Warning: fopen(/home/virtual/enm-kes/journal/upload/ip_log/ip_log_2024-04.txt): failed to open stream: Permission denied in /home/virtual/lib/view_data.php on line 88 Warning: fwrite() expects parameter 1 to be resource, boolean given in /home/virtual/lib/view_data.php on line 89 Recent Progress in the Medical Therapy of Pituitary Tumors
Skip Navigation
Skip to contents

Endocrinol Metab : Endocrinology and Metabolism

clarivate
OPEN ACCESS
SEARCH
Search

Articles

Page Path
HOME > Endocrinol Metab > Volume 32(2); 2017 > Article
Review Article
Recent Progress in the Medical Therapy of Pituitary Tumors
Fabienne Langlois1,2,3,4orcid, Shirley McCartney3,4orcid, Maria Fleseriu2,3,4orcid
Endocrinology and Metabolism 2017;32(2):162-170.
DOI: https://doi.org/10.3803/EnM.2017.32.2.162
Published online: May 19, 2017

1Department of Medicine, Endocrinology and Metabolism, University of Sherbrooke, Sherbrooke, QC, Canada.

2Department of Medicine, Oregon Health & Science University, Portland, OR, USA.

3Department of Neurological Surgery, Oregon Health & Science University, Portland, OR, USA.

4Northwest Pituitary Center, Oregon Health & Science University, Portland, OR, USA.

Corresponding author: Maria Fleseriu. Northwest Pituitary Center, Oregon Health & Science University, 3303 SW Bond Ave, Portland, OR 97239, USA. Tel: +1-503-494-4314, Fax: +1-503-346-6810, fleseriu@ohsu.edu
• Received: April 20, 2017   • Revised: May 9, 2017   • Accepted: May 10, 2017

