436
Views
9
CrossRef citations to date
0
Altmetric
Reviews

Emerging drugs for acromegaly

&

Bibliography

  • Bengtsson BA, Edén S, Ernest I, et al. Epidemiology and long-term survival in acromegaly. A study of 166 cases diagnosed between 1955 and 1984. Acta Med Scand 1988;223:327-35
  • Etxabe J, Gaztambide S, Latorre P, Vazquez JA. Acromegaly: an epidemiological study. J Endocrinol Invest 1993;16:181-7
  • Ritchie CM, Atkinson AB, Kennedy AL, et al. Ascertainment and natural history of treated acromegaly in Northern Ireland. Ulster Med J 1990;59:55-62
  • Holdaway IM, Rajasoorya C. Epidemiology of acromegaly. Pituitary 1999;2:29-41
  • Kwon O, Song YD, Kim SY, Lee EJ. Nationwide survey of acromegaly in South Korea. Clin Endocrinol (Oxf) 2013;78:577-85
  • Sesmilo G. Epidemiology of acromegaly in Spain. Endocrinol Nutr 2013;60:470-4
  • Daly AF, Rixhon M, Adam C, et al. High prevalence of pituitary adenomas: a cross-sectional study in the province of Liege, Belgium. J Clin Endocrinol Metab 2006;91:4769-75
  • Schneider HJ, Sievers C, Saller B, et al. High prevalence of biochemical acromegaly in primary care patients with elevated IGF-1 levels. Clin Endocrinol (Oxf) 2008;69:432-5
  • Nachtigall L, Delgado A, Swearingen B, et al. Changing patterns in diagnosis and therapy of acromegaly over two decades. J Clin Endocrinol Metab 2008;93:2035-41
  • Nabarro JD. Acromegaly. Clin Endocrinol (Oxf) 1987;26:481-512
  • Reid TJ, Post KD, Bruce JN, et al. Features at diagnosis of 324 patients with acromegaly did not change from 1981 to 2006: acromegaly remains under-recognized and under-diagnosed. Clin Endocrinol (Oxf) 2010;72:203-8
  • Katznelson L. Approach to the patient with persistent acromegaly after pituitary surgery. J Clin Endocrinol Metab 2010;95:4114-23
  • Schneider HJ, Kosilek RP, Günther M, et al. A novel approach to the detection of acromegaly: accuracy of diagnosis by automatic face classification. J Clin Endocrinol Metab 2011;96:2074-80
  • Melmed S, Casanueva FF, Klibanski A, et al. A consensus on the diagnosis and treatment of acromegaly complications. Pituitary 2013;16:294-302
  • Guinto G, Abdo M, Zepeda E, et al. Acromegaly: role of surgery in the therapeutic armamentarium. Int J Endocrinol 2012;2012:306094
  • Ghazi AA, Amirbaigloo A, Dezfooli AA, et al. Ectopic acromegaly due to growth hormone releasing hormone. Endocrine 2013;43:293-302
  • Losa M, von Werder K. Pathophysiology and clinical aspects of the ectopic GH-releasing hormone syndrome. Clin Endocrinol (Oxf) 1997;47:123-35
  • Garby L, Caron P, Claustrat F, et al. Clinical characteristics and outcome of acromegaly induced by ectopic secretion of growth hormone-releasing hormone (GHRH): a French nationwide series of 21 cases. J Clin Endocrinol Metab 2012;97:2093-104
  • Melmed S. Medical progress: acromegaly. N Engl J Med 2006;355:2558-73
  • Giustina A, Barkan A, Casanueva FF, et al. Criteria for cure of acromegaly: a consensus statement. J Clin Endocrinol Metab 2000;85:526-9
  • Giustina A, Chanson P, Bronstein MD, et al. A consensus on criteria for cure of acromegaly. J Clin Endocrinol Metab 2010;95:3141-8
  • Melmed S. Acromegaly pathogenesis and treatment. J Clin Invest 2009;119:3189-202
  • Colao A, Ferone D, Marzullo P, Lombardi G. Systemic complications of acromegaly: epidemiology, pathogenesis, and management. Endocr Rev 2004;25:102-52
  • Wright AD, Hill DM, Lowy C, Fraser TR. Mortality in acromegaly. Q J Med 1970;39:1-16
  • Maison P, Tropeano A-I, Macquin-Mavier I, et al. Mortality in acromegaly: a metaanalysis. J Clin Endocrinol Metab 2007;93:61-7
  • Orme SM, McNally RJ, Cartwright RA, Belchetz PE. Mortality and cancer incidence in acromegaly: a retrospective cohort study. United Kingdom Acromegaly Study Group. J Clin Endocrinol Metab 1998;83:2730-4
  • Dekkers OM, Biermasz NR, Pereira a M, et al. Mortality in acromegaly: a metaanalysis. J Clin Endocrinol Metab 2008;93:61-7
  • Holdaway IM. Factors influencing mortality in acromegaly. J Clin Endocrinol Metab 2004;89:667-74
  • Holdaway IM, Bolland MJ, Gamble GD. A meta-analysis of the effect of lowering serum levels of GH and IGF-I on mortality in acromegaly. Eur J Endocrinol 2008;159:89-95
