Skip to main content
Erschienen in: Endocrine 3/2018

31.07.2018 | Original Article

Effect of combined treatment with a pan-PI3K inhibitor or an isoform-specific PI3K inhibitor and everolimus on cell proliferation in GH-secreting pituitary tumour in an experimental setting

verfasst von: Claudia Pivonello, Roberta Patalano, Domenico Solari, Renata S. Auriemma, Federico Frio, Francesca Vitulli, Ludovica F. S. Grasso, Marialuisa Di Cera, Maria Cristina De Martino, Luigi M. Cavallo, Paolo Cappabianca, Annamaria Colao, Rosario Pivonello

Erschienen in: Endocrine | Ausgabe 3/2018

Einloggen, um Zugang zu erhalten

Abstract

Purpose

PI3K/Akt/mTOR pathway activation is common in GH-secreting pituitary tumours, and a target for treatment with mTOR inhibitors, including everolimus (EVE). The current study aimed to evaluate the efficacy of two PI3K inhibitors (PI3Ki), NVP-BKM120 and NVP-BYL719, alone and in combination with EVE in rat GH-secreting pituitary tumour cell line (GH3) and human GH-secreting pituitary tumour cell cultures.

Methods

In GH3 cell line and in six GH-secreting tumour cell cultures, the effects of PI3Ki and EVE, as single agents and in combination, were tested on cell viability and colony survival, by MTT and clonogenic assay, respectively, whereas western blot was performed to evaluate the underlying intracellular signalling pathways.

Results

PI3Ki and EVE showed a dose-dependent inhibition of cell viability in GH3 cell line, with PI3Ki displaying a synergistic effect when combined with EVE. PI3Ki and EVE inhibited colony survival in GH3 cell line with no further improvement in combination. In GH-secreting pituitary tumour cell cultures PI3Ki are effective in inhibiting cell viability increasing the slight and non significant inhibition induced by EVE as single agent, generally showing a synergistic effect. Despite in both GH3 cell line and GH-secreting pituitary tumour cell cultures combination of PI3Ki enhanced EVE effect, the study of intracellular signalling pathways revealed a different regulation of PI3K/Akt/mTOR and MAPK between the two models.