Copyright © 2017 Korean Endocrine Society

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

  • 5,262 Views
  • 62 Download
  • 27 Web of Science
  • 27 Crossref
  • 24 Scopus
  • Management of pituitary tumors is multidisciplinary, with medical therapy playing an increasingly important role. With the exception of prolactin-secreting tumors, surgery is still considered the first-line treatment for the majority of pituitary adenomas. However, medical/pharmacological therapy plays an important role in controlling hormone-producing pituitary adenomas, especially for patients with acromegaly and Cushing disease (CD). In the case of non-functioning pituitary adenomas (NFAs), pharmacological therapy plays a minor role, the main objective of which is to reduce tumor growth, but this role requires further studies. For pituitary carcinomas and atypical adenomas, medical therapy, including chemotherapy, acts as an adjuvant to surgery and radiation therapy, which is often required to control these aggressive tumors. In the last decade, knowledge about the pathophysiological mechanisms of various pituitary adenomas has increased, thus novel medical therapies that target specific pathways implicated in tumor synthesis and hormonal over secretion are now available. Advancement in patient selection and determination of prognostic factors has also helped to individualize therapy for patients with pituitary tumors. Improvements in biochemical and “tumor mass” disease control can positively affect patient quality of life, comorbidities and overall survival. In this review, the medical armamentarium for treating CD, acromegaly, prolactinomas, NFA, and carcinomas/aggressive atypical adenomas will be presented. Pharmacological therapies, including doses, mode of administration, efficacy, adverse effects, and use in special circumstances are provided. Medical therapies currently under clinical investigation are also briefly discussed.
Pituitary tumors, according to the World Health Organization, are classified as typical or atypical adenomas and carcinomas [1]. They are usually slow growing and induce clinical manifestations either by mass effect on surrounding brain structures, or by causing hormonal imbalances. Mass effect is usually managed surgically. Partial or panhypopituitarism can result from compression of normal pituitary tissue by a growing mass, and is usually managed by hormonal replacement. Pituitary hyperfunction involves a more complex interpretation of patient signs and symptoms and correlation with laboratory investigation. Careful examination of pituitary function should be pursued at diagnosis and serially during follow-up.
Except for prolactin (PRL)-secreting tumors, management of other pituitary tumors includes surgery as a first-line treatment; however, preoperative use of medical therapy in patients with acromegaly and Cushing disease (CD) is on the increase. Currently, in most countries, medical therapy is used as a first-line treatment for prolactinomas, and as a second-line treatment for other secreting pituitary tumors (either persistent or recurrent) or in tumors with an aggressive course where multiple modalities (including surgery and radiation) are required.
Medical therapy to achieve control of hypercortisolism is required if surgery is contraindicated, non-curative, delayed, or if there is no visible pituitary tumor on imaging.
There are three pathophysiological mechanism targets: central inhibition of adrenocorticotropic hormone (ACTH) secretion, adrenal-directed inhibition of steroidogenesis, and glucocorticoid-receptor blockade (Table 1).
Central inhibition of adrenocorticotropic hormone secretion
Anti-secretory and antiproliferative properties of somatostatin receptors ligands (SRLs) act at the CD source; corticotroph adenoma. However, hypercortisolism suppresses somatostatin receptor (SSTR) type 2, and first generation SRLs targeting this receptor (octreotide and lanreotide) are futile as an initial therapy. Their use, alone or in combination, once eucortisolism is achieved may be an option, but more studies are needed [2].
Pasireotide is a SRL with 40-time higher affinity for SSTR type 5, the predominant SSTR expressed in corticotroph tumors. It has been shown to suppress ACTH secretion and cell proliferation. Pasireotide (600 to 900 µg subcutaneous twice daily) induced >50% reduction of urinary free cortisol (UFC) in half the patients studied and approximately 20% achieved normal UFC [34]. A treatment response can be observed within 2 months and to date there is only one case of long-term treatment escape [5]. Tumor volume reduction (TVR) >20% has been observed in patients with baseline visible tumor on magnetic resonance imaging (MRI) [4]. Side effects include gastrointestinal symptoms, cholelithiasis; of note, hyperglycemia occurs in two-thirds of patients with CD [4]. Pasireotide long-acting release (LAR) administered monthly has also been shown in phase III trials to be effective in approximately half of patients with mild CD; overall remission rates were lower, similar with the subcutaneous formulation and with comparable incidence of hyperglycemia [6].
Cabergoline is the most effective dopamine agonist (DA) either as monotherapy or add-on combination therapy. A response rate of approximately 30% to 40% at a high mean dose of 3.5 mg/week (0.5 to 7 mg/week), has been reported [78], but with a lack of long-term response in some patients and concerns about valvulopathy at high doses.
Promising new molecular targets have been identified, including retinoic acid receptors, cyclin-dependent kinases and epidermal growth factor receptor (EGFR) with several drugs in different stages of clinical or preclinical development.
Adrenal-directed inhibition of steroidogenesis
Ketoconazole, an imidazole, has been used off-label to treat Cushing syndrome for 30 years, specifically acting on the cholesterol side-chain cleavage complex (P450scc) and 17- hydroxylase/17,20-lyase activity, reducing glucocorticoid synthesis. Concomitant inhibition of androgen synthesis can result in hypogonadism and gynecomastia in men, but can be beneficial in women with hirsutism. Ketoconazole efficacy varies from 30% to 80%, with the controversial hypothesis of an additional central modulating effect since ACTH levels remain unchanged on therapy [91011]. Approximately 20% of ketoconazole treated patients will eventually escape [9] and some will experience tumor progression. Tolerance may be limited by nausea or vomiting, and hepatotoxicity has to be regularly monitored.
Metyrapone inhibits 11β- and 19-hydroxylase and has potent cortisol-lowering effects; approximately 50% of patients will achieve eucortisolism [12]. An initial compensatory increase in ACTH might attenuate clinical response during the first months of therapy. Side effects include hyperandrogenism in women, and deoxycorticosterone accumulation can result in edema, hypokalemia, and hypertension.
Mitotane is mainly used for its adrenolytic effects in adrenocortical cancer, but at lower doses also has a slow, but potent inhibitory effect on steroid biosynthesis. Approximately 70% of patients will achieve normal UFC and about 10% may have sustained remission after discontinuation [1314]. Adrenal insufficiency (AI) is frequent and higher doses of hydrocortisone are needed for replacement, due to mitotane-induced increase in corticosterone-binding globulin and CYP3A4 inducer effect increasing cortisol metabolism. Side effects include gastrointestinal symptoms, impaired mental functions, gynecomastia, dizziness, and hyperlipidemia; thus, limiting long-term use.
Etomidate, an anesthetic agent, is the only parenteral drug. A low, non-hypnotic perfusion dose, can normalize cortisol levels in patients acutely ill or preoperatively in CD patients with uncontrolled hypercortisolism [14]. Patients have to be monitored for altered consciousness in an intensive care unit.
Medical therapies currently in clinical trial include an oral inhibitor of 18-hydroxylase; osilodrostat (LCI699). In a multicenter 22-week open-label study, normalization of UFC was attained within 10 weeks in 78% of patients. Hypokalemia is reportedly the most frequent side effect [1516] and two phase III studies are ongoing [17]. Levoketoconazole, an enantiomere of ketoconazole, is also under phase III trial and based on preclinical data, may have better efficacy and an advantageous side effect profile compared to ketoconazole [17].
Glucocorticoid-receptor blockade
Mifepristone acts as a glucocorticoid-receptor antagonist, with >10-fold affinity for glucocorticoid-receptor compared to cortisol and >3 times compared to dexamethasone [18]. Mifepristone blocks the progesterone-receptor and is contraindicated in women planning pregnancy. Patients must be monitored closely for hypokalemia, edema, and worsening hypertension. ACTH is expected to increase in two-thirds of patients without tumor growth correlation, but patients with macroadenomas should be monitored for tumor progression [19]. Importantly, laboratory evaluation for cortisol status is not reliable and doses adjustments should be based on clinical status [1820]. If AI occurs, high doses of dexamethasone (2 to 10 mg daily) are often required to overcome receptor blockade [20].
Combination therapy is sometimes needed to achieve eucorti solism in patients with more severe CD or when tolerance to a single agent is limited [2122]. Steroidogenic inhibitors can be combined, the most frequent being ketoconazole and metyrapone, with good results. Cabergoline and ketoconazole was shown to normalize UFC in almost 80% of CD patients in one study, and add-on pasireotide can provide a 10% supplementary effect (Table 1) [1723].
Medical therapy to control excess growth hormone (GH) secretion is usually reserved for patients with uncontrolled disease after an initial surgery or who are not surgical candidates. Therapeutic options include two classes of pharmacologic agents: central-acting inhibitors of GH secretion, including SRLs and DAs, and pegvisomant, a GH-receptor blocking agent (Table 2).