  • Alexander L, Appleton D, Hall R, et al. Epidemiology of acromegaly in the Newcastle region. Clin Endocrinol (Oxf) 1980;12:71-9
  • Melmed S. Acromegaly and cancer: not a problem? J Clin Endocrinol Metab 2001;86:2929-34
  • Rajasoorya C, Holdaway IM, Wrightson P, et al. Determinants of clinical outcome and survival in acromegaly. Clin Endocrinol (Oxf) 1994;41:95-102
  • Ben-Shlomo A, Sheppard MC, Stephens JM, et al. Clinical, quality of life, and economic value of acromegaly disease control. Pituitary 2011;14:284-94
  • Adelman DT, Liebert KJ, Nachtigall LB, et al. Acromegaly: the disease, its impact on patients, and managing the burden of long-term treatment. Int J Gen Med 2013;6:31-8
  • Heaney A, Edling K. An update on the treatment of acromegaly. Res Rep Endocr Disord 2013;3:1-11
  • Jallad RS, Bronstein MD. The place of medical treatment of acromegaly: current status and perspectives. Expert Opin Pharmacother 2013;14:1001-15
  • Schöfl C, Franz H, Grussendorf M, et al. Long-term outcome in patients with acromegaly: analysis of 1344 patients from the German Acromegaly Register. Eur J Endocrinol 2013;168:39-47
  • Sesmilo G, Gaztambide S, Venegas E, et al. Changes in acromegaly treatment over four decades in Spain: analysis of the Spanish Acromegaly Registry (REA). Pituitary 2013;16:115-21
  • Kauppinen-Mäkelin R, Sane T, Reunanen A, et al. A nationwide survey of mortality in acromegaly. J Clin Endocrinol Metab 2005;90:4081-6
  • Annamalai AK, Webb A, Kandasamy N, et al. A comprehensive study of clinical, biochemical, radiological, vascular, cardiac, and sleep parameters in an unselected cohort of patients with acromegaly undergoing presurgical somatostatin receptor ligand therapy. J Clin Endocrinol Metab 2013;98:1040-50
  • Colao A, Auriemma RS, Pivonello R, et al. Medical consequences of acromegaly: what are the effects of biochemical control? Rev Endocr Metab Disord 2008;9:21-31
  • Claessen KM, Ramautar SR, Pereira AM, et al. Progression of acromegalic arthropathy despite long-term biochemical control: a prospective, radiological study. Eur J Endocrinol 2012;167:235-44
  • Swearingen B. Update on pituitary surgery. J Clin Endocrinol Metab 2012;97:1073-81
  • Dorward NL. Endocrine outcomes in endoscopic pituitary surgery: a literature review. Acta Neurochir (Wien) 2010;152:1275-9
  • Jane JA, Starke RM, Elzoghby MA, et al. Endoscopic transsphenoidal surgery for acromegaly: remission using modern criteria, complications, and predictors of outcome. J Clin Endocrinol Metab 2011;96:2732-40
  • Besser GM, Burman P, Daly AF. Predictors and rates of treatment-resistant tumor growth in acromegaly. Eur J Endocrinol 2005;153:187-93
  • Ross DA, Wilson CB. Results of transsphenoidal microsurgery for growth hormone-secreting pituitary adenoma in a series of 214 patients. J Neurosurg 1988;68:854-67
  • Tindall GT, Oyesiku NM, Watts NB, et al. Transsphenoidal adenomectomy for growth hormone-secreting pituitary adenomas in acromegaly: outcome analysis and determinants of failure. J Neurosurg 1993;78:205-15
  • Gittoes NJ, Sheppard MC, Johnson a P, Stewart PM. Outcome of surgery for acromegaly–the experience of a dedicated pituitary surgeon. QJM 1999;92:741-5
  • Ahmed S, Elsheikh M, Stratton IM, et al. Outcome of transphenoidal surgery for acromegaly and its relationship to surgical experience. Clin Endocrinol (Oxf) 1999;50:561-7
  • Osman IA, James RA, Chatterjee S, et al. Factors determining the long-term outcome of surgery for acromegaly. QJM 1994;87:617-23
  • Buchfelder M, Schlaffer S-M. Intraoperative magnetic resonance imaging during surgery for pituitary adenomas: pros and cons. Endocrine 2012;42:483-95
  • Melmed S, Colao A, Barkan A, et al. Guidelines for acromegaly management: an update. J Clin Endocrinol Metab 2009;94:1509-17
  • Loeffler JS, Shih HA. Radiation therapy in the management of pituitary adenomas. J Clin Endocrinol Metab 2011;96:1992-2003
  • Minniti G, Scaringi C, Enrici RM. Radiation techniques for acromegaly. Radiat Oncol 2011;6:167
  • Jenkins PJ, Bates P, Carson MN, et al. Conventional pituitary irradiation is effective in lowering serum growth hormone and insulin-like growth factor-I in patients with acromegaly. J Clin Endocrinol Metab 2006;91:1239-45