Conclusions

The results of the current study demonstrated that PI3Ki, especially in combination with EVE, are effective in inhibiting cell proliferation, therefore representing a promising therapeutic tool for the treatment of aggressive GH-secreting pituitary tumours, not responsive to standard medical therapies.
Literatur
1.
Zurück zum Zitat S. Melmed, A. Colao, A. Barkan, M. Molitch, A.B. Grossman, D. Kleinberg et al., Guidelines for acromegaly management: an update. J. Clin. Endocrinol. Metab. 94, 1509–17 (2009).CrossRef S. Melmed, A. Colao, A. Barkan, M. Molitch, A.B. Grossman, D. Kleinberg et al., Guidelines for acromegaly management: an update. J. Clin. Endocrinol. Metab. 94, 1509–17 (2009).CrossRef
2.
Zurück zum Zitat R.N. Clayton, Cardiovascular function in acromegaly. Endocr. Rev. 24, 272–7 (2003)CrossRef R.N. Clayton, Cardiovascular function in acromegaly. Endocr. Rev. 24, 272–7 (2003)CrossRef
3.
Zurück zum Zitat R. Pivonello, R.S. Auriemma, L.F. Grasso, C. Pivonello, C. Simeoli, R. Patalano et al., Complications of acromegaly: cardiovascular, respiratory and metabolic comorbidities. Pituitary 20, 46–62 (2017).CrossRef R. Pivonello, R.S. Auriemma, L.F. Grasso, C. Pivonello, C. Simeoli, R. Patalano et al., Complications of acromegaly: cardiovascular, respiratory and metabolic comorbidities. Pituitary 20, 46–62 (2017).CrossRef
4.
Zurück zum Zitat A. Colao, D. Ferone, P. Marzullo, G. Lombardi, Systemic complications of acromegaly: epidemiology, pathogenesis, and management. Endocr. Rev. 25, 102–52 (2004)CrossRef A. Colao, D. Ferone, P. Marzullo, G. Lombardi, Systemic complications of acromegaly: epidemiology, pathogenesis, and management. Endocr. Rev. 25, 102–52 (2004)CrossRef
5.
Zurück zum Zitat L. Katznelson, E.R. Laws Jr., S. Melmed, M.E. Molitch, M.H. Murad, A. Utz et al., Acromegaly: an endocrine society clinical practice guideline. J. Clin. Endocrinol. Metab. 99, 3933–51 (2014)CrossRef L. Katznelson, E.R. Laws Jr., S. Melmed, M.E. Molitch, M.H. Murad, A. Utz et al., Acromegaly: an endocrine society clinical practice guideline. J. Clin. Endocrinol. Metab. 99, 3933–51 (2014)CrossRef
6.
Zurück zum Zitat A. Giustina, A. Barkan, F.F. Casanueva, F. Cavagnini, L. Frohman, K. Ho et al., Criteria for cure of acromegaly: a consensus statement. J. Clin. Endocrinol. Metab. 85, 526–9 (2000)PubMed A. Giustina, A. Barkan, F.F. Casanueva, F. Cavagnini, L. Frohman, K. Ho et al., Criteria for cure of acromegaly: a consensus statement. J. Clin. Endocrinol. Metab. 85, 526–9 (2000)PubMed
7.
Zurück zum Zitat E.R. Laws, G. Lanzino. Transsphenoidal Surgery. (Saunders - Elsevier, Philadelphia, 2010) E.R. Laws, G. Lanzino. Transsphenoidal Surgery. (Saunders - Elsevier, Philadelphia, 2010)
8.
Zurück zum Zitat D. Solari, L.M. Cavallo, P. Cappabianca, Surgical approach to pituitary tumors. Handb. Clin. Neurol. 124, 291–301 (2014)CrossRef D. Solari, L.M. Cavallo, P. Cappabianca, Surgical approach to pituitary tumors. Handb. Clin. Neurol. 124, 291–301 (2014)CrossRef
9.
Zurück zum Zitat A. Colao, R. Pivonello, C. Di Somma, S. Savastano, L.F. Grasso, G. Lombardi, Medical therapy of pituitary adenomas: effects on tumor shrinkage. Rev. Endocr. Metab. Disord. 10, 111–23 (2009)CrossRef A. Colao, R. Pivonello, C. Di Somma, S. Savastano, L.F. Grasso, G. Lombardi, Medical therapy of pituitary adenomas: effects on tumor shrinkage. Rev. Endocr. Metab. Disord. 10, 111–23 (2009)CrossRef
10.
Zurück zum Zitat A. Giustina, M.R. Ambrosio, P. Beck Peccoz, F. Bogazzi, S. Cannavo, L. De Marinis et al., Use of Pegvisomant in acromegaly. An Italian Society of Endocrinology guideline. J. Endocrinol. Invest. 37, 1017–30 (2014)CrossRef A. Giustina, M.R. Ambrosio, P. Beck Peccoz, F. Bogazzi, S. Cannavo, L. De Marinis et al., Use of Pegvisomant in acromegaly. An Italian Society of Endocrinology guideline. J. Endocrinol. Invest. 37, 1017–30 (2014)CrossRef
11.
Zurück zum Zitat A. Colao, R.S. Auriemma, G. Lombardi, R. Pivonello, Resistance to somatostatin analogs in acromegaly. Endocr. Rev. 32, 247–71 (2011)CrossRef A. Colao, R.S. Auriemma, G. Lombardi, R. Pivonello, Resistance to somatostatin analogs in acromegaly. Endocr. Rev. 32, 247–71 (2011)CrossRef
12.
Zurück zum Zitat M.R. Gadelha, L.E. Wildemberg, M.D. Bronstein, F. Gatto, D. Ferone, Somatostatin receptor ligands in the treatment of acromegaly. Pituitary 20, 100–8 (2017)CrossRef M.R. Gadelha, L.E. Wildemberg, M.D. Bronstein, F. Gatto, D. Ferone, Somatostatin receptor ligands in the treatment of acromegaly. Pituitary 20, 100–8 (2017)CrossRef
