drugs in the medical treatment of Cushing’s syndrome. Expert Opin. Emerg. Drugs 14, 661–671 (2009)PubMed
28.
go back to reference M. Páez-Pereda, D. Kovalovsky, U. Hopfner, M. Theodoropoulou, U. Pagotto, E. Uhl, M. Losa, J. Stalla, Y. Grübler, C. Missale, E. Arzt, G.K. Stalla, Retinoic acid prevents experimental Cushing syndrome. J. Clin. Invest. 108, 1123–1131 (2001)PubMedCentralPubMed M. Páez-Pereda, D. Kovalovsky, U. Hopfner, M. Theodoropoulou, U. Pagotto, E. Uhl, M. Losa, J. Stalla, Y. Grübler, C. Missale, E. Arzt, G.K. Stalla, Retinoic acid prevents experimental Cushing syndrome. J. Clin. Invest. 108, 1123–1131 (2001)PubMedCentralPubMed
29.
go back to reference D. Giacomini, M. Páez-Pereda, M. Theodoropoulou, M. Labeur, D. Refojo, J. Gerez, A. Chervin, S. Berner, M. Losa, M. Buchfelder, U. Renner, G.K. Stalla, E. Arzt, Bone morphogenetic protein-4 inhibits corticotroph tumor cells: involvement in the retinoic acid inhibitory action. Endocrinology 147, 247–256 (2006)PubMed D. Giacomini, M. Páez-Pereda, M. Theodoropoulou, M. Labeur, D. Refojo, J. Gerez, A. Chervin, S. Berner, M. Losa, M. Buchfelder, U. Renner, G.K. Stalla, E. Arzt, Bone morphogenetic protein-4 inhibits corticotroph tumor cells: involvement in the retinoic acid inhibitory action. Endocrinology 147, 247–256 (2006)PubMed
30.
go back to reference V. Castillo, D. Giacomini, M. Páez-Pereda, J. Stalla, M. Labeur, M. Theodoropoulou, F. Holsboer, A.B. Grossman, G.K. Stalla, E. Arzt, Retinoic acid as a novel medical therapy for Cushing’s disease in dogs. Endocrinology 147, 4438–4444 (2006)PubMed V. Castillo, D. Giacomini, M. Páez-Pereda, J. Stalla, M. Labeur, M. Theodoropoulou, F. Holsboer, A.B. Grossman, G.K. Stalla, E. Arzt, Retinoic acid as a novel medical therapy for Cushing’s disease in dogs. Endocrinology 147, 4438–4444 (2006)PubMed
31.
go back to reference F. Pecori Giraldi, A.G. Ambrogio, M. Andrioli, F. Sanguin, I. Karamouzis, S.M. Corsello, C. Scaroni, E. Arvat, A. Pontecorvi, F. Cavagnini, Potential role for retinoic acid in patients with Cushing’s disease. J. Clin. Endocrinol. Metab. 97, 3577–3583 (2012)PubMed F. Pecori Giraldi, A.G. Ambrogio, M. Andrioli, F. Sanguin, I. Karamouzis, S.M. Corsello, C. Scaroni, E. Arvat, A. Pontecorvi, F. Cavagnini, Potential role for retinoic acid in patients with Cushing’s disease. J. Clin. Endocrinol. Metab. 97, 3577–3583 (2012)PubMed
32.
go back to reference R.M. Luque, M.D. Gahete, U. Hochgeschwender, R.D. Kineman, Evidence that endogenous SST inhibits ACTH and ghrelin expression by independent pathways. Am. J. Physiol. Endocrinol. Metab. 291, E395–E403 (2006)PubMed R.M. Luque, M.D. Gahete, U. Hochgeschwender, R.D. Kineman, Evidence that endogenous SST inhibits ACTH and ghrelin expression by independent pathways. Am. J. Physiol. Endocrinol. Metab. 291, E395–E403 (2006)PubMed
33.
go back to reference D. Cervia, D. Fehlmann, D. Hoyer, Native somatostatin sst2 and sst5 receptors functionally coupled to Gi/o-protein, but not to the serum response element in AtT-20 mouse tumour corticotrophs. Naunyn Schmiedebergs Arch. Pharmacol. 367, 387–578 (2003) D. Cervia, D. Fehlmann, D. Hoyer, Native somatostatin sst2 and sst5 receptors functionally coupled to Gi/o-protein, but not to the serum response element in AtT-20 mouse tumour corticotrophs. Naunyn Schmiedebergs Arch. Pharmacol. 367, 387–578 (2003)
34.
go back to reference M.Z. Strowski, M.P. Dashkevicz, R.M. Parmar, H. Wilkinson, M. Kohler, J.M. Schaeffer, A.D. Blake, Somatostatin receptor subtypes 2 and 5 inhibit corticotropin-releasing hormone-stimulated adrenocorticotropin secretion from AtT-20 cells. Neuroendocrinology 75, 339–346 (2002)PubMed M.Z. Strowski, M.P. Dashkevicz, R.M. Parmar, H. Wilkinson, M. Kohler, J.M. Schaeffer, A.D. Blake, Somatostatin receptor subtypes 2 and 5 inhibit corticotropin-releasing hormone-stimulated adrenocorticotropin secretion from AtT-20 cells. Neuroendocrinology 75, 339–346 (2002)PubMed
35.
go back to reference S. Nielsen, S. Mellemkjaer, L.M. Rasmussen, T. Ledet, N. Olsen, M. Bojsen-Møller, J. Astrup, J. Weeke, J.O. Jørgensen, Expression of somatostatin receptors on human pituitary adenomas in vivo and ex vivo. J. Endocrinol. Invest. 24, 430–437 (2001)PubMed S. Nielsen, S. Mellemkjaer, L.M. Rasmussen, T. Ledet, N. Olsen, M. Bojsen-Møller, J. Astrup, J. Weeke, J.O. Jørgensen, Expression of somatostatin receptors on human pituitary adenomas in vivo and ex vivo. J. Endocrinol. Invest. 24, 430–437 (2001)PubMed
36.
go back to reference U.I. Richardson, A. Schonbrunn, Inhibition of adrenocorticotropin secretion by somatostatin in pituitary cells in culture. Endocrinology 108, 281–290 (1981)PubMed U.I. Richardson, A. Schonbrunn, Inhibition of adrenocorticotropin secretion by somatostatin in pituitary cells in culture. Endocrinology 108, 281–290 (1981)PubMed
37.
go back to reference A. Ben-Shlomo, K.A. Wawrowsky, I. Proekt, N.M. Wolkenfeld, S.R. Ren, J. Taylor, M.D. Culler, S. Melmed, Somatostatin receptor type 5 modulates somatostatin receptor type 2 regulation of adrenocorticotropin secretion. J. Biol. Chem. 280, 24011–24021 (2005)PubMed A. Ben-Shlomo, K.A. Wawrowsky, I. Proekt, N.M. Wolkenfeld, S.R. Ren, J. Taylor, M.D. Culler, S. Melmed, Somatostatin receptor type 5 modulates somatostatin receptor type 2 regulation of adrenocorticotropin secretion. J. Biol. Chem. 280, 24011–24021 (2005)PubMed
38.
go back to reference J. van der Hoek, M. Waaijers, P.M. van Koetsveld, D. Sprij-Mooij, R.A. Feelders, H.A. Schmid, P. Schoeffter, D. Hoyer, D. Cervia, J.E. Taylor, M.D. Culler, S.W. Lamberts, L.J. Hofland, Distinct functional properties of native somatostatin receptor subtype 5 compared with subtype 2 in the regulation of ACTH release by corticotroph tumor cells. Am. J. Physiol. Endocrinol. Metab. 289, E278–E287 (2005)PubMed J. van der Hoek, M. Waaijers, P.M. van Koetsveld, D. Sprij-Mooij, R.A. Feelders, H.A. Schmid, P. Schoeffter, D. Hoyer, D. Cervia, J.E. Taylor, M.D. Culler, S.W. Lamberts, L.J. Hofland, Distinct functional properties of native somatostatin receptor subtype 5 compared with subtype 2 in the regulation of ACTH release by corticotroph tumor cells. Am. J. Physiol. Endocrinol. Metab. 289, E278–E287 (2005)PubMed
39.
go back to reference L.J. Hofland, J. van der Hoek, R. Feelders, M.O. van Aken, P.M. van Koetsveld, M. Waaijers, D. Sprij-Mooij, C. Bruns, G. Weckbecker, W.W. de Herder, A. Beckers, S.W. Lamberts, The multi-ligand somatostatin analogue SOM230 inhibits ACTH secretion by cultured human corticotroph adenomas via somatostatin receptor type 5. Eur. J. Endocrinol. 152, 645–654 (2005)PubMed L.J. Hofland, J. van der Hoek, R. Feelders, M.O. van Aken, P.M. van Koetsveld, M. Waaijers, D. Sprij-Mooij, C. Bruns, G. Weckbecker, W.W. de Herder, A. Beckers, S.W. Lamberts, The multi-ligand somatostatin analogue SOM230 inhibits ACTH secretion by cultured human corticotroph adenomas via somatostatin receptor type 5. Eur. J. Endocrinol. 152, 645–654 (2005)PubMed
40.
go back to reference M.C. Zatelli, D. Piccin, C. Vignali, F. Tagliati, M.R. Ambrosio, M. Bondanelli, V. Cimino, A. Bianchi, H.A. Schmid, M. Scanarini, A. Pontecorvi, L. De Marinis, G. Maira, E.C. degli Uberti, 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, 91–102 (2007)PubMed M.C. Zatelli, D. Piccin, C. Vignali, F. Tagliati, M.R. Ambrosio, M. Bondanelli, V. Cimino, A. Bianchi, H.A. Schmid, M. Scanarini, A. Pontecorvi, L. De Marinis, G. Maira, E.C. degli Uberti, 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, 91–102 (2007)PubMed
41.
go back to reference A.P. Silva, P. Schoeffter, G. Weckbecker, C. Bruns, H.A. Schmid, Regulation of CRH-induced secretion of ACTH and corticosterone by SOM230 in rats. Eur. J. Endocrinol. 153, R7–R10 (2005)PubMed A.P. Silva, P. Schoeffter, G. Weckbecker, C. Bruns, H.A. Schmid, Regulation of CRH-induced secretion of ACTH and corticosterone by SOM230 in rats. Eur. J. Endocrinol. 153, R7–R10 (2005)PubMed
42.
go back to reference L.A. Nolan, H.A. Schmid, A. Levy, Octreotide and the novel multi-receptor ligand somatostatin receptor agonist pasireotide (SOM230), block the adrenalectomy-induced increase in mitotic activity in male rat anterior pituitary. Endocrinology 148, 2821–2827 (2007)PubMed L.A. Nolan, H.A. Schmid, A. Levy, Octreotide and the novel multi-receptor ligand somatostatin receptor agonist pasireotide (SOM230), block the adrenalectomy-induced increase in mitotic activity in male rat anterior pituitary. Endocrinology 148, 2821–2827 (2007)PubMed
43.
go back to reference J. van der Hoek, S.W. Lamberts, L.J. Hofland, The role of somatostatin analogs in Cushing’s disease. Pituitary 7, 257–264 (2004)PubMed J. van der Hoek, S.W. Lamberts, L.J. Hofland, The role of somatostatin analogs in Cushing’s disease. Pituitary 7, 257–264 (2004)PubMed
44.