Central inhibition of growth hormone secretion
For decades SRLs have been the first line medical treatment for acromegaly. First generation SRLs, octreotide LAR and lanreotide are considered equivalent medical therapies with minor differences in affinity for SSTR subtypes. Clinical action is mediated mainly via SSTR2A. These SRLs are effective in approximately 50% to 60% of patients in normalizing GH and insulin-like growth factor 1 (IGF-1; lower rates in non-selected or patients without surgery) [24], but also may act specifically on acromegaly-induced headache [25]. Significant TVR of >20% is also observed in ~65% of patients [2627] and shrinkage is more pronounced when SRLs are used as a first-line therapy [26]. Side effects include gastrointestinal symptoms, cholelithiasis/biliary sludge, hyper- or hypoglycemia, and bradycardia.
Preoperative SRL use may be considered in patients with severe pharyngeal thickness and sleep apnea syndrome to reduce perioperative morbidity [25]. Tumor shrinkage, without a clear improvement in surgical cure rate, has been shown, but evidence is lacking as to a precise role and duration of pre-surgical medical treatment [25].
Predictors of good response to first generation SRLs include densely granulated tumors [2829] (typically older patients and MRI T2-hypointensity signal [30]), lower Ki-67 expression and SSTR2A positive immunostaining [31]. Patients with familial acromegaly and aryl hydrocarbon receptor interacting protein (AIP) mutations tend to be resistant to first generation SRLs [32]. Rarely, disease remission has been described with first generation SRL treatment and withdrawal may be attempted in selected patients [3334]. Overall, 10% of patients are considered completely SRL-resistant and 40% to 50% will have an incomplete response, requiring alternative or combination therapies [2931].
Pasireotide, a second generation SRL, has been shown to normalize IGF-1 in 20% of patients resistant to first generation, with similar or slightly greater TVR [35] and may prove more beneficial in patients with sparsely-granulated tumors or with AIP mutations [36]. Worsening hyperglycemia, especially in patients with pre-existing diabetes, is observed in 50% of patients and close observation and treatment is needed [37].
Cabergoline can be effective alone or in combination by inhibiting GH secretion in patients with mild acromegaly (<1.5×upper limit of normal [ULN] elevation in IGF-1). Mixed GH-PRL tumors (PRL positive immunostaining) and hyperprolactinemia do not predict response [3839].
Growth hormone-receptor blockade
Pegvisomant is a GH-receptor antagonist, and blocks peripheral production of liver IGF-1 by competing with endogenous GH. Pegvisomant has a long half-life (60 to 138 hours) and can be used daily or with reduced frequency, up to once weekly. Dose-dependent normalization of IGF-1 levels was achieved in 60% of patients at mean dose of 18 mg/day, and up to 90% of cases at doses of up to 40 mg/day [4041]. Patients with insulin-treated diabetes tend to require higher doses because of a possible up-regulation of the hepatic GH-receptor by hyperinsulinemia [42]. Injection-site rotation is important as patients may rarely develop lipodystrophy (especially women), and liver function has to be regularly monitored. Mild transaminases elevation (2 to 5 times normal) is expected; however, if more severe (>5 times normal) dose reductions or discontinuation is required. By blocking IGF-1 production, GH will increase and thus cannot be monitored for disease-control. Approximately 3% to 5% of patients may experience tumor growth, and serial imaging is important in patients with residual tumors [40].
For patients needing combination therapy, first generation SRL and pegvisomant results in 80% to 97% normal IGF-1 at lower doses of pegvisomant, with better tolerability and at lower cost, but patients require close monitoring, notably for liver enzyme elevations [4344]. Also, the addition of cabergoline to an SRL, can have additional effect in achieving control in patients with mild (<1.5 times ULN) elevation in IGF-1 [43].
Medical therapies currently in clinical trial include a phase III study of an oral form of octreotide where a sustained response was noted in 85% of patients initially controlled on the injectable form, with similar tolerance [45]. Long-acting lanreotide (up to 3 months interval), subcutaneous long-acting octreotide, octreotide implants, somatoprim (SRL with additional SSTR type 4 affinity and less insulin inhibition), and antisense oligonucleotides, which downregulates GH-signaling, are also in different stages of clinical or preclinical development.
Dopamine has an inhibitory effect on lactotroph cells acting through dopamine-receptor subtype 2 (D2). Bromocriptine and cabergoline (Table 3) act more specifically on this receptor subtype, and are therefore the cornerstone of therapy for PRL-secreting tumors. DAs have a dual action; inhibition of PRL secretion per se, and induction of apoptosis in lactotroph cells [46]. They have quick onset of action and usually TVR parallels the decrease in serum PRL, but response can differ depending on prolactinoma subtype. If there is erosion of the sella floor by a large macroprolactinoma, cerebrospinal fluid leak can be a consequence of rapid tumor shrinkage and patient awareness of new, persistent rhinorrhea is advised. Nausea and dizziness are usually transient and improve if medication is taken with food, before bedtime. An increase in compulsive disorders (e.g., hypersexuality and gambling) has been noted in 5% of patients on DAs [47]. Cabergoline is more potent with better tolerance profile than bromocriptine [46]; however, safety data in pregnancy is greater with bromocriptine. DAs are usually discontinued as soon as pregnancy is confirmed, but sometimes have to be reinstituted if there is significant tumor progression during pregnancy. Of note, for smaller prolactinomas without mass effect, replacement with testosterone or estrogen±progesterone can be a reasonable option for hypogonadism [46] with careful biochemical and structural monitoring since estrogen can stimulate tumor growth.
DAs resistance is defined as failure to normalize PRL levels or failure to achieve TVR of >50% at maximal usual doses of medication (bromocriptine 7.5 mg/day or cabergoline 2 mg/week). Treatment resistance can be due to poor compliance, concomitant estrogen or rarely testosterone replacement, or tumor transformation to a more atypical/aggressive type [48]. Structural and/or biochemical resistance can be as high as 25% on bromocriptine, and if so, switching to cabergoline may achieve further response in 70% to 80% of those patients. Increasing the cabergoline dose up to 4 to 6 mg/week in patients with objective response is a good approach; and some groups reported doses as high as 12 mg/week [49]. Higher doses may induce cardiac valvular abnormalities by a serotonin-mediated stimulatory effect on fibroblasts [48]. Recent prospective studies did not show a clear association with smaller doses [50], but should be monitored on cabergoline doses >2 mg/week [46]. Overall, 10% of patients will remain DA resistant and will require surgery, which can also be considered for patients intolerant to medication or with larger tumors pre-pregnancy.
Lapatinib, a tyrosine-kinase inhibitor targeting EGFR and erbB2 tyrosine-kinase, approved in the treatment of breast cancer, has been shown in animal models to inhibit PRL secretion, expression and EGFR/HER2 signaling [51]. Cooper et al. [52] reported a positive response in two patients resistant to high-dose cabergoline; a phase III study of lapatinib and cabergoline in resistant prolactinomas is ongoing.
Most non-functioning pituitary adenomas express D2-receptors and tumor growth can be potentially controlled by cabergoline [53]. Study results are controversial and initial studies included non-progressing tumors; therefore, treatment efficacy is difficult to ascertain [5455]. A recent study by Greenman et al. [56] showed a promising response rate of 58% in enlarging tumors; however, the study was limited by its retrospective design, group imbalances, and inherent selection and observation bias. Thus, with the absence of randomized placebo-controlled trials, use of cabergoline in patients with non-operable tumors or with enlarging residual tumors cannot be universally recommended.
Atypical adenomas are extremely invasive and rapidly progressing pituitary tumors. Characterized by pathologic features such as high Ki-67 (more than 3% to 10%) with extensive positive expression of p53 protein [57], they represent 5% to 10% of pituitary tumors. Temozolomide (TMZ), an oral chemotherapy, used alone [58] or in combination with DAs [59] or SRLs [60] may have an additional effect in tumor control. Data to support TMZ use is scarce; however, it should be considered alongside surgery and radiation therapy in patients with aggressive tumors.
Pituitary carcinomas, defined by presence of widespread metastasis, are exceedingly rare. TMZ response rate in a meta-analysis on pituitary carcinomas was 65% with improved overall 5-year survival of 92% versus 54% (P=0.08) [58]. Response is usually, but not always, inversely proportional to the tumoral expression (immunostaining) of 6-methyguanine-DNA methyltransferase, a DNA repair enzyme that interferes with TMZ action.
Knowledge of tumor pathophysiology and biology is rapidly increasing and pharmacotherapy for pituitary tumors is evolving. Targeting specific receptors and genes implicated in tumors pathogenesis and determining predictors of response using radiological, pathological, and clinical characteristics will lead to more personalized medicine. It is our hope that care of patients with pituitary tumors will continue to advance over time, achieving both biochemical and tumor response, but also significant improvement in quality of life and comorbidities.