  • Barrande G, Pittino-Lungo M, Coste J, et al. Hormonal and metabolic effects of radiotherapy in acromegaly: long-term results in 128 patients followed in a single center. J Clin Endocrinol Metab 2000;85:3779-85
  • Pollock BE, Jacob JT, Brown PD, Nippoldt TB. Radiosurgery of growth hormone-producing pituitary adenomas: factors associated with biochemical remission. J Neurosurg 2007;106:833-8
  • Jagannathan J, Sheehan JP, Pouratian N, et al. Gamma knife radiosurgery for acromegaly: outcomes after failed transsphenoidal surgery. Neurosurgery 2008;62:1262-70
  • Melmed S, Jackson I, Kleinberg D, Klibanski A. Current treatment guidelines for acromegaly. J Clin Endocrinol Metab 1998;83:2646-52
  • Vance ML, Laws ER. Role of medical therapy in the management of acromegaly. Neurosurgery 2005;56:877-85
  • Zhang Y, Chan HF, Leong KW. Advanced materials and processing for drug delivery: the past and the future. Adv Drug Deliv Rev 2013;65:104-20
  • Unger N, Ueberberg B, Schulz S, et al. Differential expression of somatostatin receptor subtype 1-5 proteins in numerous human normal tissues. Exp Clin Endocrinol Diabetes 2012;120:482-9
  • Patel YC, Greenwood MT, Panetta R, et al. The somatostatin receptor family. Life Sci 1995;57:1249-65
  • Freda PU. Somatostatin analogs in acromegaly. J Clin Endocrinol Metab 2002;87:3013-18
  • Stewart PM, Kane KF, Stewart SE, et al. Depot long-acting somatostatin analog (Sandostatin-LAR) is an effective treatment for acromegaly. J Clin Endocrinol Metab 1995;80:3267-72
  • Salvatori R, Woodmansee W, Clary C, et al. Efficacy and safety in the Somatuline® depot (lanreotide) injection for acromegaly (SODA) study conducted in the U.S.: overall year 1 data and comparison of home injection versus office injection. Endocr Rev 2012;33:SUN-746
  • Bevan JS. Clinical review: the antitumoral effects of somatostatin analog therapy in acromegaly. J Clin Endocrinol Metab 2005;90:1856-63
  • Maiza JC, Vezzosi D, Matta M, et al. Long-term (up to 18 years) effects on GH/IGF-1 hypersecretion and tumour size of primary somatostatin analogue (SSTa) therapy in patients with GH-secreting pituitary adenoma responsive to SSTa. Clin Endocrinol (Oxf) 2007;67:282-9
  • Caron P, Bevan JS, Clermont A, Maisonobe P. Early and sustained tumour volume reduction and GH/IGF1 control in patients with GH-secreting pituitary macroadenoma primarily treated with lanreotide Autogel 120 mg for 48 weeks: the PRIMARYS study. Endocr Abstr 2013;32:P840
  • Colao A, Auriemma RS, Lombardi G, Pivonello R. Resistance to somatostatin analogs in acromegaly. Endocr Rev 2011;32:247-71
  • Freda PU, Katznelson L, van der Lely AJ, et al. Long-acting somatostatin analog therapy of acromegaly: a meta-analysis. J Clin Endocrinol Metab 2005;90:4465-73
  • Murray RD, Melmed S. A critical analysis of clinically available somatostatin analog formulations for therapy of acromegaly. J Clin Endocrinol Metab 2008;93:2957-68
  • Barnett P. Somatostatin and somatostatin receptor physiology. Endocrine 2003;20:255-64
  • Manjila S, Wu OC, Khan FR, et al. Pharmacological management of acromegaly: a current perspective. Neurosurg Focus 2010;29:E14
  • Plöckinger U, Dienemann D, Quabbe HJ. Gastrointestinal side-effects of octreotide during long-term treatment of acromegaly. J Clin Endocrinol Metab 1990;71:1658-62
  • Alexopoulou O, Abrams P, Verhelst J, et al. Efficacy and tolerability of lanreotide Autogel therapy in acromegalic patients previously treated with octreotide LAR. Eur J Endocrinol 2004;151:317-24
  • Ben-Shlomo A, Melmed S. Somatostatin agonists for treatment of acromegaly. Mol Cell Endocrinol 2008;286:192-8
  • Di Cianni A, Carotenuto A, Brancaccio D, et al. Novel octreotide dicarba-analogues with high affinity and different selectivity for somatostatin receptors. J Med Chem 2010;53:6188-97
  • Carmichael JD. Lanreotide depot deep subcutaneous injection: a new method of delivery and its associated benefits. Patient Prefer Adherence 2012;6:73-82
  • Schopohl J, Strasburger CJ, Caird D, et al. Efficacy and acceptability of lanreotide Autogel® 120 mg at different dose intervals in patients with acromegaly previously treated with octreotide LAR. Exp Clin Endocrinol Diabetes 2011;119:156-62