go back to reference L.J. Hofland, S.W.J. Lamberts, R.A. Feelders, Role of somatostatin receptors in normal and tumoral pituitary corticotropic cells. Neuroendocrinology 92(suppl 1), 11–16 (2010)PubMed L.J. Hofland, S.W.J. Lamberts, R.A. Feelders, Role of somatostatin receptors in normal and tumoral pituitary corticotropic cells. Neuroendocrinology 92(suppl 1), 11–16 (2010)PubMed
45.
go back to reference M.R. Ambrosio, M. Campo, M.C. Zatelli, S.G. Cella, G. Trasforini, A. Margutti, A.E. Rigamonti, E.E. Müller, E.C. degli Uberti, Unexpected activation of pituitary-adrenal axis in healthy young and elderly subjects during somatostatin infusion. Neuroendocrinology 68, 123–128 (1998)PubMed M.R. Ambrosio, M. Campo, M.C. Zatelli, S.G. Cella, G. Trasforini, A. Margutti, A.E. Rigamonti, E.E. Müller, E.C. degli Uberti, Unexpected activation of pituitary-adrenal axis in healthy young and elderly subjects during somatostatin infusion. Neuroendocrinology 68, 123–128 (1998)PubMed
46.
go back to reference C. Invitti, F. Pecori Giraldi, A. Dubini, M. Piolini, F. Cavagnini, Effect of Sandostatin on CRF-stimulated secretion of ACTH, beta-lipotropin and beta-endorphin. Horm. Metab. Res. 23, 233–235 (1991)PubMed C. Invitti, F. Pecori Giraldi, A. Dubini, M. Piolini, F. Cavagnini, Effect of Sandostatin on CRF-stimulated secretion of ACTH, beta-lipotropin and beta-endorphin. Horm. Metab. Res. 23, 233–235 (1991)PubMed
47.
go back to reference P.J. Stafford, P.J. Kopelman, K. Davidson, L. McLoughlin, A. White, L.H. Rees, G.M. Besser, D.H. Coy, A. Grossman, The pituitary-adrenal response to CRF-41 is unaltered by intravenous somatostatin in normal subjects. Clin. Endocrinol. (Oxf). 30, 661–666 (1989)PubMed P.J. Stafford, P.J. Kopelman, K. Davidson, L. McLoughlin, A. White, L.H. Rees, G.M. Besser, D.H. Coy, A. Grossman, The pituitary-adrenal response to CRF-41 is unaltered by intravenous somatostatin in normal subjects. Clin. Endocrinol. (Oxf). 30, 661–666 (1989)PubMed
48.
go back to reference H.L. Fehm, K.H. Voigt, R. Lang, K.E. Beinert, S. Raptis, E.F. Pfeiffer, Somatostatin: a potent inhibitor of ACTH-hypersecretion in adrenal insufficiency. Klin Wochenschr. 54, 173–175 (1976)PubMed H.L. Fehm, K.H. Voigt, R. Lang, K.E. Beinert, S. Raptis, E.F. Pfeiffer, Somatostatin: a potent inhibitor of ACTH-hypersecretion in adrenal insufficiency. Klin Wochenschr. 54, 173–175 (1976)PubMed
49.
go back to reference G. Benker, K. Hackenberg, B. Hamburger, D. Reinwein, Effects of growth hormone release-inhibiting hormone and bromocryptine (CB 154) in states of abnormal pituitary-adrenal function. Clin. Endocrinol. (Oxf). 5, 187–190 (1976)PubMed G. Benker, K. Hackenberg, B. Hamburger, D. Reinwein, Effects of growth hormone release-inhibiting hormone and bromocryptine (CB 154) in states of abnormal pituitary-adrenal function. Clin. Endocrinol. (Oxf). 5, 187–190 (1976)PubMed
50.
go back to reference G.K. Stalla, S.J. Brockmeier, U. Renner, C. Newton, M. Buchfelder, J. Stalla, O.A. Müller, Octreotide exerts different effects in vivo and in vitro in Cushing’s disease. Eur. J. Endocrinol. 130, 125–131 (1994)PubMed G.K. Stalla, S.J. Brockmeier, U. Renner, C. Newton, M. Buchfelder, J. Stalla, O.A. Müller, Octreotide exerts different effects in vivo and in vitro in Cushing’s disease. Eur. J. Endocrinol. 130, 125–131 (1994)PubMed
51.
go back to reference B. Ambrosi, D. Bochicchio, C. Fadin, P. Colombo, G. Faglia, Failure of somatostatin and octreotide to acutely affect the hypothalamic–pituitary–adrenal function in patients with corticotropin hypersecretion. J. Endocrinol. Invest. 13, 257–261 (1990)PubMed B. Ambrosi, D. Bochicchio, C. Fadin, P. Colombo, G. Faglia, Failure of somatostatin and octreotide to acutely affect the hypothalamic–pituitary–adrenal function in patients with corticotropin hypersecretion. J. Endocrinol. Invest. 13, 257–261 (1990)PubMed
52.
go back to reference S.W. Lamberts, J. Zuyderwijk, F. den Holder, P. van Koetsveld, L. Hofland, Studies on the conditions determining the inhibitory effect of somatostatin on adrenocorticotropin, prolactin and thyrotropin release by cultured rat pituitary cells. Neuroendocrinology 50, 44–50 (1989)PubMed S.W. Lamberts, J. Zuyderwijk, F. den Holder, P. van Koetsveld, L. Hofland, Studies on the conditions determining the inhibitory effect of somatostatin on adrenocorticotropin, prolactin and thyrotropin release by cultured rat pituitary cells. Neuroendocrinology 50, 44–50 (1989)PubMed
53.
go back to reference J. Julesz, F. Laczi, T. Janáky, F. László, Effects of somatostatin and bromocryptine on the plasma ACTH level in bilaterally adrenalectomized patients with Cushing’s disease. Endokrinologie 76, 68–72 (1980)PubMed J. Julesz, F. Laczi, T. Janáky, F. László, Effects of somatostatin and bromocryptine on the plasma ACTH level in bilaterally adrenalectomized patients with Cushing’s disease. Endokrinologie 76, 68–72 (1980)PubMed
54.
go back to reference C. de Bruin, R.A. Feelders, A.M. Waaijers, P.M. van Koetsveld, D.M. Sprij-Mooij, S.W.J. Lamberts, L.J. Hofland, Differential regulation of human dopamine D2 and somatostatin receptor subtype expression by glucocorticoids in vitro. J. Mol. Endocrinol. 42, 47–56 (2009)PubMed C. de Bruin, R.A. Feelders, A.M. Waaijers, P.M. van Koetsveld, D.M. Sprij-Mooij, S.W.J. Lamberts, L.J. Hofland, Differential regulation of human dopamine D2 and somatostatin receptor subtype expression by glucocorticoids in vitro. J. Mol. Endocrinol. 42, 47–56 (2009)PubMed
55.
go back to reference A. Saveanu, P. Jaquet, Somatostatin-dopamine ligands in the treatment of pituitary adenomas. Rev. Endocr. Metab. Disord. 10, 83–90 (2009)PubMed A. Saveanu, P. Jaquet, Somatostatin-dopamine ligands in the treatment of pituitary adenomas. Rev. Endocr. Metab. Disord. 10, 83–90 (2009)PubMed
56.
go back to reference C. de Bruin, A.M. Pereira, R.A. Feelders, J.A. Romijn, F. Roelfsema, D.M. Sprij-Mooij, M.O. van Aken, A.J. van der Lelij, W.W. de Herder, S.W. Lamberts, L.J. Hofland, Coexpression of dopamine and somatostatin receptor subtypes in corticotroph adenomas. J. Clin. Endocrinol. Metab. 94, 1118–1124 (2009)PubMed C. de Bruin, A.M. Pereira, R.A. Feelders, J.A. Romijn, F. Roelfsema, D.M. Sprij-Mooij, M.O. van Aken, A.J. van der Lelij, W.W. de Herder, S.W. Lamberts, L.J. Hofland, Coexpression of dopamine and somatostatin receptor subtypes in corticotroph adenomas. J. Clin. Endocrinol. Metab. 94, 1118–1124 (2009)PubMed
57.
go back to reference T. Tateno, M. Kato, Y. Tani, K. Oyama, S. Yamada, Y. Hirata, Differential expression of somatostatin and dopamine receptor subtype genes in adrenocorticotropin (ACTH)-secreting pituitary tumors and silent corticotroph adenomas. Endocr. J. 56, 579–584 (2009)PubMed T. Tateno, M. Kato, Y. Tani, K. Oyama, S. Yamada, Y. Hirata, Differential expression of somatostatin and dopamine receptor subtype genes in adrenocorticotropin (ACTH)-secreting pituitary tumors and silent corticotroph adenomas. Endocr. J. 56, 579–584 (2009)PubMed
58.
go back to reference D.L. Batista, X. Zhang, R. Gejman, P.J. Ansell, Y. Zhou, S.A. Johnson, B. Swearingen, E.T. Hedley-Whyte, C.A. Stratakis, A. Klibanski, The effects of SOM230 on cell proliferation and adrenocorticotropin secretion in human corticotroph pituitary adenomas. J. Clin. Endocrinol. Metab. 91, 4482–4488 (2006)PubMed D.L. Batista, X. Zhang, R. Gejman, P.J. Ansell, Y. Zhou, S.A. Johnson, B. Swearingen, E.T. Hedley-Whyte, C.A. Stratakis, A. Klibanski, The effects of SOM230 on cell proliferation and adrenocorticotropin secretion in human corticotroph pituitary adenomas. J. Clin. Endocrinol. Metab. 91, 4482–4488 (2006)PubMed
59.
go back to reference E. Hubina, A.M. Nanzer, M.R. Hanson, E. Ciccarelli, M. Losa, D. Gaia, M. Papotti, M.R. Terreni, S. Khalaf, S. Jordan, S. Czirják, Z. Hanzély, G.M. Nagy, M.I. Góth, A.B. Grossman, M. Korbonits, Somatostatin analogues stimulate p27 expression and inhibit the MAP kinase pathway in pituitary tumours. Eur. J. Endocrinol. 155, 371–379 (2006)PubMed E. Hubina, A.M. Nanzer, M.R. Hanson, E. Ciccarelli, M. Losa, D. Gaia, M. Papotti, M.R. Terreni, S. Khalaf, S. Jordan, S. Czirják, Z. Hanzély, G.M. Nagy, M.I. Góth, A.B. Grossman, M. Korbonits, Somatostatin analogues stimulate p27 expression and inhibit the MAP kinase pathway in pituitary tumours. Eur. J. Endocrinol. 155, 371–379 (2006)PubMed
60.
go back to reference A. Ben-Shlomo, H. Schmid, K. Wawrowsky, O. Pichurin, E. Hubina, V. Chesnokova, N.A. Liu, M. Culler, S. Melmed, Differential ligand-mediated pituitary somatostatin receptor subtype signaling: implications for corticotroph tumor therapy. J. Clin. Endocrinol. Metab. 94, 4342–4350 (2006) A. Ben-Shlomo, H. Schmid, K. Wawrowsky, O. Pichurin, E. Hubina, V. Chesnokova, N.A. Liu, M. Culler, S. Melmed, Differential ligand-mediated pituitary somatostatin receptor subtype signaling: implications for corticotroph tumor therapy. J. Clin. Endocrinol. Metab. 94, 4342–4350 (2006)
61.