CONFLICTS OF INTEREST: Maria Fleseriu has been a principal investigator with research grants to OHSU and scientific consultant to Chiasma, Novartis, Pfizer, Strongbridge. Fabienne Langlois and Shirley McCartney have no conflict of interest.

  • 1. Heaney AP. Clinical review. Pituitary carcinoma: difficult diagnosis and treatment. J Clin Endocrinol Metab 2011;96:3649–3660. ArticlePubMedPMCPDF
  • 2. de Bruin C, Hofland LJ, Nieman LK, van Koetsveld PM, Waaijers AM, Sprij-Mooij DM, et al. Mifepristone effects on tumor somatostatin receptor expression in two patients with Cushing's syndrome due to ectopic adrenocorticotropin secretion. J Clin Endocrinol Metab 2012;97:455–462. ArticlePubMed
  • 3. Pivonello R, Petersenn S, Newell-Price J, Findling JW, Gu F, Maldonado M, et al. Pasireotide treatment significantly improves clinical signs and symptoms in patients with Cushing's disease: results from a phase III study. Clin Endocrinol (Oxf) 2014;81:408–417. ArticlePubMed
  • 4. Colao A, Petersenn S, Newell-Price J, Findling JW, Gu F, Maldonado M, et al. A 12-month phase 3 study of pasireotide in Cushing's disease. N Engl J Med 2012;366:914–924. ArticlePubMed
  • 5. Gabalec F, Husek P, Pacovsky J, Cap J. Escape from response to long-term pasireotide treatment in recurrent Cushing's disease. Endocr Abstr 2015;37:EP716.Article
  • 6. Lacroix A, Petersenn S, Biller BM, Arnaldez F, Roughton M, Ravichandran S, et al. SAT-546: Monthly pasireotide LAR improves urinary free cortisol (UFC) in patients with Cushing's disease: results from a randomized, double-blind, multicenter, phase III study In: Poster presented at: Endocrine Society's 98th Annual Meeting and Expo. 2016 Apr 1-4; Boston, MA.
  • 7. Burman P, Eden-Engstrom B, Ekman B, Karlsson FA, Schwarcz E, Wahlberg J. Limited value of cabergoline in Cushing's disease: a prospective study of a 6-week treatment in 20 patients. Eur J Endocrinol 2016;174:17–24. ArticlePubMed
  • 8. Godbout A, Manavela M, Danilowicz K, Beauregard H, Bruno OD, Lacroix A. Cabergoline monotherapy in the long-term treatment of Cushing's disease. Eur J Endocrinol 2010;163:709–716. ArticlePubMed
  • 9. Castinetti F, Guignat L, Giraud P, Muller M, Kamenicky P, Drui D, et al. Ketoconazole in Cushing's disease: is it worth a try? J Clin Endocrinol Metab 2014;99:1623–1630. ArticlePubMedPDF
  • 10. Cuevas-Ramos D, Lim DS, Fleseriu M. Update on medical treatment for Cushing's disease. Clin Diabetes Endocrinol 2016;2:16.ArticlePubMedPMC
  • 11. Pivonello R, De Leo M, Cozzolino A, Colao A. The treatment of Cushing's disease. Endocr Rev 2015;36:385–486. ArticlePubMedPMCPDF
  • 12. Daniel E, Aylwin S, Mustafa O, Ball S, Munir A, Boelaert K, et al. Effectiveness of metyrapone in treating Cushing's syndrome: a retrospective multicenter study in 195 patients. J Clin Endocrinol Metab 2015;100:4146–4154. ArticlePubMedPMC
  • 13. Baudry C, Coste J, Bou Khalil R, Silvera S, Guignat L, Guibourdenche J, et al. Efficiency and tolerance of mitotane in Cushing's disease in 76 patients from a single center. Eur J Endocrinol 2012;167:473–481. ArticlePubMed
  • 14. Molitch ME. Current approaches to the pharmacological management of Cushing's disease. Mol Cell Endocrinol 2015;408:185–189. ArticlePubMed
  • 15. Fleseriu M. Medical treatment of Cushing disease: new targets, new hope. Endocrinol Metab Clin North Am 2015;44:51–70. ArticlePubMed
  • 16. Fleseriu M, Pivonello R, Young J, Hamrahian AH, Molitch ME, Shimizu C, et al. Osilodrostat, a potent oral 11beta-hydroxylase inhibitor: 22-week, prospective, phase II study in Cushing's disease. Pituitary 2016;19:138–148. ArticlePubMedPDF
  • 17. Fleseriu M, Castinetti F. Updates on the role of adrenal steroidogenesis inhibitors in Cushing's syndrome: a focus on novel therapies. Pituitary 2016;19:643–653. ArticlePubMedPMCPDF
  • 18. Fleseriu M, Biller BM, Findling JW, Molitch ME, Schteingart DE, Gross C, et al. Mifepristone, a glucocorticoid receptor antagonist, produces clinical and metabolic benefits in patients with Cushing's syndrome. J Clin Endocrinol Metab 2012;97:2039–2049. ArticlePubMedPDF
  • 19. Fleseriu M, Findling JW, Koch CA, Schlaffer SM, Buchfelder M, Gross C. Changes in plasma ACTH levels and corticotroph tumor size in patients with Cushing's disease during long-term treatment with the glucocorticoid receptor antagonist mifepristone. J Clin Endocrinol Metab 2014;99:3718–3727. ArticlePubMedPMC
  • 20. Fleseriu M, Molitch ME, Gross C, Schteingart DE, Vaughan TB 3rd, Biller BM. A new therapeutic approach in the medical treatment of Cushing's syndrome: glucocorticoid receptor blockade with mifepristone. Endocr Pract 2013;19:313–326. ArticlePubMedPDF
  • 21. Nieman LK, Biller BM, Findling JW, Murad MH, Newell-Price J, Savage MO, et al. Treatment of Cushing's syndrome: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab 2015;100:2807–2831. ArticlePubMedPMC
  • 22. Cuevas-Ramos D, Fleseriu M. Treatment of Cushing’s disease: a mechanistic update. J Endocrinol 2014;223:R19–R39. ArticlePubMed
  • 23. Feelders RA, de Bruin C, Pereira AM, Romijn JA, Netea-Maier RT, Hermus AR, et al. Pasireotide alone or with cabergoline and ketoconazole in Cushing's disease. N Engl J Med 2010;362:1846–1848. ArticlePubMed
  • 24. Carmichael JD, Bonert VS, Nuno M, Ly D, Melmed S. Acromegaly clinical trial methodology impact on reported biochemical efficacy rates of somatostatin receptor ligand treatments: a meta-analysis. J Clin Endocrinol Metab 2014;99:1825–1833. ArticlePubMedPMCPDF
  • 25. Katznelson L, Laws ER Jr, Melmed S, Molitch ME, Murad MH, Utz A, et al. Acromegaly: an endocrine society clinical practice guideline. J Clin Endocrinol Metab 2014;99:3933–3951. ArticlePubMed
  • 26. Caron PJ, Bevan JS, Petersenn S, Flanagan D, Tabarin A, Prevost G, et al. Tumor shrinkage with lanreotide Autogel 120 mg as primary therapy in acromegaly: results of a prospective multicenter clinical trial. J Clin Endocrinol Metab 2014;99:1282–1290. ArticlePubMedPDF
  • 27. Giustina A, Mazziotti G, Torri V, Spinello M, Floriani I, Melmed S. Meta-analysis on the effects of octreotide on tumor mass in acromegaly. PLoS One 2012;7:e36411ArticlePubMedPMC
  • 28. Kiseljak-Vassiliades K, Carlson NE, Borges MT, Kleinschmidt-DeMasters BK, Lillehei KO, Kerr JM, et al. Growth hormone tumor histological subtypes predict response to surgical and medical therapy. Endocrine 2015;49:231–241. ArticlePubMedPDF
  • 29. Cuevas-Ramos D, Fleseriu M. Somatostatin receptor ligands and resistance to treatment in pituitary adenomas. J Mol Endocrinol 2014;52:R223–R240. ArticlePubMed