  • Abrams P, Alexopoulou O, Abs R, et al. Optimalization and cost management of lanreotide-Autogel therapy in acromegaly. Eur J Endocrinol 2007;157:571-7
  • Salvatori R, Nachtigall LB, Cook DM, et al. Effectiveness of self- or partner-administration of an extended-release aqueous-gel formulation of lanreotide in lanreotide-naïve patients with acromegaly. Pituitary 2010;13:115-22
  • Bevan JS, Newell-Price J, Wass JA, et al. Home administration of lanreotide Autogel by patients with acromegaly, or their partners, is safe and effective. Clin Endocrinol (Oxf) 2008;68:343-9
  • Ronchi CL, Rizzo E, Lania AG, et al. Preliminary data on biochemical remission of acromegaly after somatostatin analogs withdrawal. Eur J Endocrinol 2008;158:19-25
  • Ramírez C, Vargas G, González B, et al. Discontinuation of octreotide LAR after long term, successful treatment of patients with acromegaly: is it worth trying? Eur J Endocrinol 2012;166:21-6
  • Oberg K. Future aspects of somatostatin-receptor-mediated therapy. Neuroendocrinology 2004;80(Suppl 1):57-61
  • Colao A, Pivonello R, Auriemma RS, et al. Beneficial effect of dose escalation of octreotide-LAR as first-line therapy in patients with acromegaly. Eur J Endocrinol 2007;157:579-87
  • Giustina A, Bonadonna S, Bugari G, et al. High-dose intramuscular octreotide in patients with acromegaly inadequately controlled on conventional somatostatin analogue therapy: a randomised controlled trial. Eur J Endocrinol 2009;161:331-8
  • Ludlam WH, Anthony L. Safety review: dose optimization of somatostatin analogs in patients with acromegaly and neuroendocrine tumors. Adv Ther 2011;28:825-41
  • Fleseriu M. Clinical efficacy and safety results for dose escalation of somatostatin receptor ligands in patients with acromegaly: a literature review. Pituitary 2011;14:184-93
  • Thorner MO, Strasburger CJ, Wu Z, et al. Growth hormone (GH) receptor blockade with a PEG-modified GH (B2036-PEG) lowers serum insulin-like growth factor-I but does not acutely stimulate serum GH. J Clin Endocrinol Metab 1999;84:2098-103
  • Cunningham BC, Wells JA. High-resolution epitope mapping of hGH-receptor interactions by alanine-scanning mutagenesis. Science 1989;244:1081-5
  • Kang JS, Deluca PP, Lee KC. Emerging PEGylated drugs. Expert Opin Emerg Drugs 2009;14:363-80
  • Ross RJ, Leung KC, Maamra M, et al. Binding and functional studies with the growth hormone receptor antagonist, B2036-PEG (pegvisomant), reveal effects of pegylation and evidence that it binds to a receptor dimer. J Clin Endocrinol Metab 2001;86:1716-23
  • Zhang Y, Jiang J, Kopchick JJ, Frank SJ. Disulfide linkage of growth hormone (GH) receptors (GHR) reflects GH-induced GHR dimerization. Association of JAK2 with the GHR is enhanced by receptor dimerization. J Biol Chem 1999;274:33072-84
  • Harding PA, Wang X, Okada S, et al. Growth hormone (GH) and a GH antagonist promote GH receptor dimerization and internalization. J Biol Chem 1996;271:6708-12
  • Maamra M, Finidori J, Von Laue S, et al. Studies with a growth hormone antagonist and dual-fluorescent confocal microscopy demonstrate that the full-length human growth hormone receptor, but not the truncated isoform, is very rapidly internalized independent of Jak2-Stat5 signaling. J Biol Chem 1999;274:14791-8
  • Govers R, ten Broeke T, van Kerkhof P, et al. Identification of a novel ubiquitin conjugation motif, required for ligand-induced internalization of the growth hormone receptor. EMBO J 1999;18:28-36
  • Govers R, van Kerkhof P, Schwartz AL, Strous GJ. Di-leucine-mediated internalization of ligand by a truncated growth hormone receptor is independent of the ubiquitin conjugation system. J Biol Chem 1998;273:16426-33
  • Govers R, van Kerkhof P, Schwartz a L, Strous GJ. Linkage of the ubiquitin-conjugating system and the endocytic pathway in ligand-induced internalization of the growth hormone receptor. EMBO J 1997;16:4851-8
  • Trainer PJ, Drake WM, Katznelson L, et al. Treatment of acromegaly with the growth hormone-receptor antagonist pegvisomant. N Engl J Med 2000;342:1171-7
  • Van der Lely AJ, Hutson RK, Trainer PJ, et al. Long-term treatment of acromegaly with pegvisomant, a growth hormone receptor antagonist. Lancet 2001;358:1754-9
  • Buchfelder M, Schlaffer S, Droste M, et al. The German ACROSTUDY: past and present. Eur J Endocrinol 2009;161:S3-S10