go back to reference S. Lesche, D. Lehmann, F. Nagel, H.A. Schmid, S. Schulz, Differential effects of octreotide and pasireotide on somatostatin receptor internalization and trafficking in vitro. J. Clin. Endocrinol. Metab. 94, 654–661 (2009)PubMed S. Lesche, D. Lehmann, F. Nagel, H.A. Schmid, S. Schulz, Differential effects of octreotide and pasireotide on somatostatin receptor internalization and trafficking in vitro. J. Clin. Endocrinol. Metab. 94, 654–661 (2009)PubMed
62.
go back to reference M.C. Zatelli, F. Tagliati, J.E. Taylor, D. Piccin, M.D. Culler, E.C. degli Uberti, Somatostatin, but not somatostatin receptor subtypes 2 and 5 selective agonists, inhibits calcitonin secretion and gene expression in the human medullary thyroid carcinoma cell line, TT. Horm. Metab. Res. 34, 229–233 (2002)PubMed M.C. Zatelli, F. Tagliati, J.E. Taylor, D. Piccin, M.D. Culler, E.C. degli Uberti, Somatostatin, but not somatostatin receptor subtypes 2 and 5 selective agonists, inhibits calcitonin secretion and gene expression in the human medullary thyroid carcinoma cell line, TT. Horm. Metab. Res. 34, 229–233 (2002)PubMed
63.
go back to reference A. Colao, S. Petersenn, J. Newell-Price, J.W. Findling, F. Gu, M. Maldonado, U. Schoenherr, D. Mills, L.R. Salgado, B.M. Biller, Pasireotide B2305 Study Group, A 12-month phase 3 study of pasireotide in Cushing’s disease. N. Engl. J. Med. 366, 914–924 (2012)PubMed A. Colao, S. Petersenn, J. Newell-Price, J.W. Findling, F. Gu, M. Maldonado, U. Schoenherr, D. Mills, L.R. Salgado, B.M. Biller, Pasireotide B2305 Study Group, A 12-month phase 3 study of pasireotide in Cushing’s disease. N. Engl. J. Med. 366, 914–924 (2012)PubMed
64.
go back to reference R.A. Feelders, C. de Bruin, A.M. Pereira, J.A. Romijn, R.T. Netea-Maier, A.R. Hermus, P.M. Zelissen, F.H. de Jong, A.J. van der Lely, W.W. de Herder, L.J. Hofland, S.W.J. Lamberts, Stepwise medical treatment of Cushing’s disease with pasireotide mono- or combination therapy with cabergoline and low-dose ketoconazole. N. Engl. J. Med. 362, 1846–1848 (2010)PubMed R.A. Feelders, C. de Bruin, A.M. Pereira, J.A. Romijn, R.T. Netea-Maier, A.R. Hermus, P.M. Zelissen, F.H. de Jong, A.J. van der Lely, W.W. de Herder, L.J. Hofland, S.W.J. Lamberts, Stepwise medical treatment of Cushing’s disease with pasireotide mono- or combination therapy with cabergoline and low-dose ketoconazole. N. Engl. J. Med. 362, 1846–1848 (2010)PubMed
65.
go back to reference R. Pivonello, D. Ferone, G. Lombardi, A. Colao, S.W. Lamberts, L.J. Hofland, Novel insights in dopamine receptor physiology. Eur. J. Endocrinol. 156(Suppl 1), S13–S21 (2007)PubMed R. Pivonello, D. Ferone, G. Lombardi, A. Colao, S.W. Lamberts, L.J. Hofland, Novel insights in dopamine receptor physiology. Eur. J. Endocrinol. 156(Suppl 1), S13–S21 (2007)PubMed
66.
go back to reference M. Boschetti, F. Gatto, M. Arvigo, D. Esposito, A. Rebora, M. Talco, M. Albertelli, E. Nazzari, U. Goglia, F. Minuto, D. Ferone, Role of dopamine receptors in normal and tumoral pituitary corticotropic cells and adrenal cells. Neuroendocrinology 92(Suppl 1), 17–22 (2010)PubMed M. Boschetti, F. Gatto, M. Arvigo, D. Esposito, A. Rebora, M. Talco, M. Albertelli, E. Nazzari, U. Goglia, F. Minuto, D. Ferone, Role of dopamine receptors in normal and tumoral pituitary corticotropic cells and adrenal cells. Neuroendocrinology 92(Suppl 1), 17–22 (2010)PubMed
67.
go back to reference A. Saiardi, E. Borrelli, Absence of dopaminergic control on melanotrophs leads to Cushing’s-like syndrome in mice. Mol. Endocrinol. 12, 1133–1139 (1998)PubMed A. Saiardi, E. Borrelli, Absence of dopaminergic control on melanotrophs leads to Cushing’s-like syndrome in mice. Mol. Endocrinol. 12, 1133–1139 (1998)PubMed
68.
go back to reference D. Ferone, R. Pivonello, E. Resmini, M. Boschetti, A. Rebora, M. Albertelli, V. Albanese, A. Colao, M.D. Culler, F. Minuto, Preclinical and clinical experiences with the role of dopamine receptors in the treatment of pituitary adenomas. Eur. J. Endocrinol. 156(Suppl 1), S37–S43 (2007)PubMed D. Ferone, R. Pivonello, E. Resmini, M. Boschetti, A. Rebora, M. Albertelli, V. Albanese, A. Colao, M.D. Culler, F. Minuto, Preclinical and clinical experiences with the role of dopamine receptors in the treatment of pituitary adenomas. Eur. J. Endocrinol. 156(Suppl 1), S37–S43 (2007)PubMed
69.
go back to reference D. Ferone, F. Gatto, M. Arvigo, E. Resmini, M. Boschetti, C. Teti, D. Esposito, F. Minuto, The clinical-molecular interface of somatostatin, dopamine and their receptors in pituitary pathophysiology. J. Mol. Endocrinol. 42, 361–370 (2009)PubMed D. Ferone, F. Gatto, M. Arvigo, E. Resmini, M. Boschetti, C. Teti, D. Esposito, F. Minuto, The clinical-molecular interface of somatostatin, dopamine and their receptors in pituitary pathophysiology. J. Mol. Endocrinol. 42, 361–370 (2009)PubMed
70.
go back to reference W.E. Farrell, A.J. Clark, M.F. Stewart, S.R. Crosby, A. White, Bromocriptine inhibits pro-opiomelanocortin mRNA and ACTH precursor secretion in small cell lung cancer cell lines. J Clin Invest. 90, 705–710 (1992)PubMedCentralPubMed W.E. Farrell, A.J. Clark, M.F. Stewart, S.R. Crosby, A. White, Bromocriptine inhibits pro-opiomelanocortin mRNA and ACTH precursor secretion in small cell lung cancer cell lines. J Clin Invest. 90, 705–710 (1992)PubMedCentralPubMed
71.
go back to reference D. Yin, S. Kondo, J. Takeuchi, T. Morimura, Induction of apoptosis in murine ACTH-secreting pituitary adenoma cells by bromocriptine. FEBS Lett. 339, 73–75 (1994)PubMed D. Yin, S. Kondo, J. Takeuchi, T. Morimura, Induction of apoptosis in murine ACTH-secreting pituitary adenoma cells by bromocriptine. FEBS Lett. 339, 73–75 (1994)PubMed
72.
go back to reference R. Pivonello, D. Ferone, W.W. de Herder, J.M. Kros, M.L. De Caro, M. Arvigo, L. Annunziato, G. Lombardi, A. Colao, L.J. Hofland, S.W. Lamberts, Dopamine receptor expression and function in corticotroph pituitary tumors. J. Clin. Endocrinol. Metab. 89, 2452–2462 (2004)PubMed R. Pivonello, D. Ferone, W.W. de Herder, J.M. Kros, M.L. De Caro, M. Arvigo, L. Annunziato, G. Lombardi, A. Colao, L.J. Hofland, S.W. Lamberts, Dopamine receptor expression and function in corticotroph pituitary tumors. J. Clin. Endocrinol. Metab. 89, 2452–2462 (2004)PubMed
73.
go back to reference M. Rocheville, D.C. Lange, U. Kumar, S.C. Patel, R.C. Patel, Y.C. Patel, Receptors for dopamine and somatostatin: formation of hetero-oligomers with enhanced functional activity. Science 288, 154–157 (2000)PubMed M. Rocheville, D.C. Lange, U. Kumar, S.C. Patel, R.C. Patel, Y.C. Patel, Receptors for dopamine and somatostatin: formation of hetero-oligomers with enhanced functional activity. Science 288, 154–157 (2000)PubMed
74.
go back to reference D. Ferone, A. Saveanu, M.D. Culler, M. Arvigo, A. Rebora, F. Gatto, F. Minuto, P. Jaquet, Novel chimeric somatostatin analogs: facts and perspectives. Eur. J. Endocrinol. 156(Suppl 1), S23–S28 (2007)PubMed D. Ferone, A. Saveanu, M.D. Culler, M. Arvigo, A. Rebora, F. Gatto, F. Minuto, P. Jaquet, Novel chimeric somatostatin analogs: facts and perspectives. Eur. J. Endocrinol. 156(Suppl 1), S23–S28 (2007)PubMed
75.
go back to reference A. Saveanu, G. Gunz, S. Guillen, H. Dufour, M.D. Culler, P. Jaquet, Somatostatin and dopamine-somatostatin multiple ligands directed towards somatostatin and dopamine receptors in pituitary adenomas. Neuroendocrinology 83, 258–263 (2006)PubMed A. Saveanu, G. Gunz, S. Guillen, H. Dufour, M.D. Culler, P. Jaquet, Somatostatin and dopamine-somatostatin multiple ligands directed towards somatostatin and dopamine receptors in pituitary adenomas. Neuroendocrinology 83, 258–263 (2006)PubMed
76.
go back to reference D. Ferone, M. Arvigo, C. Semino, P. Jaquet, A. Saveanu, J.E. Taylor, J.P. Moreau, M.D. Culler, M. Albertelli, F. Minuto, A. Barreca, Somatostatin and dopamine receptor expression in lung carcinoma cells and effects of chimeric somatostatin-dopamine molecules on cell proliferation. Am. J. Physiol. Endocrinol. Metab. 289, E1044–E1050 (2005)PubMed D. Ferone, M. Arvigo, C. Semino, P. Jaquet, A. Saveanu, J.E. Taylor, J.P. Moreau, M.D. Culler, M. Albertelli, F. Minuto, A. Barreca, Somatostatin and dopamine receptor expression in lung carcinoma cells and effects of chimeric somatostatin-dopamine molecules on cell proliferation. Am. J. Physiol. Endocrinol. Metab. 289, E1044–E1050 (2005)PubMed
77.
go back to reference C. De Bruin, R.A. Feelders, S.W. Lamberts, L.J. Hofland, Somatostatin and dopamine receptors as targets for medical treatment of Cushing’s syndrome. Rev. Endocr. Metab. Disord. 10, 91–102 (2009)PubMed C. De Bruin, R.A. Feelders, S.W. Lamberts, L.J. Hofland, Somatostatin and dopamine receptors as targets for medical treatment of Cushing’s syndrome. Rev. Endocr. Metab. Disord. 10, 91–102 (2009)PubMed
78.