  • 30. Potorac I, Petrossians P, Daly AF, Alexopoulou O, Borot S, Sahnoun-Fathallah M, et al. T2-weighted MRI signal predicts hormone and tumor responses to somatostatin analogs in acromegaly. Endocr Relat Cancer 2016;23:871–881. ArticlePubMed
  • 31. Colao A, Auriemma RS, Lombardi G, Pivonello R. Resistance to somatostatin analogs in acromegaly. Endocr Rev 2011;32:247–271. ArticlePubMedPDF
  • 32. Fleseriu M. Advances in the pharmacotherapy of patients with acromegaly. Discov Med 2014;17:329–338. PubMed
  • 33. Vilar L, Fleseriu M, Naves LA, Albuquerque JL, Gadelha PS, dos Santos Faria M, et al. Can we predict long-term remission after somatostatin analog withdrawal in patients with acromegaly? Results from a multicenter prospective trial. Endocrine 2014;46:577–584. ArticlePubMedPDF
  • 34. Casagrande A, Bronstein MD, Jallad RS, Moraes AB, Elias PC, Castro M, et al. Long-term remission of acromegaly after octreotide withdrawal is an uncommon and frequently unsustainable event. Neuroendocrinology 2017;104:273–279. ArticlePubMed
  • 35. Gadelha MR, Bronstein MD, Brue T, Coculescu M, Fleseriu M, Guitelman M, et al. Pasireotide versus continued treatment with octreotide or lanreotide in patients with inadequately controlled acromegaly (PAOLA): a randomised, phase 3 trial. Lancet Diabetes Endocrinol 2014;2:875–884. ArticlePubMed
  • 36. Iacovazzo D, Carlsen E, Lugli F, Chiloiro S, Piacentini S, Bianchi A, et al. Factors predicting pasireotide responsiveness in somatotroph pituitary adenomas resistant to first-generation somatostatin analogues: an immunohistochemical study. Eur J Endocrinol 2016;174:241–250. ArticlePubMed
  • 37. Schmid HA, Brue T, Colao A, Gadelha MR, Shimon I, Kapur K, et al. Effect of pasireotide on glucose- and growth hormone-related biomarkers in patients with inadequately controlled acromegaly. Endocrine 2016;53:210–219. ArticlePubMedPMCPDF
  • 38. Sandret L, Maison P, Chanson P. Place of cabergoline in acromegaly: a meta-analysis. J Clin Endocrinol Metab 2011;96:1327–1335. ArticlePubMed
  • 39. Kuhn E, Chanson P. Cabergoline in acromegaly. Pituitary 2017;20:121–128. ArticlePubMedPDF
  • 40. van der Lely AJ, Biller BM, Brue T, Buchfelder M, Ghigo E, Gomez R, et al. Long-term safety of pegvisomant in patients with acromegaly: comprehensive review of 1288 subjects in ACROSTUDY. J Clin Endocrinol Metab 2012;97:1589–1597. ArticlePubMedPDF
  • 41. Trainer PJ, Drake WM, Katznelson L, Freda PU, Herman-Bonert V, van der Lely AJ, et al. Treatment of acromegaly with the growth hormone-receptor antagonist pegvisomant. N Engl J Med 2000;342:1171–1177. ArticlePubMed
  • 42. Droste M, Domberg J, Buchfelder M, Mann K, Schwanke A, Stalla G, et al. Therapy of acromegalic patients exacerbated by concomitant type 2 diabetes requires higher pegvisomant doses to normalise IGF1 levels. Eur J Endocrinol 2014;171:59–68. ArticlePubMed
  • 43. Lim DS, Fleseriu M. The role of combination medical therapy in the treatment of acromegaly. Pituitary 2017;20:136–148. ArticlePubMedPDF
  • 44. Franck SE, Muhammad A, van der Lely AJ, Neggers SJ. Combined treatment of somatostatin analogues with pegvisomant in acromegaly. Endocrine 2016;52:206–213. ArticlePubMedPDF
  • 45. Melmed S, Popovic V, Bidlingmaier M, Mercado M, van der Lely AJ, Biermasz N, et al. Safety and efficacy of oral octreotide in acromegaly: results of a multicenter phase III trial. J Clin Endocrinol Metab 2015;100:1699–1708. ArticlePubMed
  • 46. Melmed S, Casanueva FF, Hoffman AR, Kleinberg DL, Montori VM, Schlechte JA, et al. Diagnosis and treatment of hyperprolactinemia: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab 2011;96:273–288. ArticlePubMedPDF
  • 47. Noronha S, Stokes V, Karavitaki N, Grossman A. Treating prolactinomas with dopamine agonists: always worth the gamble? Endocrine 2016;51:205–210. ArticlePubMedPDF
  • 48. Molitch ME. Management of medically refractory prolactinoma. J Neurooncol 2014;117:421–428. ArticlePubMedPDF
  • 49. Ono M, Miki N, Kawamata T, Makino R, Amano K, Seki T, et al. Prospective study of high-dose cabergoline treatment of prolactinomas in 150 patients. J Clin Endocrinol Metab 2008;93:4721–4727. ArticlePubMedPDF
  • 50. Drake WM, Stiles CE, Bevan JS, Karavitaki N, Trainer PJ, Rees DA, et al. A follow-up study of the prevalence of valvular heart abnormalities in hyperprolactinemic patients treated with cabergoline. J Clin Endocrinol Metab 2016;101:4189–4194. ArticlePubMed
  • 51. Liu X, Kano M, Araki T, Cooper O, Fukuoka H, Tone Y, et al. ErbB receptor-driven prolactinomas respond to targeted lapatinib treatment in female transgenic mice. Endocrinology 2015;156:71–79. ArticlePubMed
  • 52. Cooper O, Mamelak A, Bannykh S, Carmichael J, Bonert V, Lim S, et al. Prolactinoma ErbB receptor expression and targeted therapy for aggressive tumors. Endocrine 2014;46:318–327. ArticlePubMedPDF
  • 53. Gagliano T, Filieri C, Minoia M, Buratto M, Tagliati F, Ambrosio MR, et al. Cabergoline reduces cell viability in non functioning pituitary adenomas by inhibiting vascular endothelial growth factor secretion. Pituitary 2013;16:91–100. ArticlePubMedPDF
  • 54. Colao A, Di Somma C, Pivonello R, Faggiano A, Lombardi G, Savastano S. Medical therapy for clinically non-functioning pituitary adenomas. Endocr Relat Cancer 2008;15:905–915. ArticlePubMed
  • 55. Garcia EC, Naves LA, Silva AO, de Castro LF, Casulari LA, Azevedo MF. Short-term treatment with cabergoline can lead to tumor shrinkage in patients with nonfunctioning pituitary adenomas. Pituitary 2013;16:189–194. ArticlePubMedPDF
  • 56. Greenman Y, Cooper O, Yaish I, Robenshtok E, Sagiv N, Jonas-Kimchi T, et al. Treatment of clinically nonfunctioning pituitary adenomas with dopamine agonists. Eur J Endocrinol 2016;175:63–72. ArticlePubMed
  • 57. Sav A, Rotondo F, Syro LV, Di Ieva A, Cusimano MD, Kovacs K. Invasive, atypical and aggressive pituitary adenomas and carcinomas. Endocrinol Metab Clin North Am 2015;44:99–104. ArticlePubMed
  • 58. Ji Y, Vogel RI, Lou E. Temozolomide treatment of pituitary carcinomas and atypical adenomas: systematic review of case reports. Neurooncol Pract 2016;3:188–195. ArticlePubMedPDF
  • 59. 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 (Oxf) 2012;76:877–886. ArticlePubMed
  • 60. Ceccato F, Lombardi G, Manara R, Emanuelli E, Denaro L, Milanese L, et al. Temozolomide and pasireotide treatment for aggressive pituitary adenoma: expertise at a tertiary care center. J Neurooncol 2015;122:189–196. ArticlePubMedPDF
Table 1