  • Bianchi A, Valentini F, Iuorio R, et al. Long-term treatment of somatostatin analog-refractory growth hormone-secreting pituitary tumors with pegvisomant alone or combined with long-acting somatostatin analogs: a retrospective analysis of clinical practice and outcomes. J Exp Clin Cancer Res 2013;32:40
  • Van der Lely a J, Biller BMK, Brue T, et al. Long-term safety of pegvisomant in patients with acromegaly: comprehensive review of 1288 subjects in ACROSTUDY. J Clin Endocrinol Metab 2012;97:1589-97
  • Higham CE, Thomas JDJ, Bidlingmaier M, et al. Successful use of weekly pegvisomant administration in patients with acromegaly. Eur J Endocrinol 2009;161:21-5
  • Jehle S, Reyes CM, Sundeen RE, Freda PU. Alternate-day administration of pegvisomant maintains normal serum insulin-like growth factor-I levels in patients with acromegaly. J Clin Endocrinol Metab 2005;90:1588-93
  • Biering H, Saller B, Bauditz J, et al. Elevated transaminases during medical treatment of acromegaly: a review of the German pegvisomant surveillance experience and a report of a patient with histologically proven chronic mild active hepatitis. Eur J Endocrinol 2006;154:213-20
  • Madsen M, Krusenstjerna-Hafstrøm T, Møller L, et al. Fat content in liver and skeletal muscle changes in a reciprocal manner in patients with acromegaly during combination therapy with a somatostatin analog and a GH receptor antagonist: a randomized clinical trial. J Clin Endocrinol Metab 2012;97:1227-35
  • Buchfelder M, Weigel D, Droste M, et al. Pituitary tumor size in acromegaly during pegvisomant treatment: experience from MR re-evaluations of the German Pegvisomant Observational Study. Eur J Endocrinol 2009;161:27-35
  • Fedele M, Pierantoni GM, Fusco A. The genetics of pituitary adenomas. In: Diamanti-Kandarakis E, editor. Contemporary aspects of endocrinology. InTech; Rijeka; 2011
  • Abs R, Verhelst J, Maiter D, et al. Cabergoline in the treatment of acromegaly: a study in 64 patients. J Clin Endocrinol Metab 1998;83:374-8
  • Moyes VJ, Metcalfe KA, Drake WM. Clinical use of cabergoline as primary and adjunctive treatment for acromegaly. Eur J Endocrinol 2008;159:541-5
  • Sandret L, Maison P, Chanson P. Place of cabergoline in acromegaly: a meta-analysis. J Clin Endocrinol Metab 2011;96:1327-35
  • Zanettini R, Antonini A, Gatto G, et al. Valvular heart disease and the use of dopamine agonists for Parkinson's disease. N Engl J Med 2007;356:39-46
  • Auriemma RS, Pivonello R, Perone Y, et al. Safety of long-term treatment with cabergoline on cardiac valve disease in patients with prolactinomas. Eur J Endocrinol 2013;169:359-66
  • Maione L, Garcia C, Bouchachi A, et al. No evidence of a detrimental effect of cabergoline therapy on cardiac valves in patients with acromegaly. J Clin Endocrinol Metab 2012;97:E1714-19
  • Fleseriu M. The role of combination medical therapy in acromegaly: hope for the nonresponsive patient. Curr Opin Endocrinol Diabetes Obes 2013;20:321-9
  • Moore DJ, Adi Y, Connock MJ, Bayliss S. Clinical effectiveness and cost-effectiveness of pegvisomant for the treatment of acromegaly: a systematic review and economic evaluation. BMC Endocr Disord 2009;9:20
  • Higham CE, Atkinson a B, Aylwin S, et al. Effective combination treatment with cabergoline and low-dose pegvisomant in active acromegaly: a Prospective Clinical Trial. J Clin Endocrinol Metab 2012;97:1187-93
  • Knutzen R, Ezzat S. The cost of medical care for the acromegalic patient. Neuroendocrinology 2006;83:139-44
  • Wilson LS, Shin JL, Ezzat S. Longitudinal assessment of economic burden and clinical outcomes in acromegaly. Endocr Pract 2001;7:170-80
  • Roset M, Merino-Montero S, Luque-Ramírez M, et al. Cost of clinical management of acromegaly in Spain. Clin Drug Investig 2012;32:235-45
  • Sowiński J, Sawicka N, Piątek K, Zybek A. Pharmacoeconomic aspects of the treatment of pituitary gland tumours. Contemp Oncol 2013;17:137-43
  • Luque-Ramírez M, Paramo C, Varela da Costa C, García-Mayor RV. Cost of management of invasive growth hormone-secreting macroadenoma. J Endocrinol Invest 2007;30:541-5