go back to reference M. Barbot, N. Albiger, F. Ceccato, M. Zilio, A.C. Frigo, L. Denaro, F. Mantero, C. Scaroni, Combination therapy for Cushing’s disease: effectiveness of two schedules of treatment. Should we start with cabergoline or ketoconazole? Pituitary (2013). doi:10.1007/s11102-013-0475-3 M. Barbot, N. Albiger, F. Ceccato, M. Zilio, A.C. Frigo, L. Denaro, F. Mantero, C. Scaroni, Combination therapy for Cushing’s disease: effectiveness of two schedules of treatment. Should we start with cabergoline or ketoconazole? Pituitary (2013). doi:10.​1007/​s11102-013-0475-3
79.
go back to reference E.S. Newlands, G.R.P. Blackledge, J.A. Slack, G.J.S. Rustin, D.B. Smith, N.S.A. Stuart, C.P. Quarterman, R. Hoffman, M.F.G. Stevens, M.H. Brampton, A.C. Gibson, Phase I trial of temozolomide (CCRG 81045: M&B 39831: NSC 362856). Br. J. Cancer 65, 287–291 (1992)PubMedCentralPubMed E.S. Newlands, G.R.P. Blackledge, J.A. Slack, G.J.S. Rustin, D.B. Smith, N.S.A. Stuart, C.P. Quarterman, R. Hoffman, M.F.G. Stevens, M.H. Brampton, A.C. Gibson, Phase I trial of temozolomide (CCRG 81045: M&B 39831: NSC 362856). Br. J. Cancer 65, 287–291 (1992)PubMedCentralPubMed
80.
go back to reference S. Neidle, D.E. Thurston, Chemical approaches to the discovery and development of cancer therapies. Nat. Rev. Cancer 5, 285–296 (2005)PubMed S. Neidle, D.E. Thurston, Chemical approaches to the discovery and development of cancer therapies. Nat. Rev. Cancer 5, 285–296 (2005)PubMed
81.
go back to reference S. Lim, H. Shahinian, M.M. Maya, W. Yong, A.P. Heaney, Temozolomide: a novel treatment for pituitary carcinoma. Lancet Oncol. 7, 518–520 (2006)PubMed S. Lim, H. Shahinian, M.M. Maya, W. Yong, A.P. Heaney, Temozolomide: a novel treatment for pituitary carcinoma. Lancet Oncol. 7, 518–520 (2006)PubMed
82.
go back to reference F.J. Rodriguez, S.N. Thibodeau, R.B. Jenkins, K.V. Schowalter, B.L. Caron, B.P. O’neill, C.D. James, S. Passe, J. Slezak, C. Giannini, MGMT immunohistochemical expression and promoter methylation in human glioblastoma. Appl. Immunohistochem. Mol. Morphol. 16, 59–65 (2008)PubMed F.J. Rodriguez, S.N. Thibodeau, R.B. Jenkins, K.V. Schowalter, B.L. Caron, B.P. O’neill, C.D. James, S. Passe, J. Slezak, C. Giannini, MGMT immunohistochemical expression and promoter methylation in human glioblastoma. Appl. Immunohistochem. Mol. Morphol. 16, 59–65 (2008)PubMed
83.
go back to reference S. Sharma, F. Salehi, B.W. Scheithauer, F. Rotondo, L.V. Syro, K. Kovacs, Role of MGMT in tumor development, progression, diagnosis, treatment and prognosis. Anticancer Res. 29, 3759–3768 (2009)PubMed S. Sharma, F. Salehi, B.W. Scheithauer, F. Rotondo, L.V. Syro, K. Kovacs, Role of MGMT in tumor development, progression, diagnosis, treatment and prognosis. Anticancer Res. 29, 3759–3768 (2009)PubMed
84.
go back to reference L.V. Syro, L.D. Ortiz, B.W. Scheithauer, R. Lloyd, Q. Lau, R. Gonzalez, H. Uribe, M. Cusimano, K. Kovacs, E. Horvath, Treatment of pituitary neoplasms with temozolomide: a review. Cancer 117, 454–462 (2011)PubMed L.V. Syro, L.D. Ortiz, B.W. Scheithauer, R. Lloyd, Q. Lau, R. Gonzalez, H. Uribe, M. Cusimano, K. Kovacs, E. Horvath, Treatment of pituitary neoplasms with temozolomide: a review. Cancer 117, 454–462 (2011)PubMed
85.
go back to reference A. Takeshita, N. Inoshita, M. Taguchi, C. Okuda, N. Fukuhara, K. Oyama, K. Ohashi, T. Sano, Y. Takeuchi, S. Yamada, High incidence of low O(6)-methylguanine DNA methyltransferase expression in invasive macroadenomas of Cushing’s disease. Eur. J. Endocrinol. 161, 553–559 (2009)PubMed A. Takeshita, N. Inoshita, M. Taguchi, C. Okuda, N. Fukuhara, K. Oyama, K. Ohashi, T. Sano, Y. Takeuchi, S. Yamada, High incidence of low O(6)-methylguanine DNA methyltransferase expression in invasive macroadenomas of Cushing’s disease. Eur. J. Endocrinol. 161, 553–559 (2009)PubMed
86.
go back to reference F. Salehi, B.W. Scheithauer, K. Kovacs, E. Horvath, L.V. Syro, S. Sharma, B. Manoranjan, M. Cusimano, O-6-methylguanine-DNA methyltransferase (MGMT) immunohistochemical expression in pituitary corticotroph adenomas. Neurosurgery 70, 491–496 (2012)PubMed F. Salehi, B.W. Scheithauer, K. Kovacs, E. Horvath, L.V. Syro, S. Sharma, B. Manoranjan, M. Cusimano, O-6-methylguanine-DNA methyltransferase (MGMT) immunohistochemical expression in pituitary corticotroph adenomas. Neurosurgery 70, 491–496 (2012)PubMed
87.
go back to reference G. Raverot, F. Castinetti, E. Jouanneau, I. Morange, D. Figarella-Branger, H. Dufour, J. Trouillas, T. Brue, Pituitary carcinomas and aggressive pituitary tumours: merits and pitfalls of temozolomide treatment. Clin. Endocrinol. 76, 769–775 (2012) G. Raverot, F. Castinetti, E. Jouanneau, I. Morange, D. Figarella-Branger, H. Dufour, J. Trouillas, T. Brue, Pituitary carcinomas and aggressive pituitary tumours: merits and pitfalls of temozolomide treatment. Clin. Endocrinol. 76, 769–775 (2012)
88.
go back to reference K. Kovacs, B.W. Scheithauer, M. Lombardero, R.E. McLendon, L.V. Syro, H. Uribe, L.D. Ortiz, L.C. Penagos, MGMT immunoexpression predicts responsiveness of pituitary tumors to temozolomide therapy. Acta Neuropathol. 115, 261–262 (2008)PubMed K. Kovacs, B.W. Scheithauer, M. Lombardero, R.E. McLendon, L.V. Syro, H. Uribe, L.D. Ortiz, L.C. Penagos, MGMT immunoexpression predicts responsiveness of pituitary tumors to temozolomide therapy. Acta Neuropathol. 115, 261–262 (2008)PubMed
89.
go back to reference S.S. Agarwala, J.M. Kirkwood, Temozolomide, a novel alkylating agent with activity in the central nervous system, may improve the treatment of advanced metastatic melanoma. Oncologist 5, 144–151 (2000)PubMed S.S. Agarwala, J.M. Kirkwood, Temozolomide, a novel alkylating agent with activity in the central nervous system, may improve the treatment of advanced metastatic melanoma. Oncologist 5, 144–151 (2000)PubMed
90.
go back to reference J. Sheehan, J. Rainey, J. Nguyen, R. Grimsdale, S. Han, Temozolomide-induced inhibition of pituitary adenoma cells. J. Neurosurg. 114, 354–358 (2011)PubMed J. Sheehan, J. Rainey, J. Nguyen, R. Grimsdale, S. Han, Temozolomide-induced inhibition of pituitary adenoma cells. J. Neurosurg. 114, 354–358 (2011)PubMed
91.
go back to reference A. Spada, Growth factors and human pituitary adenomas. Eur. J. Endocrinol. 138, 255–257 (1998)PubMed A. Spada, Growth factors and human pituitary adenomas. Eur. J. Endocrinol. 138, 255–257 (1998)PubMed
92.
go back to reference M. Niveiro, F.I. Aranda, G. Peiró, C. Alenda, A. Picó, Immunohistochemical analysis of tumor angiogenic factors in human pituitary adenomas. Hum. Pathol. 36, 1090–1095 (2005)PubMed M. Niveiro, F.I. Aranda, G. Peiró, C. Alenda, A. Picó, Immunohistochemical analysis of tumor angiogenic factors in human pituitary adenomas. Hum. Pathol. 36, 1090–1095 (2005)PubMed
93.
go back to reference C.J. McCabe, K. Boelaert, L.A. Tannahill, A.P. Heaney, A.L. Stratford, J.S. Khaira, S. Hussain, M.C. Sheppard, J.A. Franklyn, N.J. Gittoes, Vascular endothelial growth factor, its receptor KDR/Flk-1, and pituitary tumor transforming gene in pituitary tumors. J. Clin. Endocrinol. Metab. 87, 4238–4244 (2002)PubMed C.J. McCabe, K. Boelaert, L.A. Tannahill, A.P. Heaney, A.L. Stratford, J.S. Khaira, S. Hussain, M.C. Sheppard, J.A. Franklyn, N.J. Gittoes, Vascular endothelial growth factor, its receptor KDR/Flk-1, and pituitary tumor transforming gene in pituitary tumors. J. Clin. Endocrinol. Metab. 87, 4238–4244 (2002)PubMed
94.
go back to reference L.D. Ortiz, L.V. Syro, B.W. Scheithauer, A. Ersen, H. Uribe, C.E. Fadul, F. Rotondo, E. Horvath, K. Kovacs, Anti-VEGF therapy in pituitary carcinoma. Pituitary 15, 445–449 (2012)PubMed L.D. Ortiz, L.V. Syro, B.W. Scheithauer, A. Ersen, H. Uribe, C.E. Fadul, F. Rotondo, E. Horvath, K. Kovacs, Anti-VEGF therapy in pituitary carcinoma. Pituitary 15, 445–449 (2012)PubMed
95.
go back to reference S. Ezzat, L. Zheng, H.S. Smyth, S.L. Asa, The c-erbB-2/neu proto-oncogene in human pituitary tumours. Clin. Endocrinol. (Oxf). 46, 599–606 (1997)PubMed S. Ezzat, L. Zheng, H.S. Smyth, S.L. Asa, The c-erbB-2/neu proto-oncogene in human pituitary tumours. Clin. Endocrinol. (Oxf). 46, 599–606 (1997)PubMed
96.
go back to reference G. Kontogeorgos, L. Stefaneanu, K. Kovacs, Z. Cheng, Localization of epidermal growth factor (EGF) and epidermal growth factor receptor (EGFr) in human pituitary adenomas and nontumorous pituitaries: an immunocytochemical study. Endocr. Pathol. 7, 63–70 (1996)PubMed G. Kontogeorgos, L. Stefaneanu, K. Kovacs, Z. Cheng, Localization of epidermal growth factor (EGF) and epidermal growth factor receptor (EGFr) in human pituitary adenomas and nontumorous pituitaries: an immunocytochemical study. Endocr. Pathol. 7, 63–70 (1996)PubMed
97.