Medical Therapy of Cushing Disease

enm-32-162-i001.jpg
Variable Route Usual dose Mode of administration Side effect Efficacy (approx) Pregnancy categorya Additional information
Adrenal steroidogenesis inhibitors
 Ketoconazole Oral 400–1,200 mg/day 2–4 times/day Transaminases elevation, N/D, rash, dizziness, AI, gynecomastia (men) 50% C Absorption needs acid gastric pH
 Levoketoconazole Oral 400 mg/day 2 times/day Headaches, back pain, nausea TBD C Phase III study ongoing
 Metyrapone Oral 0.5–6.0 g/day 4 times/day Hirsutism, acne, HTN, hypokaliemia, edema, N, AI 50%–80% C Limited availability in most countries
 Etomidate IV 0.03 mg/kg bolus followed by 0.02–0.08 mg/kg/hr Sedation, myoclonus, N/V, AI 100% C Monitor patient in ICU
 Mitotane Oral 2–4 g/day 3–4 times/day Lethargy, dizziness, weakness, N/V/D, anorexia, AI 70%–90% D Mostly used in adrenal carcinoma
 Osilodrostat Oral 4–60 mg/day 2 times/day N/D, asthenia, AI, hirsutism, acne, headache, hypokaliemia TBD NA Phase III studies ongoing
Centrally-acting agent
 Pasireotide SC 300–1,800 µg/day 2 times/day N/V/D, constipation, abdominal pain, cholelithiasis/biliary sludge, bloating, bradycardia, hyperglycemia (~60%) 20%–30% C More efficacious in mild CD or in combination
 Pasireotide LAR IM 40–60 mg Monthly TBD C Phase III study ongoing
 Cabergoline Oral 1–4 mg Bi-weekly up to daily Nausea, dizziness, orthostatic hypotension 30%–50% B Risk of tachyphylaxis or treatment escape
Glucocorticoid-receptor blocker
 Mifepristone Oral 300–1,200 mg/day Daily Hypokaliemia, edema, HTN, vaginal bleeding, N/V, fatigue, dizziness, headaches 60%–87% X Approved in United States for CS with glucose intolerance or diabetes

N, nausea; D, diarrhea; AI, adrenal insufficiency; TBD, to be determined; HTN, hypertension; IV, intravenous; V, vomiting; ICU, intensive care unit; NA, not available; SC, subcutaneous; CD, Cushing disease; LAR, long-acting release; IM, intramuscular; CS, Cushing syndrome.

aPregnancy categories: A (Generally acceptable. Controlled studies in pregnant women show no evidence of fetal risk), B (May be acceptable. Animal studies showed minor risks and human studies showed no risk), C (Use with caution if benefits outweigh risks. Either no studies available or only animal studies show risk and human studies not available), D (Positive evidence of human fetal risk. Use in last resort, as salvage therapy for emergencies when no safer drug available), X (Do not use in pregnancy. Risks outweigh benefits, use alternatives).