  • Marty R, Roze S, Kurth H. Decision-tree model for health economic comparison of two long-acting somatostatin receptor ligand devices in France, Germany, and the UK. Med Devices (Auckl) 2012;5:39-44
  • Marko NF, LaSota E, Hamrahian AH, Weil RJ. Comparative effectiveness review of treatment options for pituitary microadenomas in acromegaly. J Neurosurg 2012;117:522-38
  • Novartis Corporate publications [Internet]
  • IPSEN Group Website Publications [Internet]
  • Pfizer Financial Reports [Internet]
  • Didoni G, Grottol S, Gasco V, et al. Cost-of-illness study in acromegalic patients in Italy. J Endocrinol Invest 2004;27:1034-9
  • Colao A, Cappabianca P, Caron P, et al. Octreotide LAR vs. surgery in newly diagnosed patients with acromegaly: a randomized, open-label, multicentre study. Clin Endocrinol (Oxf) 2009;70:757-68
  • Mercado M, Borges F, Bouterfa H, et al. A prospective, multicentre study to investigate the efficacy, safety and tolerability of octreotide LAR (long-acting repeatable octreotide) in the primary therapy of patients with acromegaly. Clin Endocrinol (Oxf) 2007;66:859-68
  • Sherlock M, Woods C, Sheppard MC. Medical therapy in acromegaly. Nat Rev Endocrinol 2011;7:291-300
  • Kasuki L, Marques NV, Nuez MJ, et al. Acromegalic patients lost to follow-up: a pilot study. Pituitary 2013;16:245-50
  • Melmed S. Pathogenesis of pituitary tumors. Nat Rev Endocrinol 2011;7:257-66
  • Attwooll C, Lazzerini Denchi E, Helin K. The E2F family: specific functions and overlapping interests. EMBO J 2004;23:4709-16
  • Lania A, Mantovani G, Spada A. Genetics of pituitary tumors: focus on G-protein mutations. Exp Biol Med (Maywood) 2003;228:1004-17
  • Spada A, Lania A, Ballarè E. G protein abnormalities in pituitary adenomas. Mol Cell Endocrinol 1998;142:1-14
  • Lopes MB. Growth hormone-secreting adenomas: pathology and cell biology. Neurosurg Focus 2010;29:E2
  • Sharpless NE, DePinho RA. Telomeres, stem cells, senescence, and cancer. J Clin Invest 2004;113:160-8
  • Schmitt CA, Fridman JS, Yang M, et al. A senescence program controlled by p53 and p16INK4a contributes to the outcome of cancer therapy. Cell 2002;109:335-46
  • Campisi J. Suppressing cancer: the importance of being senescent. Science 2005;309:886-7
  • Serrano M, Lin AW, McCurrach ME, et al. Oncogenic ras provokes premature cell senescence associated with accumulation of p53 and p16INK4a. Cell 1997;88:593-602
  • Bartkova J, Rezaei N, Liontos M, et al. Oncogene-induced senescence is part of the tumorigenesis barrier imposed by DNA damage checkpoints. Nature 2006;444:633-7
  • Vlotides G, Eigler T, Melmed S. Pituitary tumor-transforming gene: physiology and implications for tumorigenesis. Endocr Rev 2007;28:165-86
  • Reubi JC, Kvols L, Krenning E, Lamberts SW. Distribution of somatostatin receptors in normal and tumor tissue. Metabolism 1990;39:78-81
  • Narayanan RP, Siddals KW, Heald AH, Gibson JM. Interactions of the IGF system with diabetes and its vascular complications. Exp Clin Endocrinol Diabetes 2013;121:255-61
  • Brown RJ, Adams JJ, Pelekanos RA, et al. Model for growth hormone receptor activation based on subunit rotation within a receptor dimer. Nat Struct Mol Biol 2005;12:814-21
  • Argetsinger LS, Campbell GS, Yang X, et al. Identification of JAK2 as a growth hormone receptor-associated tyrosine kinase. Cell 1993;74:237-44
  • Greenhalgh CJ, Rico-Bautista E, Lorentzon M, et al. SOCS2 negatively regulates growth hormone action in vitro and in vivo. J Clin Invest 2005;115:397-406
  • Rowlinson SW, Yoshizato H, Barclay JL, et al. An agonist-induced conformational change in the growth hormone receptor determines the choice of signalling pathway. Nat Cell Biol 2008;10:740-7
  • Brooks AJ, Wooh JW, Tunny KA, Waters MJ. Growth hormone receptor; mechanism of action. Int J Biochem Cell Biol 2008;40:1984-9
  • Dworakowska D, Wlodek E, Leontiou CA, et al. Activation of RAF/MEK/ERK and PI3K/AKT/mTOR pathways in pituitary adenomas and their effects on downstream effectors. Endocr Relat Cancer 2009;16:1329-38
  • Suojun Z, Feng W, Dongsheng G, Ting L. Targeting Raf/MEK/ERK pathway in pituitary adenomas. Eur J Cancer 2012;48:389-95
  • Bruns C, Lewis I, Briner U, et al. SOM230: a novel somatostatin peptidomimetic with broad somatotropin release inhibiting factor (SRIF) receptor binding and a unique antisecretory profile. Eur J Endocrinol 2002;146:707-16