go back to reference O. Cooper, G. Vlotides, H. Fukuoka, M.I. Greene, S. Melmed, Expression and function of ErbB receptors and ligands in the pituitary. Endocr. Relat. Cancer 18, R197–R211 (2011)PubMedCentralPubMed O. Cooper, G. Vlotides, H. Fukuoka, M.I. Greene, S. Melmed, Expression and function of ErbB receptors and ligands in the pituitary. Endocr. Relat. Cancer 18, R197–R211 (2011)PubMedCentralPubMed
98.
go back to reference D. Lubke, W. Saeger, D.K. Ludecke, Proliferation markers and EGF in ACTH-secreting adenomas and carcinomas of the pituitary. Endocr. Pathol. 6, 45–55 (1995)PubMed D. Lubke, W. Saeger, D.K. Ludecke, Proliferation markers and EGF in ACTH-secreting adenomas and carcinomas of the pituitary. Endocr. Pathol. 6, 45–55 (1995)PubMed
99.
go back to reference P.A. van Wijk, J.W. van Neck, A. Rijnberk, R.J. Croughs, J.A. Mol, Proliferation of the murine corticotropic tumour cell line AtT20 is affected by hypophysiotrophic hormones, growth factors and glucocorticoids. Mol. Cell. Endocrinol. 111, 13–19 (1995)PubMed P.A. van Wijk, J.W. van Neck, A. Rijnberk, R.J. Croughs, J.A. Mol, Proliferation of the murine corticotropic tumour cell line AtT20 is affected by hypophysiotrophic hormones, growth factors and glucocorticoids. Mol. Cell. Endocrinol. 111, 13–19 (1995)PubMed
100.
go back to reference H. Fukuoka, O. Cooper, A. Ben-Shlomo, A. Mamelak, S.G. Ren, D. Bruyette, S. Melmed, EGFR as a therapeutic target for human, canine, and mouse ACTH-secreting pituitary adenomas. J. Clin. Invest. 121, 4712–4721 (2011)PubMedCentralPubMed H. Fukuoka, O. Cooper, A. Ben-Shlomo, A. Mamelak, S.G. Ren, D. Bruyette, S. Melmed, EGFR as a therapeutic target for human, canine, and mouse ACTH-secreting pituitary adenomas. J. Clin. Invest. 121, 4712–4721 (2011)PubMedCentralPubMed
101.
go back to reference A. Gorshtein, H. Rubinfeld, E. Kendler, M. Theodoropoulou, V. Cerovac, G.K. Stalla, Z.R. Cohen, M. Hadani, I. Shimon, Mammalian target of rapamycin inhibitors rapamycin and RAD001 (everolimus) induce anti-proliferative effects in GHsecreting pituitary tumor cells in vitro. Endocr. Relat. Cancer 16, 1017–1027 (2009)PubMed A. Gorshtein, H. Rubinfeld, E. Kendler, M. Theodoropoulou, V. Cerovac, G.K. Stalla, Z.R. Cohen, M. Hadani, I. Shimon, Mammalian target of rapamycin inhibitors rapamycin and RAD001 (everolimus) induce anti-proliferative effects in GHsecreting pituitary tumor cells in vitro. Endocr. Relat. Cancer 16, 1017–1027 (2009)PubMed
102.
go back to reference M.C. Zatelli, M. Minoia, C. Filieri, F. Tagliati, M. Buratto, M.R. Ambrosio, M. Lapparelli, M. Scanarini, E.C. degli Uberti, Effect of everolimus on cell viability in nonfunctioning pituitary adenomas. J. Clin. Endocrinol. Metab. 95, 968–976 (2010)PubMed M.C. Zatelli, M. Minoia, C. Filieri, F. Tagliati, M. Buratto, M.R. Ambrosio, M. Lapparelli, M. Scanarini, E.C. degli Uberti, Effect of everolimus on cell viability in nonfunctioning pituitary adenomas. J. Clin. Endocrinol. Metab. 95, 968–976 (2010)PubMed
103.
go back to reference D. Dworakowska, E. Wlodek, C.A. Leontiou, S. Igreja, M. Cakir, M. Teng, N. Prodromou, M.I. Goth, S. Grozinsky-Glasberg, M. Gueorguiev, B. Kola, M. Korbonits, A.B. Grossman, Activation of RAF/MEK/ERK and PI3 K/AKT/mTOR pathways in pituitary adenomas and their effects on downstream effectors. Endocr. Relat. Cancer 16, 1329–1338 (2009)PubMed D. Dworakowska, E. Wlodek, C.A. Leontiou, S. Igreja, M. Cakir, M. Teng, N. Prodromou, M.I. Goth, S. Grozinsky-Glasberg, M. Gueorguiev, B. Kola, M. Korbonits, A.B. Grossman, Activation of RAF/MEK/ERK and PI3 K/AKT/mTOR pathways in pituitary adenomas and their effects on downstream effectors. Endocr. Relat. Cancer 16, 1329–1338 (2009)PubMed
104.
go back to reference E. Jouanneau, A. Wierinckx, F. Ducray, V. Favrel, F. Borson-Chazot, J. Honnorat, J. Trouillas, G. Raverot, New targeted therapies in pituitary carcinoma resistant to temozolomide. Pituitary 15, 37–43 (2012)PubMed E. Jouanneau, A. Wierinckx, F. Ducray, V. Favrel, F. Borson-Chazot, J. Honnorat, J. Trouillas, G. Raverot, New targeted therapies in pituitary carcinoma resistant to temozolomide. Pituitary 15, 37–43 (2012)PubMed
105.
go back to reference V. Cerovac, J. Monteserin-Garcia, H. Rubinfeld, M. Buchfelder, M. Losa, T. Florio, M. Paez-Pereda, G.K. Stalla, M. Theodoropoulou, The somatostatin analogue octreotide confers sensitivity to rapamycin treatment on pituitary tumor cells. Cancer Res. 70, 666–674 (2010)PubMed V. Cerovac, J. Monteserin-Garcia, H. Rubinfeld, M. Buchfelder, M. Losa, T. Florio, M. Paez-Pereda, G.K. Stalla, M. Theodoropoulou, The somatostatin analogue octreotide confers sensitivity to rapamycin treatment on pituitary tumor cells. Cancer Res. 70, 666–674 (2010)PubMed
106.
go back to reference K.E. O’Reilly, F. Rojo, Q.B. She, D. Solit, G.B. Mills, D. Smith, H. Lane, F. Hofmann, D.J. Hicklin, D.L. Ludwig, J. Baselga, N. Rosen, mTOR inhibition induces upstream receptor tyrosine kinase signaling and activates Akt. Cancer Res. 66, 1500–1508 (2006)PubMedCentralPubMed K.E. O’Reilly, F. Rojo, Q.B. She, D. Solit, G.B. Mills, D. Smith, H. Lane, F. Hofmann, D.J. Hicklin, D.L. Ludwig, J. Baselga, N. Rosen, mTOR inhibition induces upstream receptor tyrosine kinase signaling and activates Akt. Cancer Res. 66, 1500–1508 (2006)PubMedCentralPubMed
107.
go back to reference P.M. Stewart, S. Petersenn, Rationale for treatment and therapeutic options in Cushing’s disease. Best Pract. Res. Clin. Endocrinol. Metab. 23(Suppl 1), S15–S22 (2009)PubMed P.M. Stewart, S. Petersenn, Rationale for treatment and therapeutic options in Cushing’s disease. Best Pract. Res. Clin. Endocrinol. Metab. 23(Suppl 1), S15–S22 (2009)PubMed
108.
go back to reference M. Boscaro, N. Sonino, A. Rampazzo, F. Mantero, Response of pituitary-adrenal axis to corticotrophin releasing hormone in patients with Cushing’s disease before and after ketoconazole treatment. Clin. Endocrinol. (Oxf). 27, 461–467 (1987)PubMed M. Boscaro, N. Sonino, A. Rampazzo, F. Mantero, Response of pituitary-adrenal axis to corticotrophin releasing hormone in patients with Cushing’s disease before and after ketoconazole treatment. Clin. Endocrinol. (Oxf). 27, 461–467 (1987)PubMed
109.
go back to reference P. Loli, M.E. Berselli, M. Tagliaferri, Use of ketoconazole in the treatment of Cushing’s syndrome. J. Clin. Endocrinol. Metab. 63, 1365–1371 (1986)PubMed P. Loli, M.E. Berselli, M. Tagliaferri, Use of ketoconazole in the treatment of Cushing’s syndrome. J. Clin. Endocrinol. Metab. 63, 1365–1371 (1986)PubMed
110.
go back to reference N. Sonino, The use of ketoconazole as an inhibitor of steroid production. N. Engl. J. Med. 317, 812–818 (1987)PubMed N. Sonino, The use of ketoconazole as an inhibitor of steroid production. N. Engl. J. Med. 317, 812–818 (1987)PubMed
111.
go back to reference R.A. Feelders, L.J. Hofland, W.W. de Herder, Medical treatment of Cushing’s syndrome: adrenal-blocking drugs and ketaconazole. Neuroendocrinology 92(Suppl 1), 111–115 (2010)PubMed R.A. Feelders, L.J. Hofland, W.W. de Herder, Medical treatment of Cushing’s syndrome: adrenal-blocking drugs and ketaconazole. Neuroendocrinology 92(Suppl 1), 111–115 (2010)PubMed
112.
go back to reference G.K. Stalla, J. Stalla, M. Huber, J.P. Loeffler, V. Hollt, K. von Werder, O.A. Muller, Ketoconazole inhibits corticotropic cell function in vitro. Endocrinology 122, 618–623 (1988)PubMed G.K. Stalla, J. Stalla, M. Huber, J.P. Loeffler, V. Hollt, K. von Werder, O.A. Muller, Ketoconazole inhibits corticotropic cell function in vitro. Endocrinology 122, 618–623 (1988)PubMed
113.
go back to reference A. Angeli, R. Frairia, Ketoconazole therapy in Cushing’s disease. Lancet 1, 821 (1985)PubMed A. Angeli, R. Frairia, Ketoconazole therapy in Cushing’s disease. Lancet 1, 821 (1985)PubMed
114.
go back to reference M. Boscaro, N. Sonino, A. Rampazzo, F. Mantero, Response of pituitary-adrenal axis to corticotrophin releasing hormone in patients with Cushing’s disease before and after ketoconazole treatment. Clin. Endocrinol. (Oxf). 27, 461–467 (1987)PubMed M. Boscaro, N. Sonino, A. Rampazzo, F. Mantero, Response of pituitary-adrenal axis to corticotrophin releasing hormone in patients with Cushing’s disease before and after ketoconazole treatment. Clin. Endocrinol. (Oxf). 27, 461–467 (1987)PubMed
115.
go back to reference P. Loli, M.E. Berselli, M. Tagliaferri, Use of ketoconazole in the treatment of Cushing’s syndrome. J. Clin. Endocrinol. Metab. 63, 1365–1371 (1986)PubMed P. Loli, M.E. Berselli, M. Tagliaferri, Use of ketoconazole in the treatment of Cushing’s syndrome. J. Clin. Endocrinol. Metab. 63, 1365–1371 (1986)PubMed
116.
go back to reference D. Engelhardt, M.M. Weber, Therapy of Cushing’s syndrome with steroid biosynthesis inhibitors. J. Steroid Biochem. Mol. Biol. 49, 261–267 (1994)PubMed D. Engelhardt, M.M. Weber, Therapy of Cushing’s syndrome with steroid biosynthesis inhibitors. J. Steroid Biochem. Mol. Biol. 49, 261–267 (1994)PubMed
117.