Table 2

Medical Therapy of Acromegaly

enm-32-162-i002.jpg
Variable Route Usual dose Dosage Side effect Efficacy (approx) Pregnancy categorya Additional information
Centrally acting agents
 Octreotide SC 50–400 µg/day 1–4 times/day N/V/D, constipation, abdominal pain, cholelithiasis/biliary sludge, bloating, bradycardia, fatigue, headache, alopecia, dysglycemia 50%–60% C May be used specifically to treat headaches
 Octreotide LAR IM 20–40 mg Monthly C Administered by a health care professional
 Lanreotide Deep SC 60–120 mg Monthly (4–6 weeks) C Pre-filled syringe, may be self-administered
 Pasireotide LAR IM 40–60 mg Monthly Same as above, with more hyperglycemia Up to 80% C Responders identified within 3 months on therapy
 Cabergoline Oral 1–4 mg Bi-weekly up to daily Nausea, dizziness, orthostatic hypotension 30%–40% B Used at higher doses compared to prolactinomas
 Oral octreotide Oral 40–80 mg 2 times/day N/V/D, dyspepsia, cholelithiasis, headaches, dizziness, dysglycemia 65% NA Studies ongoing
GH receptor blocker
 Pegvisomant SC 10–40 mg Daily to once weekly (less frequent dosage in combination) Transaminases elevation, lipodystrophy, arthralgias 60%–90% C Improves insulin resistance, patients on hypoglycemic drugs may require monitoring

SC, subcutaneous; N, nausea; V, vomiting; D, diarrhea; LAR, long-acting release; IM, intramuscular; NA, not available; GH, growth hormone.

aPregnancy categories: A (Generally acceptable. Controlled studies in pregnant women show no evidence of fetal risk), B (May be acceptable. Animal studies showed minor risks and human studies showed no risk), C (Use with caution if benefits outweigh risks. Either no studies available or only animal studies show risk and human studies not available), D (Positive evidence of human fetal risk. Use in last resort, as salvage therapy for emergencies when no safer drug available), X (Do not use in pregnancy. Risks outweigh benefits, use alternatives).

Table 3

Medical Therapy of Prolactinomas

enm-32-162-i003.jpg
Variable Route Usual dose Dosage Side effect Efficacy (approx) Pregnancy categorya Additional information
Bromocriptine Oral 2.5–7.5 mg/day 1–2 times/day N/V, dizziness, orthostatic hypotension, nasal congestion, headache, compulsive behavior 60%–80% B First choice in women planning pregnancy based on available data
Cabergoline Oral 0.5–2 mg Bi-weekly 80%–90% B More potent and generally better tolerated
Lapatinib Oral 1,250 mg Daily Fatigue, GI disturbances, acroparesthesias, insomnia TBD NA Also in study for patients with CD

N, nausea; V, vomiting; GI, gastrointestinal; TBD, to be determined; NA, not available; CD, Cushing disease.

aPregnancy categories: A (Generally acceptable. Controlled studies in pregnant women show no evidence of fetal risk), B (May be acceptable. Animal studies showed minor risks and human studies showed no risk), C (Use with caution if benefits outweigh risks. Either no studies available or only animal studies show risk and human studies not available), D (Positive evidence of human fetal risk. Use in last resort, as salvage therapy for emergencies when no safer drug available), X (Do not use in pregnancy. Risks outweigh benefits, use alternatives).