  • Petersenn S, Schopohl J, Barkan A, et al. Pasireotide (SOM230) demonstrates efficacy and safety in patients with acromegaly: a randomized, multicenter, phase II trial. J Clin Endocrinol Metab 2010;95:2781-9
  • Dietrich H, Hu K, Ruffin M, et al. Safety, tolerability, and pharmacokinetics of a single dose of pasireotide long-acting release in healthy volunteers: a single-center phase I study. Eur J Endocrinol 2012;166:821-8
  • Golor G, Hu K, Ruffin M, et al. A first-in-man study to evaluate the safety, tolerability, and pharmacokinetics of pasireotide (SOM230), a multireceptor-targeted somatostatin analog, in healthy volunteers. Drug Des Devel Ther 2012;6:71-9
  • Henry RR, Ciaraldi TP, Armstrong D, et al. Hyperglycemia associated with pasireotide: results from a mechanistic study in healthy volunteers. J Clin Endocrinol Metab 2013;98:3446-53
  • Henry RR, Mudaliar S, Wetli-Hermosillo K, et al. Mechanism and management of hyperglycemia associated with pasireotide: results from studies in healthy volunteers. Endocr Abstr 2011;260:P260
  • Reznik Y, Bertherat J, Borson-Chazot F, et al. Management of hyperglycaemia in Cushing's disease: experts' proposals on the use of pasireotide. Diabetes Metab 2013;39:34-41
  • Colao A, Bronstein MD, Freda PU, et al. Pasireotide LAR is significantly more effective than octreotide LAR at inducing biochemical control in patients with acromegaly: results of a 12-month randomized, double-blind, multicenter, phase III study. Endocr Abstr 2012;29:OC1.1
  • Cano-Cebrián MJ, Zornoza T, Granero L, Polache A. Intestinal absorption enhancement via the paracellular route by fatty acids, chitosans and others: a target for drug delivery. Curr Drug Deliv 2005;2:9-22
  • Tuvia S, Salama P, Weinstein I, et al. OR14,80 OctreolinTM, a safe oral alternative for parenteral octreotide treatment. Growth Horm IGF Res 2010;20:S35-6
  • Tuvia S, Atsmon J, Teichman SL, et al. Oral octreotide absorption in human subjects: comparable pharmacokinetics to parenteral octreotide and effective growth hormone suppression. J Clin Endocrinol Metab 2012;97:1-8
  • Shimon I, Rubinek T, Hadani M, Alhadef N. PTR-3173 (somatoprim), a novel somatostatin analog with affinity for somatostatin receptors 2, 4 and 5 is a potent inhibitor of human GH secretion. J Endocrinol Invest 2004;27:721-7
  • Plöckinger U, Hoffmann U, Geese M, et al. DG3173 (somatoprim), a unique somatostatin receptor subtypes 2-, 4- and 5-selective analogue, effectively reduces GH secretion in human GH-secreting pituitary adenomas even in Octreotide non-responsive tumours. Eur J Endocrinol 2012;166:223-34
  • Afargan M, Janson ET, Gelerman G, et al. Novel long-acting somatostatin analog with endocrine selectivity: potent suppression of growth hormone but not of insulin. Endocrinology 2001;142:477-86
  • Maggio ET, Grasso P. Oral delivery of octreotide acetate in Intravail® improves uptake, half-life, and bioavailability over subcutaneous administration in male Swiss webster mice. Regul Pept 2011;167:233-8
  • Gadelha MR, Chieffo C, Bai SA, et al. A subcutaneous octreotide hydrogel implant for the treatment of acromegaly. Endocr Pract 2012;18:870-81
  • Chieffo C, Cook D, Xiang Q, Frohman LA. Efficacy and safety of an octreotide implant in the treatment of patients with acromegaly. J Clin Endocrinol Metab 2013;98:4047-54
  • Jaquet P, Gunz G, Saveanu A, et al. BIM-23A760, a chimeric molecule directed towards somatostatin and dopamine receptors, vs universal somatostatin receptors ligands in GH-secreting pituitary adenomas partial responders to octreotide. J Endocrinol Invest 2005;28:21-7
  • Culler MD. Somatostatin-dopamine chimeras: a novel approach to treatment of neuroendocrine tumors. Horm Metab Res 2011;43:854-7
  • Jaquet P, Gunz G, Saveanu A, et al. Efficacy of chimeric molecules directed towards multiple somatostatin and dopamine receptors on inhibition of GH and prolactin secretion from GH-secreting pituitary adenomas classified as partially responsive to somatostatin analog therapy. Eur J Endocrinol 2005;153:135-41
  • Gruszka A, Culler MD, Melmed S. Somatostatin analogs and chimeric somatostatin-dopamine molecules differentially regulate human growth hormone and prolactin gene expression and secretion in vitro. Mol Cell Endocrinol 2012;362:104-9