go back to reference T.A. Miettinen, Cholesterol metabolism during ketoconazole treatment in man. J. Lipid Res. 29, 43–51 (1988)PubMed T.A. Miettinen, Cholesterol metabolism during ketoconazole treatment in man. J. Lipid Res. 29, 43–51 (1988)PubMed
118.
go back to reference M.I. Dushkin, N.K. Zenkov, E.B. Menshikova, E.N. Pivovarova, G. Lyubimov, N.N. Volsky, Ketoconazole inhibits oxidative modification of low density lipoprotein. Atherosclerosis 114, 9–18 (1995)PubMed M.I. Dushkin, N.K. Zenkov, E.B. Menshikova, E.N. Pivovarova, G. Lyubimov, N.N. Volsky, Ketoconazole inhibits oxidative modification of low density lipoprotein. Atherosclerosis 114, 9–18 (1995)PubMed
119.
go back to reference X. Bertagna, L. Guignat, L. Groussin, J. Bertherat, Cushing’s disease. Best Pract. Res. Clin. Endocrinol. Metab. 23, 607–623 (2009)PubMed X. Bertagna, L. Guignat, L. Groussin, J. Bertherat, Cushing’s disease. Best Pract. Res. Clin. Endocrinol. Metab. 23, 607–623 (2009)PubMed
120.
go back to reference C.N. Dang, O. Trainer, Pharmacological management of Cushing’s syndrome: an update. Arq. Bras. Endocrinol. Metabol. 51, 1339–1348 (2007)PubMed C.N. Dang, O. Trainer, Pharmacological management of Cushing’s syndrome: an update. Arq. Bras. Endocrinol. Metabol. 51, 1339–1348 (2007)PubMed
121.
go back to reference B.A. Gross, S.A. Mindea, A.J. Pick, J.P. Chandler, H.H. Batjer, Medical management of Cushing disease. Neurosurg. Focus 23, E10 (2007)PubMed B.A. Gross, S.A. Mindea, A.J. Pick, J.P. Chandler, H.H. Batjer, Medical management of Cushing disease. Neurosurg. Focus 23, E10 (2007)PubMed
122.
go back to reference J.A. Verhelst, P.J. Trainer, T.A. Howlett, L. Perry, L.H. Rees, A.B. Grossman, J.A. Wass, G.M. Besser, Short and long-term responses to metyrapone in the medical management of 91 patients with Cushing’s syndrome. Clin. Endocrinol. (Oxf). 35, 169–178 (1991)PubMed J.A. Verhelst, P.J. Trainer, T.A. Howlett, L. Perry, L.H. Rees, A.B. Grossman, J.A. Wass, G.M. Besser, Short and long-term responses to metyrapone in the medical management of 91 patients with Cushing’s syndrome. Clin. Endocrinol. (Oxf). 35, 169–178 (1991)PubMed
123.
go back to reference T. Mancini, T. Porcelli, A. Giustina, Treatment of Cushing disease: overview and recent findings. Ther. Clin. Risk Manag. 6, 505–516 (2010)PubMedCentralPubMed T. Mancini, T. Porcelli, A. Giustina, Treatment of Cushing disease: overview and recent findings. Ther. Clin. Risk Manag. 6, 505–516 (2010)PubMedCentralPubMed
124.
go back to reference L.K. Nieman, Medical therapy of Cushing’s disease. Pituitary 5, 77–82 (2002)PubMed L.K. Nieman, Medical therapy of Cushing’s disease. Pituitary 5, 77–82 (2002)PubMed
125.
go back to reference M. Terzolo, A. Angeli, M. Fassnacht, F. Daffara, L. Tauchmanova, P.A. Conton, R. Rossetto, L. Buci, P. Sperone, E. Grossrubatscher, G. Reimondo, E. Bollito, M. Papotti, W. Saeger, S. Hahner, A.C. Koschker, E. Arvat, B. Ambrosi, P. Loli, G. Lombardi, M. Mannelli, P. Bruzzi, F. Mantero, B. Allolio, L. Dogliotti, A. Berruti, Adjuvant mitotane treatment for adrenocortical carcinoma. N. Engl. J. Med. 356, 2372–2380 (2007)PubMed M. Terzolo, A. Angeli, M. Fassnacht, F. Daffara, L. Tauchmanova, P.A. Conton, R. Rossetto, L. Buci, P. Sperone, E. Grossrubatscher, G. Reimondo, E. Bollito, M. Papotti, W. Saeger, S. Hahner, A.C. Koschker, E. Arvat, B. Ambrosi, P. Loli, G. Lombardi, M. Mannelli, P. Bruzzi, F. Mantero, B. Allolio, L. Dogliotti, A. Berruti, Adjuvant mitotane treatment for adrenocortical carcinoma. N. Engl. J. Med. 356, 2372–2380 (2007)PubMed
126.
go back to reference E. Gentilin, F. Tagliati, M. Terzolo, M. Zoli, M. Lapparelli, M. Minoia, M.R. Ambrosio, E.C. degli Uberti, M.C. Zatelli, Mitotane reduces human and mouse ACTH-secreting pituitary cells viability and function. J. Endocrinol. 218, 275–285 (2013)PubMed E. Gentilin, F. Tagliati, M. Terzolo, M. Zoli, M. Lapparelli, M. Minoia, M.R. Ambrosio, E.C. degli Uberti, M.C. Zatelli, Mitotane reduces human and mouse ACTH-secreting pituitary cells viability and function. J. Endocrinol. 218, 275–285 (2013)PubMed
127.
go back to reference M.C. Zatelli, E. Gentilin, F. Daffara, F. Tagliati, G. Reimondo, G. Carandina, M.R. Ambrosio, M. Terzolo, E.C. degli Uberti, Therapeutic concentrations of mitotane (o,p′-DDD) inhibit thyrotroph cell viability and TSH expression and secretion in a mouse cell line model. Endocrinology 151, 2453–2461 (2010)PubMed M.C. Zatelli, E. Gentilin, F. Daffara, F. Tagliati, G. Reimondo, G. Carandina, M.R. Ambrosio, M. Terzolo, E.C. degli Uberti, Therapeutic concentrations of mitotane (o,p′-DDD) inhibit thyrotroph cell viability and TSH expression and secretion in a mouse cell line model. Endocrinology 151, 2453–2461 (2010)PubMed
128.
go back to reference J.P. Luton, J.A. Mahoudeau, P. Bouchard, P. Thieblot, M. Hautecouverture, D. Simon, M.H. Laudat, Y. Touitou, H. Bricaire, Treatment of Cushing’s disease by o,p′DDD. Survey of 62 cases. N. Engl. J. Med. 300, 459–464 (1979)PubMed J.P. Luton, J.A. Mahoudeau, P. Bouchard, P. Thieblot, M. Hautecouverture, D. Simon, M.H. Laudat, Y. Touitou, H. Bricaire, Treatment of Cushing’s disease by o,p′DDD. Survey of 62 cases. N. Engl. J. Med. 300, 459–464 (1979)PubMed
129.
go back to reference D.E. Schteingart, H.S. Tsao, C.I. Taylor, A. McKenzie, R. Victoria, B.A. Therrien, Sustained remission of Cushing’s disease with mitotane and pituitary irradiation. Ann. Intern. Med. 92, 613–619 (1980)PubMed D.E. Schteingart, H.S. Tsao, C.I. Taylor, A. McKenzie, R. Victoria, B.A. Therrien, Sustained remission of Cushing’s disease with mitotane and pituitary irradiation. Ann. Intern. Med. 92, 613–619 (1980)PubMed
130.
go back to reference D. LaSala, Y. Shibanaka, A.Y. Jeng, Coexpression of CYP11B2 or CYP11B1 with adrenodoxin and adrenodoxin reductase for assessing the potency and selectivity of aldosterone synthase inhibitors. Anal. Biochem. 394, 56–61 (2009)PubMed D. LaSala, Y. Shibanaka, A.Y. Jeng, Coexpression of CYP11B2 or CYP11B1 with adrenodoxin and adrenodoxin reductase for assessing the potency and selectivity of aldosterone synthase inhibitors. Anal. Biochem. 394, 56–61 (2009)PubMed
131.
go back to reference L. Amar, M. Azizi, J. Menard, S. Peyrard, C. Watson, P.F. Plouin, Aldosterone synthase inhibition with LCI699: a proof-of-concept study in patients with primary aldosteronism. Hypertension 56, 831–838 (2010)PubMed L. Amar, M. Azizi, J. Menard, S. Peyrard, C. Watson, P.F. Plouin, Aldosterone synthase inhibition with LCI699: a proof-of-concept study in patients with primary aldosteronism. Hypertension 56, 831–838 (2010)PubMed
132.
go back to reference D.A. Calhoun, W.B. White, H. Krum, W. Guo, G. Bermann, A. Trapani, M.P. Lefkowitz, J. Menard, Effects of a novel aldosterone synthase inhibitor for treatment of primary hypertension: results of a randomized, double-blind, placebo- and active-controlled phase 2 trial. Circulation 124, 1945–1955 (2011)PubMed D.A. Calhoun, W.B. White, H. Krum, W. Guo, G. Bermann, A. Trapani, M.P. Lefkowitz, J. Menard, Effects of a novel aldosterone synthase inhibitor for treatment of primary hypertension: results of a randomized, double-blind, placebo- and active-controlled phase 2 trial. Circulation 124, 1945–1955 (2011)PubMed
133.
go back to reference N.A. Tritos, B.M. Biller, Advances in medical therapies for Cushing’s syndrome. Discov. Med. 13, 171–179 (2011) N.A. Tritos, B.M. Biller, Advances in medical therapies for Cushing’s syndrome. Discov. Med. 13, 171–179 (2011)
134.
go back to reference X. Bertagna, R. Pivonello, M. Fleseriu, J. Zhang, P. Robinson, A. Taylor, C. Watson, M. Maldonado, A. Hamraian, M. Boscaro, Patients with Cushing’s disease achieve normal urinary cortisol with LCI699, a potent 11B-hydroxylase inhibitor: preliminary results from a multicenter, proof-of-concept study, in 15th International and 14th European Congress of Endocrinology (ICE/ECE 2012), Florence, Italy (abstract OC1.2) (2012) X. Bertagna, R. Pivonello, M. Fleseriu, J. Zhang, P. Robinson, A. Taylor, C. Watson, M. Maldonado, A. Hamraian, M. Boscaro, Patients with Cushing’s disease achieve normal urinary cortisol with LCI699, a potent 11B-hydroxylase inhibitor: preliminary results from a multicenter, proof-of-concept study, in 15th International and 14th European Congress of Endocrinology (ICE/ECE 2012), Florence, Italy (abstract OC1.2) (2012)
135.
go back to reference P. Dewis, D.C. Anderson, D.E. Bu’lock, R. Earnshaw, W.F. Kelly, Experience with trilostane in the treatment of Cushing’s syndrome. Clin. Endocrinol. (Oxf). 18, 533–540 (1983)PubMed P. Dewis, D.C. Anderson, D.E. Bu’lock, R. Earnshaw, W.F. Kelly, Experience with trilostane in the treatment of Cushing’s syndrome. Clin. Endocrinol. (Oxf). 18, 533–540 (1983)PubMed
136.