Figure & Data

References

    Citations

    Citations to this article as recorded by  
    • Multiomics Approach to Acromegaly: Unveiling Translational Insights for Precision Medicine
      Kyungwon Kim, Cheol Ryong Ku, Eun Jig Lee
      Endocrinology and Metabolism.2023; 38(5): 463.     CrossRef
    • PDCD10 promotes the aggressive behaviors of pituitary adenomas by up-regulating CXCR2 and activating downstream AKT/ERK signaling
      Jingdian Liu, Junwen Wang, Weidong Tian, Yu Xu, Ran Li, Kai Zhao, Chao You, Yuan Zhu, Joerg Walter Bartsch, Hongquan Niu, Huaqiu Zhang, Kai Shu, Ting Lei
      Aging.2022; 14(15): 6066.     CrossRef
    • LncRNA MYMLR promotes pituitary adenoma development by upregulating carbonyl reductase 1 via sponging miR-197-3p
      Tuo Wang, Ping Mao, Yan Zhang, Bo Cui, Mao-De Wang, Ya Li, Ke Gao
      Anti-Cancer Drugs.2022; 33(10): 1058.     CrossRef
    • Comparative Proteomic Study Shows the Expression of Hint-1 in Pituitary Adenomas
      Carolina Carrillo-Najar, Daniel Rembao-Bojórquez, Martha L. Tena-Suck, Sergio Zavala-Vega, Noemí Gelista-Herrera, Miguel A. Ramos-Peek, Juan L. Gómez-Amador, Febe Cazares-Raga, Fidel de la Cruz Hernández-Hernández, Alma Ortiz-Plata
      Diagnostics.2021; 11(2): 330.     CrossRef
    • Recent Understanding and Future Directions of Recurrent Corticotroph Tumors
      José Miguel Hinojosa-Amaya, César Ernesto Lam-Chung, Daniel Cuevas-Ramos
      Frontiers in Endocrinology.2021;[Epub]     CrossRef
    • What to Do with Incidentally Discovered Pituitary Abnormalities?
      Fabienne Langlois, Maria Fleseriu
      Medical Clinics of North America.2021; 105(6): 1081.     CrossRef
    • Aggressive prolactinoma (Review)
      Ana Valea, Florica Sandru, Aida Petca, Mihai Dumitrascu, Mara Carsote, Razvan-Cosmin Petca, Adina Ghemigian
      Experimental and Therapeutic Medicine.2021;[Epub]     CrossRef
    • Diagnosis and Treatment of Pituitary Adenomas
      O. A. Beylerli, Zhao Shiguang, I. F. Gareev, Chen Xin
      Creative surgery and oncology.2020; 9(4): 311.     CrossRef
    • Management of patients with persistent or recurrent Cushing’s disease after initial pituitary surgery
      Cristina Capatina, José Miguel Hinojosa-Amaya, Catalina Poiana, Maria Fleseriu
      Expert Review of Endocrinology & Metabolism.2020; 15(5): 321.     CrossRef
    • Modern principles of the diagnosis and conservative treatment of Cushing disease
      Ashraf Abdali, Ludmila Astafeva, Pavel Kalinin, Yuriy Trunin, I. V. Chernov, G. E. Сhmutin, Abdali Badshahzar, GulSharif Sherzad, Simfukwe Keith
      Vestnik nevrologii, psihiatrii i nejrohirurgii (Bulletin of Neurology, Psychiatry and Neurosurgery).2020; (10): 67.     CrossRef
    • Effects of Oxytocin on Cell Proliferation in a Corticotroph Adenoma Cell Line
      Jung Soo Lim, Young Woo Eom, Eun Soo Lee, Hyeong Ju Kwon, Ja-Young Kwon, Junjeong Choi, Choon Hee Chung, Young Suk Jo, Eun Jig Lee
      Endocrinology and Metabolism.2019; 34(3): 302.     CrossRef
    • Pituitary Adenomas: What Are the Key Features? What Are the Current Treatments? Where Is the Future Taking Us?
      Domenico Solari, Rosario Pivonello, Chiara Caggiano, Elia Guadagno, Carmela Chiaramonte, Giovanni Miccoli, Luigi M. Cavallo, Marialaura Del Basso De Caro, Annamaria Colao, Paolo Cappabianca
      World Neurosurgery.2019; 127: 695.     CrossRef
    • Expression of Cyclin E/Cdk2/p27Kip1 in Growth Hormone Adenomas
      Wei Dong, Haibo Zhu, Hua Gao, Wenjian Shi, Yu Zhang, Hongyun Wang, Chuzhong Li, Guidong Song, Yazhuo Zhang
      World Neurosurgery.2019; 121: e45.     CrossRef
    • Acromegaly
      Annamaria Colao, Ludovica F. S. Grasso, Andrea Giustina, Shlomo Melmed, Philippe Chanson, Alberto M. Pereira, Rosario Pivonello
      Nature Reviews Disease Primers.2019;[Epub]     CrossRef
    • Differenzialdiagnostik und Therapie der Hypophysenadenome
      M. Hlaváč, F. Sommer, G. Karpel-Massler, R. Wirtz, T. Hoffmann, A. Paľa
      HNO.2019; 67(4): 307.     CrossRef
    • Medical Management of Cushing’s Syndrome: Current and Emerging Treatments
      José Miguel Hinojosa-Amaya, Daniel Cuevas-Ramos, Maria Fleseriu
      Drugs.2019; 79(9): 935.     CrossRef
    • Aggressive and Malignant Prolactinomas
      Nicoleta Cristina Olarescu, Luis G. Perez-Rivas, Federico Gatto, Thomas Cuny, Maria A. Tichomirowa, Gianluca Tamagno, Manuel D. Gahete
      Neuroendocrinology.2019; 109(1): 57.     CrossRef
    • New Insights in Cushing Disease Treatment With Focus on a Derivative of Vitamin A
      Mariana Fuertes, Julieta Tkatch, Josefina Rosmino, Leandro Nieto, Mirtha Adriana Guitelman, Eduardo Arzt
      Frontiers in Endocrinology.2018;[Epub]     CrossRef
    • Prenatal alcohol exposure increases the susceptibility to develop aggressive prolactinomas in the pituitary gland
      Shaima Jabbar, Kenneth Reuhl, Dipak K. Sarkar
      Scientific Reports.2018;[Epub]     CrossRef
    • Aggressive and malignant pituitary tumours: state-of-the-art
      Dorota Dworakowska, Ashley B Grossman
      Endocrine-Related Cancer.2018; 25(11): R559.     CrossRef
    • Epidemiología y caracterización general de los tumores cerebrales primarios en el adulto
      Maria Isabel Ocampo Navia, Juan Carlos Gómez Vega, Oscar Hernando Feo Lee
      Universitas Médica.2018;[Epub]     CrossRef
    • Triptolide inhibits pituitary adenoma cell viability, migration and invasion via ADAM12/EGFR signaling pathway
      Junwen Wang, Zhuo Zhang, Ran Li, Wei Sun, Juan Chen, Huaqiu Zhang, Kai Shu, Ting Lei
      Life Sciences.2018; 194: 150.     CrossRef
    • Emerging roles of bexarotene in the prevention, treatment and anti-drug resistance of cancers
      Danyang Shen, Xiaoming Yu, Yan Wu, Yuanlei Chen, Gonghui Li, Feng Cheng, Liqun Xia
      Expert Review of Anticancer Therapy.2018; 18(5): 487.     CrossRef
    • New Molecular Targets for Treatment of Cushing’s Disease
      Elizabeth Foulkes, John Newell-Price
      Endocrinology and Metabolism Clinics of North America.2018; 47(2): 367.     CrossRef
    • Pituitary-Directed Therapies for Cushing’s Disease
      Fabienne Langlois, Jennifer Chu, Maria Fleseriu
      Frontiers in Endocrinology.2018;[Epub]     CrossRef
    • Updates in Diagnosis and Treatment of Acromegaly
      Roula Zahr, Maria Fleseriu
      European Endocrinology.2018; 14(2): 57.     CrossRef
    • Root cause analysis of diagnostic and surgical failures in the treatment of suspected Cushing’s disease
      Hasan A. Zaidi, David L. Penn, David J. Cote, Edward R. Laws
      Journal of Clinical Neuroscience.2018; 53: 153.     CrossRef


    Endocrinol Metab : Endocrinology and Metabolism