  • Tachas G, Lofthouse S, Wraight CJ, et al. A GH receptor antisense oligonucleotide inhibits hepatic GH receptor expression, IGF-I production and body weight gain in normal mice. J Endocrinol 2006;189:147-54
  • Stevens MF, Hickman JA, Langdon SP, et al. Antitumor activity and pharmacokinetics in mice of 8-carbamoyl-3-methyl-imidazo[5,1-d]-1,2,3,5-tetrazin-4(3H)-one (CCRG 81045; M & B 39831), a novel drug with potential as an alternative to dacarbazine. Cancer Res 1987;47:5846-52
  • Hegi ME, Diserens A-C, Gorlia T, et al. MGMT gene silencing and benefit from temozolomide in glioblastoma. N Engl J Med 2005;352:997-1003
  • Kovacs K, Horvath E, Syro L V, et al. Temozolomide therapy in a man with an aggressive prolactin-secreting pituitary neoplasm: morphological findings. Hum Pathol 2007;38:185-9
  • Byrne S, Karapetis C, Vrodos N. A novel use of temozolomide in a patient with malignant prolactinoma. J Clin Neurosci 2009;16:1694-6
  • Losa M, Mazza E, Terreni MR, et al. Salvage therapy with temozolomide in patients with aggressive or metastatic pituitary adenomas: experience in six cases. Eur J Endocrinol 2010;163:843-51
  • Melmed S, Casanueva FF, Hoffman AR, et al. Diagnosis and treatment of hyperprolactinemia: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab 2011;96:273-88
  • Jacinto F V, Esteller M. MGMT hypermethylation: a prognostic foe, a predictive friend. DNA Repair (Amst) 2007;6:1155-60
  • McCormack AI, McDonald KL, Gill AJ, et al. Low O6-methylguanine-DNA methyltransferase (MGMT) expression and response to temozolomide in aggressive pituitary tumours. Clin Endocrinol (Oxf) 2009;71:226-33
  • Raverot G, Sturm N, de Fraipont F, et al. Temozolomide treatment in aggressive pituitary tumors and pituitary carcinomas: a French multicenter experience. J Clin Endocrinol Metab 2010;95:4592-9
  • Fadul CE, Kominsky AL, Meyer LP, et al. Long-term response of pituitary carcinoma to temozolomide. Report of two cases. J Neurosurg 2006;105:621-6
  • Bush ZM, Longtine JA, Cunningham T, et al. Temozolomide treatment for aggressive pituitary tumors: correlation of clinical outcome with O(6)-methylguanine methyltransferase (MGMT) promoter methylation and expression. J Clin Endocrinol Metab 2010;95:E280-90
  • Thearle MS, Freda PU, Bruce JN, et al. Temozolomide (Temodar®) and capecitabine (Xeloda®) treatment of an aggressive corticotroph pituitary tumor. Pituitary 2011;14:418-24
  • Mohammed S, Kovacs K, Mason W, et al. Use of temozolomide in aggressive pituitary tumors: case report. Neurosurgery 2009;64:E773-4
  • Hagen C, Schroeder HD, Hansen S, Andersen M. Temozolomide treatment of a pituitary carcinoma and two pituitary macroadenomas resistant to conventional therapy. Eur J Endocrinol 2009;161:631-7
  • Morin E, Berthelet F, Weisnagel J, et al. Failure of temozolomide and conventional doses of pegvisomant to attain biochemical control in a severe case of acromegaly. Pituitary 2012;15:97-100
  • Barbieri F, Bajetto A, Pattarozzi A, et al. Peptide receptor targeting in cancer: the somatostatin paradigm. Int J Pept 2013;2013:926295
  • Theodoropoulou M, Stalla GK. Somatostatin receptors: from signaling to clinical practice. Front Neuroendocrinol 2013;34:228-52
  • Mayr BM, Buslei R, Theodoropoulou M, et al. Molecular and functional properties of densely and sparsely granulated GH-producing pituitary adenomas. Eur J Endocrinol 2013;169(4):391-400
  • Gadelha MR, Kasuki L, Korbonits M. Novel pathway for somatostatin analogs in patients with acromegaly. Trends Endocrinol Metab 2013;24:238-46
  • Horvath E, Kovacs K. Pathology of acromegaly. Neuroendocrinology 2006;83:161-5
  • Larkin SJ, Reddy RG, Karavitaki N, et al. Granulation pattern, but not GSP or GHR mutation, is associated with clinical characteristics in somatostatin-naive patients with somatotroph adenomas. Eur J Endocrinol 2013;168(4):491-9
  • Reubi J, Waser B, Schaer J-C, Laissue J. Somatostatin receptor sst1-sst5 expression in normal and neoplastic human tissues using receptor autoradiography with subtype-selective ligands. Eur J Nucl Med Mol Imaging 2001;28:836-46

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.