go back to reference P. Komanicky, R.F. Spark, J.C. Melby, Treatment of Cushing’s syndrome with trilostane (WIN 24,540), an inhibitor of adrenal steroid biosynthesis. J. Clin. Endocrinol. Metab. 47, 1042–1051 (1978)PubMed P. Komanicky, R.F. Spark, J.C. Melby, Treatment of Cushing’s syndrome with trilostane (WIN 24,540), an inhibitor of adrenal steroid biosynthesis. J. Clin. Endocrinol. Metab. 47, 1042–1051 (1978)PubMed
137.
go back to reference S. Hahner, A. Stürmer, M. Fassnacht, R.W. Hartmann, K. Schewe, S. Cochran, M. Zink, A. Schirbel, B. Allolio, Etomidate unmasks intraadrenal regulation of steroidogenesis and proliferation in adrenal cortical cell lines. Horm. Metab. Res. 42, 528–534 (2010)PubMed S. Hahner, A. Stürmer, M. Fassnacht, R.W. Hartmann, K. Schewe, S. Cochran, M. Zink, A. Schirbel, B. Allolio, Etomidate unmasks intraadrenal regulation of steroidogenesis and proliferation in adrenal cortical cell lines. Horm. Metab. Res. 42, 528–534 (2010)PubMed
138.
go back to reference N. Mettauer, J. Brierley, A novel use of etomidate for intentional adrenal suppression to control severe hypercortisolemia in childhood. Pediatr. Crit. Care Med. 10, e37–e40 (2009)PubMed N. Mettauer, J. Brierley, A novel use of etomidate for intentional adrenal suppression to control severe hypercortisolemia in childhood. Pediatr. Crit. Care Med. 10, e37–e40 (2009)PubMed
139.
go back to reference P. Kamenicky, C. Droumaguet, S. Salenave, A. Blanchard, C. Jublanc, J.F. Gautier, S. Brailly-Tabard, S. Leboulleux, M. Schlumberger, E. Baudin, P. Chanson, J. Young, Mitotane, metyrapone, and ketoconazole combination therapy as an alternative to rescue adrenalectomy for severe ACTH-dependent Cushing’s syndrome. J. Clin. Endocrinol. Metab. 96, 2796–2804 (2011)PubMed P. Kamenicky, C. Droumaguet, S. Salenave, A. Blanchard, C. Jublanc, J.F. Gautier, S. Brailly-Tabard, S. Leboulleux, M. Schlumberger, E. Baudin, P. Chanson, J. Young, Mitotane, metyrapone, and ketoconazole combination therapy as an alternative to rescue adrenalectomy for severe ACTH-dependent Cushing’s syndrome. J. Clin. Endocrinol. Metab. 96, 2796–2804 (2011)PubMed
140.
go back to reference S. Johanssen, B. Allolio, Mifepristone (RU 486) in Cushing’s syndrome. Eur. J. Endocrinol. 157, 561–569 (2007)PubMed S. Johanssen, B. Allolio, Mifepristone (RU 486) in Cushing’s syndrome. Eur. J. Endocrinol. 157, 561–569 (2007)PubMed
141.
go back to reference M.C. Lebeau, E.E. Baulieu, Steroid antagonists and receptor-associated proteins. Hum. Reprod. 9, 437–444 (1994)PubMed M.C. Lebeau, E.E. Baulieu, Steroid antagonists and receptor-associated proteins. Hum. Reprod. 9, 437–444 (1994)PubMed
142.
go back to reference C.M. Bamberger, G.P. Chrousos, The glucocorticoid receptor and RU 486 in man. Ann. N. Y. Acad. Sci. 761, 296–310 (1995)PubMed C.M. Bamberger, G.P. Chrousos, The glucocorticoid receptor and RU 486 in man. Ann. N. Y. Acad. Sci. 761, 296–310 (1995)PubMed
143.
go back to reference M.D. Heitzer, I.M. Wolf, E.R. Sanchez, S.F. Witchel, D.B. DeFranco, Glucocorticoid receptor physiology. Rev. Endocr. Metab. Disord. 8, 321–330 (2007)PubMed M.D. Heitzer, I.M. Wolf, E.R. Sanchez, S.F. Witchel, D.B. DeFranco, Glucocorticoid receptor physiology. Rev. Endocr. Metab. Disord. 8, 321–330 (2007)PubMed
144.
go back to reference H.F. Yang-Yen, J.C. Chambard, Y.L. Sun, T. Smeal, T.J. Schmidt, J. Drouin, M. Karin, Transcriptional interference between c-Jun and the glucocorticoid receptor: mutual inhibition of DNA binding due to direct protein–protein interaction. Cell 62, 1205–1215 (1990)PubMed H.F. Yang-Yen, J.C. Chambard, Y.L. Sun, T. Smeal, T.J. Schmidt, J. Drouin, M. Karin, Transcriptional interference between c-Jun and the glucocorticoid receptor: mutual inhibition of DNA binding due to direct protein–protein interaction. Cell 62, 1205–1215 (1990)PubMed
145.
go back to reference C.M. Jewell, J.C. Webster, K.L. Burnstein, M. Sar, J.E. Bodwell, J.A. Cidlowski, Immunocytochemical analysis of hormone mediated nuclear translocation of wild type and mutant glucocorticoid receptors. J. Steroid Biochem. Mol. Biol. 55, 135–146 (1995)PubMed C.M. Jewell, J.C. Webster, K.L. Burnstein, M. Sar, J.E. Bodwell, J.A. Cidlowski, Immunocytochemical analysis of hormone mediated nuclear translocation of wild type and mutant glucocorticoid receptors. J. Steroid Biochem. Mol. Biol. 55, 135–146 (1995)PubMed
146.
go back to reference B.W. Peeters, G.S. Ruigt, M. Craighead, P. Kitchener, Differential effects of the new glucocorticoid receptor antagonist ORG 34517 and RU486 (mifepristone) on glucocorticoid receptor nuclear translocation in the AtT20 cell line. Ann. N. Y. Acad. Sci. 1148, 536–541 (2008)PubMed B.W. Peeters, G.S. Ruigt, M. Craighead, P. Kitchener, Differential effects of the new glucocorticoid receptor antagonist ORG 34517 and RU486 (mifepristone) on glucocorticoid receptor nuclear translocation in the AtT20 cell line. Ann. N. Y. Acad. Sci. 1148, 536–541 (2008)PubMed
147.
go back to reference F. Spiga, D.M. Knight, S.K. Droste, B. Conway-Campbell, Y. Kershaw, C.P. MacSweeney, F.J. Thomson, M. Craighead, B.W. Peeters, S.L. Lightman, Differential effect of glucocorticoid receptor antagonists on glucocorticoid receptor nuclear translocation and DNA binding. J. Psychopharmacol. 25, 211–221 (2011)PubMed F. Spiga, D.M. Knight, S.K. Droste, B. Conway-Campbell, Y. Kershaw, C.P. MacSweeney, F.J. Thomson, M. Craighead, B.W. Peeters, S.L. Lightman, Differential effect of glucocorticoid receptor antagonists on glucocorticoid receptor nuclear translocation and DNA binding. J. Psychopharmacol. 25, 211–221 (2011)PubMed
148.
go back to reference R. Sitruk-Ware, I.M. Spitz, Pharmacological properties of mifepristone: toxicology and safety in animal and human studies. Contraception 68, 409–420 (2003)PubMed R. Sitruk-Ware, I.M. Spitz, Pharmacological properties of mifepristone: toxicology and safety in animal and human studies. Contraception 68, 409–420 (2003)PubMed
149.
go back to reference X. Bertagna, C. Bertagna, M.H. Laudat, J.M. Husson, F. Girard, J.P. Luton, Pituitary-adrenal response to the antiglucocorticoid action of RU 486 in Cushing’s syndrome. J. Clin. Endocrinol. Metab. 63, 639–643 (1986)PubMed X. Bertagna, C. Bertagna, M.H. Laudat, J.M. Husson, F. Girard, J.P. Luton, Pituitary-adrenal response to the antiglucocorticoid action of RU 486 in Cushing’s syndrome. J. Clin. Endocrinol. Metab. 63, 639–643 (1986)PubMed
150.
go back to reference J.W. Chu, D.F. Matthias, J. Belanoff, A. Schatzberg, A.R. Hoffman, D. Feldman, Successful long-term treatment of refractory Cushing’s disease with high-dose mifepristone (RU 486). J. Clin. Endocrinol. Metab. 86, 3568–3573 (2001)PubMed J.W. Chu, D.F. Matthias, J. Belanoff, A. Schatzberg, A.R. Hoffman, D. Feldman, Successful long-term treatment of refractory Cushing’s disease with high-dose mifepristone (RU 486). J. Clin. Endocrinol. Metab. 86, 3568–3573 (2001)PubMed
151.
go back to reference A.M. Isidori, G.A. Kaltsas, C. Pozza, V. Frajese, J. Newell-Price, R.H. Reznek, P.J. Jenkins, J.P. Monson, A.B. Grossman, G.M. Besser, The ectopic adrenocorticotropin syndrome: clinical features, diagnosis, management, and long-term follow-up. J. Clin. Endocrinol. Metab. 91, 371–377 (2006)PubMed A.M. Isidori, G.A. Kaltsas, C. Pozza, V. Frajese, J. Newell-Price, R.H. Reznek, P.J. Jenkins, J.P. Monson, A.B. Grossman, G.M. Besser, The ectopic adrenocorticotropin syndrome: clinical features, diagnosis, management, and long-term follow-up. J. Clin. Endocrinol. Metab. 91, 371–377 (2006)PubMed
152.
go back to reference F. Castinetti, M. Fassnacht, S. Johanssen, M. Terzolo, P. Bouchard, P. Chanson, C. Do Cao, I. Morange, A. Pico, S. Ouzounian, J. Young, S. Hahner, T. Brue, B. Allolio, B. Conte-Devolx, Merits and pitfalls of mifepristone in Cushing’s syndrome. Eur. J. Endocrinol. 160, 1003–1010 (2009)PubMed F. Castinetti, M. Fassnacht, S. Johanssen, M. Terzolo, P. Bouchard, P. Chanson, C. Do Cao, I. Morange, A. Pico, S. Ouzounian, J. Young, S. Hahner, T. Brue, B. Allolio, B. Conte-Devolx, Merits and pitfalls of mifepristone in Cushing’s syndrome. Eur. J. Endocrinol. 160, 1003–1010 (2009)PubMed
153.
go back to reference F. Castinetti, B. Conte-Devolx, T. Brue, Medical treatment of Cushing’s syndrome: glucocorticoid receptor antagonists and mifepristone. Neuroendocrinology 92(Suppl 1), 125–130 (2010)PubMed F. Castinetti, B. Conte-Devolx, T. Brue, Medical treatment of Cushing’s syndrome: glucocorticoid receptor antagonists and mifepristone. Neuroendocrinology 92(Suppl 1), 125–130 (2010)PubMed
154.
go back to reference M. Fleseriu, B.M. Biller, J.W. Findling, M.E. Molitch, D.E. Schteingart, C. Gross, Mifepristone, a glucocorticoid receptor antagonist, produces clinical and metabolic benefits in patients with Cushing’s syndrome. J. Clin. Endocrinol. Metab. 97, 2039–2049 (2012)PubMed M. Fleseriu, B.M. Biller, J.W. Findling, M.E. Molitch, D.E. Schteingart, C. Gross, Mifepristone, a glucocorticoid receptor antagonist, produces clinical and metabolic benefits in patients with Cushing’s syndrome. J. Clin. Endocrinol. Metab. 97, 2039–2049 (2012)PubMed
155.
go back to reference P.M. Stewart, B.R. Walker, G. Holder, D. O’Halloran, C.H. Shackleton, 11 beta-Hydroxysteroid dehydrogenase activity in Cushing’s syndrome: explaining the mineralocorticoid excess state of the ectopic adrenocorticotropin syndrome. J. Clin. Endocrinol. Metab. 80, 3617–3620 (1995)PubMed P.M. Stewart, B.R. Walker, G. Holder, D. O’Halloran, C.H. Shackleton, 11 beta-Hydroxysteroid dehydrogenase activity in Cushing’s syndrome: explaining the mineralocorticoid excess state of the ectopic adrenocorticotropin syndrome. J. Clin. Endocrinol. Metab. 80, 3617–3620 (1995)PubMed
156.
go back to reference S.H. van Uum, J.W. Lenders, A.R. Hermus, Cortisol, 11beta-hydroxysteroid dehydrogenases, and hypertension. Semin. Vasc. Med. 4, 121–128 (2004)PubMed S.H. van Uum, J.W. Lenders, A.R. Hermus, Cortisol, 11beta-hydroxysteroid dehydrogenases, and hypertension. Semin. Vasc. Med. 4, 121–128 (2004)PubMed
157.
go back to reference J.D. Carmichael, M. Fleseriu, Mifepristone: is there a place in the treatment of Cushing’s disease? Endocrine 44, 20–32 (2013)PubMed J.D. Carmichael, M. Fleseriu, Mifepristone: is there a place in the treatment of Cushing’s disease? Endocrine 44, 20–32 (2013)PubMed
158.
go back to reference G.A. Kaltsas, P. Nomikos, G. Kontogeorgos, M. Buchfelder, A.B. Grossman, Clinical review: diagnosis and management of pituitary carcinomas. J. Clin. Endocrinol. Metab. 90, 3089–3099 (2005)PubMed G.A. Kaltsas, P. Nomikos, G. Kontogeorgos, M. Buchfelder, A.B. Grossman, Clinical review: diagnosis and management of pituitary carcinomas. J. Clin. Endocrinol. Metab. 90, 3089–3099 (2005)PubMed
159.
go back to reference B.T. Ragel, W.T. Couldwell, Pituitary carcinoma: a review of the literature. Neurosurg. Focus 16, 1–9 (2004) B.T. Ragel, W.T. Couldwell, Pituitary carcinoma: a review of the literature. Neurosurg. Focus 16, 1–9 (2004)
160.
go back to reference R.E. Landman, M. Horwith, R.E. Peterson, A.G. Khandji, S.L. Wardlaw, Long-term survival with ACTH-secreting carcinoma of the pituitary: a case report and review of the literature. J. Clin. Endocrinol. Metab. 87, 3084–3089 (2002)PubMed R.E. Landman, M. Horwith, R.E. Peterson, A.G. Khandji, S.L. Wardlaw, Long-term survival with ACTH-secreting carcinoma of the pituitary: a case report and review of the literature. J. Clin. Endocrinol. Metab. 87, 3084–3089 (2002)PubMed
161.
go back to reference G.A. Kaltsas, J.J. Mukherjee, The role of cytotoxic chemotherapy in the management of aggressive and malignant pituitary tumors. J. Clin. Endocrinol. Metab. 83, 4233–4238 (1998)PubMed G.A. Kaltsas, J.J. Mukherjee, The role of cytotoxic chemotherapy in the management of aggressive and malignant pituitary tumors. J. Clin. Endocrinol. Metab. 83, 4233–4238 (1998)PubMed
162.
go back to reference A.A. van der Klaauw, T. Kienitz, C.J. Strasburger, J.W. Smit, J.A. Romijn, Malignant pituitary corticotroph adenomas: report of two cases and a comprehensive review of the literature. Pituitary 12, 57–69 (2009)PubMed A.A. van der Klaauw, T. Kienitz, C.J. Strasburger, J.W. Smit, J.A. Romijn, Malignant pituitary corticotroph adenomas: report of two cases and a comprehensive review of the literature. Pituitary 12, 57–69 (2009)PubMed
163.
go back to reference K. Cukier, R. Tewari, F. Kurth, H.A. Schmid, C. Lai, D.J. Torpy, Significant response to pasireotide (SOM230) in the treatment of a patient with persistent, refractory Cushing’s disease. Clin. Endocrinol. (Oxf). 71, 305–307 (2009)PubMed K. Cukier, R. Tewari, F. Kurth, H.A. Schmid, C. Lai, D.J. Torpy, Significant response to pasireotide (SOM230) in the treatment of a patient with persistent, refractory Cushing’s disease. Clin. Endocrinol. (Oxf). 71, 305–307 (2009)PubMed
164.
go back to reference H. Bode, M. Seiz, A. Lammert, M.A. Brockmann, W. Back, H.P. Hammes, C. Thomé, SOM230 (pasireotide) and temozolomide achieve sustained control of tumour progression and ACTH secretion in pituitary carcinoma with widespread metastases. Exp. Clin. Endocrinol. Diabetes 118, 760–763 (2010)PubMed H. Bode, M. Seiz, A. Lammert, M.A. Brockmann, W. Back, H.P. Hammes, C. Thomé, SOM230 (pasireotide) and temozolomide achieve sustained control of tumour progression and ACTH secretion in pituitary carcinoma with widespread metastases. Exp. Clin. Endocrinol. Diabetes 118, 760–763 (2010)PubMed
165.
go back to reference G. Raverot, N. Sturm, F. de Fraipont, M. Muller, S. Salenave, P. Caron, O. Chabre, P. Chanson, C. Cortet-Rudelli, R. Assaker, H. Dufour, S. Gaillard, P. François, E. Jouanneau, J.G. Passagia, M. Bernier, A. Cornélius, D. Figarella-Branger, J. Trouillas, F. Borson-Chazot, T. Brue, Temozolomide treatment in aggressive pituitary tumors and pituitary carcinomas: a French multicenter experience. J. Clin. Endocrinol. Metab. 95, 4592–4599 (2010)PubMed G. Raverot, N. Sturm, F. de Fraipont, M. Muller, S. Salenave, P. Caron, O. Chabre, P. Chanson, C. Cortet-Rudelli, R. Assaker, H. Dufour, S. Gaillard, P. François, E. Jouanneau, J.G. Passagia, M. Bernier, A. Cornélius, D. Figarella-Branger, J. Trouillas, F. Borson-Chazot, T. Brue, Temozolomide treatment in aggressive pituitary tumors and pituitary carcinomas: a French multicenter experience. J. Clin. Endocrinol. Metab. 95, 4592–4599 (2010)PubMed
166.
go back to reference C.E. Fadul, A.L. Kominsky, L.P. Meyer, L.S. Kingman, W.B. Kinlaw, C.H. Rhodes, C.J. Eskey, N.E. Simmons, Long-term response of pituitary carcinoma to temozolamide. Report of two cases. J. Neurosurg. 105, 621–626 (2006)PubMed C.E. Fadul, A.L. Kominsky, L.P. Meyer, L.S. Kingman, W.B. Kinlaw, C.H. Rhodes, C.J. Eskey, N.E. Simmons, Long-term response of pituitary carcinoma to temozolamide. Report of two cases. J. Neurosurg. 105, 621–626 (2006)PubMed
167.
go back to reference C. Hagen, H.D. Schroder, S. Hansen, C. Hagen, M. Andersen, Temozolamide treatment of a pituitary carcinoma and two pituitary macroadenomas resistant to conventional therapy. Eur. J. Endocrinol. 161, 631–637 (2009)PubMed C. Hagen, H.D. Schroder, S. Hansen, C. Hagen, M. Andersen, Temozolamide treatment of a pituitary carcinoma and two pituitary macroadenomas resistant to conventional therapy. Eur. J. Endocrinol. 161, 631–637 (2009)PubMed
168.
go back to reference A.I. McCormack, K.L. McDonald, A.J. Gill, S.J. Clark, M.G. Burt, K.A. Campbell, W.J. Braund, N.S. Little, R.J. Cook, A.B. Grossman, B.G. Robinson, R.J. Clifton-Bligh, Low O6-methylguanine-DNA methyltransferase (MGMT) expression and response to temozolomide in aggressive pituitary tumours. Clin. Endocrinol. 71, 226–233 (2009) A.I. McCormack, K.L. McDonald, A.J. Gill, S.J. Clark, M.G. Burt, K.A. Campbell, W.J. Braund, N.S. Little, R.J. Cook, A.B. Grossman, B.G. Robinson, R.J. Clifton-Bligh, Low O6-methylguanine-DNA methyltransferase (MGMT) expression and response to temozolomide in aggressive pituitary tumours. Clin. Endocrinol. 71, 226–233 (2009)
169.
go back to reference M. Thearle, P. Freda, J. Bruce, S. Isaacson, Y. Lee, R. Fine, Temozolomide (Temodar®) and capecitabine (Xeloda®) treatment of an aggressive corticotroph pituitary tumor. Pituitary 14, 418–424 (2011)PubMedCentralPubMed M. Thearle, P. Freda, J. Bruce, S. Isaacson, Y. Lee, R. Fine, Temozolomide (Temodar®) and capecitabine (Xeloda®) treatment of an aggressive corticotroph pituitary tumor. Pituitary 14, 418–424 (2011)PubMedCentralPubMed
170.
go back to reference M. Basina, H. Liu, A.R. Hoffman, D. Feldman, Successful long-term treatment of Cushing disease with mifepristone (RU486). Endocr. Pract. 18, e114–e120 (2012)PubMed M. Basina, H. Liu, A.R. Hoffman, D. Feldman, Successful long-term treatment of Cushing disease with mifepristone (RU486). Endocr. Pract. 18, e114–e120 (2012)PubMed
171.
go back to reference L.F. Chan, M. Vaidya, B. Westphal, J. Allgrove, L. Martin, F. Afshar, P.C. Hindmarsh, M.O. Savage, A.B. Grossman, H.L. Storr, Use of intravenous etomidate to control acute psychosis induced by the hypercortisolaemia in severe paediatric Cushing’s disease. Horm. Res. Paediatr. 75, 441–446 (2011)PubMed L.F. Chan, M. Vaidya, B. Westphal, J. Allgrove, L. Martin, F. Afshar, P.C. Hindmarsh, M.O. Savage, A.B. Grossman, H.L. Storr, Use of intravenous etomidate to control acute psychosis induced by the hypercortisolaemia in severe paediatric Cushing’s disease. Horm. Res. Paediatr. 75, 441–446 (2011)PubMed
Metadata
Title
Molecular basis of pharmacological therapy in Cushing’s disease
Authors
Diego Ferone
Claudia Pivonello
Giovanni Vitale
Maria Chiara Zatelli
Annamaria Colao
Rosario Pivonello
Publication date
01-06-2014
Publisher
Springer US
Published in
Endocrine / Issue 2/2014
Print ISSN: 1355-008X
Electronic ISSN: 1559-0100
DOI
https://doi.org/10.1007/s12020-013-0098-5

Other articles of this Issue 2/2014

Endocrine 2/2014 Go to the issue

Endocrine Imaging

Lingual thyroid

Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discusses last year's major advances in heart failure and cardiomyopathies.