13.
Zurück zum Zitat C. Beauregard, U. Truong, J. Hardy, O. Serri, Long-term outcome and mortality after transsphenoidal adenomectomy for acromegaly. Clin. Endocrinol. 58, 86–91 (2003)CrossRef C. Beauregard, U. Truong, J. Hardy, O. Serri, Long-term outcome and mortality after transsphenoidal adenomectomy for acromegaly. Clin. Endocrinol. 58, 86–91 (2003)CrossRef
14.
Zurück zum Zitat G. Minniti, M.L. Jaffrain-Rea, V. Esposito, A. Santoro, G. Tamburrano, G. Cantore, Evolving criteria for post-operative biochemical remission of acromegaly: can we achieve a definitive cure? An audit of surgical results on a large series and a review of the literature. Endocr. Relat. Cancer 10, 611–9 (2003)CrossRef G. Minniti, M.L. Jaffrain-Rea, V. Esposito, A. Santoro, G. Tamburrano, G. Cantore, Evolving criteria for post-operative biochemical remission of acromegaly: can we achieve a definitive cure? An audit of surgical results on a large series and a review of the literature. Endocr. Relat. Cancer 10, 611–9 (2003)CrossRef
15.
Zurück zum Zitat F. Di Nicolantonio, S. Arena, J. Tabernero, S. Grosso, F. Molinari, T. Macarulla et al., Deregulation of the PI3K and KRAS signaling pathways in human cancer cells determines their response to everolimus. J. Clin. Invest. 120, 2858–66 (2010)CrossRef F. Di Nicolantonio, S. Arena, J. Tabernero, S. Grosso, F. Molinari, T. Macarulla et al., Deregulation of the PI3K and KRAS signaling pathways in human cancer cells determines their response to everolimus. J. Clin. Invest. 120, 2858–66 (2010)CrossRef
16.
Zurück zum Zitat Y. Lin, X. Jiang, Y. Shen, M. Li, H. Ma, M. Xing et al., Frequent mutations and amplifications of the PIK3CA gene in pituitary tumors. Endocr. Relat. Cancer 16, 301–10 (2009)CrossRef Y. Lin, X. Jiang, Y. Shen, M. Li, H. Ma, M. Xing et al., Frequent mutations and amplifications of the PIK3CA gene in pituitary tumors. Endocr. Relat. Cancer 16, 301–10 (2009)CrossRef
17.
Zurück zum Zitat K.E. O’Reilly, F. Rojo, Q.B. She, D. Solit, G.B. Mills, D. Smith et al., mTOR inhibition induces upstream receptor tyrosine kinase signaling and activates Akt. Cancer Res. 66, 1500–8 (2006)CrossRef K.E. O’Reilly, F. Rojo, Q.B. She, D. Solit, G.B. Mills, D. Smith et al., mTOR inhibition induces upstream receptor tyrosine kinase signaling and activates Akt. Cancer Res. 66, 1500–8 (2006)CrossRef
18.
Zurück zum Zitat I. Vivanco, C.L. Sawyers, The phosphatidylinositol 3-Kinase AKT pathway in human cancer. Nat. Rev. Cancer 2, 489–501 (2002)CrossRef I. Vivanco, C.L. Sawyers, The phosphatidylinositol 3-Kinase AKT pathway in human cancer. Nat. Rev. Cancer 2, 489–501 (2002)CrossRef
19.
Zurück zum Zitat D.A. Altomare, J.R. Testa, Perturbations of the AKT signaling pathway in human cancer. Oncogene 24, 7455–64 (2005)CrossRef D.A. Altomare, J.R. Testa, Perturbations of the AKT signaling pathway in human cancer. Oncogene 24, 7455–64 (2005)CrossRef
20.
Zurück zum Zitat K.H. Khan, T.A. Yap, L. Yan, D. Cunningham, Targeting the PI3K-AKT-mTOR signaling network in cancer. Chin. J. Cancer 32, 253–65 (2013)CrossRef K.H. Khan, T.A. Yap, L. Yan, D. Cunningham, Targeting the PI3K-AKT-mTOR signaling network in cancer. Chin. J. Cancer 32, 253–65 (2013)CrossRef
21.
Zurück zum Zitat C. Pivonello, M. Negri, M.C. De Martino, M. Napolitano, C. de Angelis, D.P. Provvisiero et al., The dual targeting of insulin and insulin-like growth factor 1 receptor enhances the mTOR inhibitor-mediated antitumor efficacy in hepatocellular carcinoma. Oncotarget 7, 9718–31 (2016)CrossRef C. Pivonello, M. Negri, M.C. De Martino, M. Napolitano, C. de Angelis, D.P. Provvisiero et al., The dual targeting of insulin and insulin-like growth factor 1 receptor enhances the mTOR inhibitor-mediated antitumor efficacy in hepatocellular carcinoma. Oncotarget 7, 9718–31 (2016)CrossRef
22.
Zurück zum Zitat L. Zhao, P.K. Vogt, Class I PI3K in oncogenic cellular transformation. Oncogene 27, 5486–96 (2008)CrossRef L. Zhao, P.K. Vogt, Class I PI3K in oncogenic cellular transformation. Oncogene 27, 5486–96 (2008)CrossRef
23.
Zurück zum Zitat T.L. Yuan, L.C. Cantley, PI3K pathway alterations in cancer: variations on a theme. Oncogene 27, 5497–510 (2008)CrossRef T.L. Yuan, L.C. Cantley, PI3K pathway alterations in cancer: variations on a theme. Oncogene 27, 5497–510 (2008)CrossRef
24.
Zurück zum Zitat A. Carracedo, L. Ma, J. Teruya-Feldstein, F. Rojo, L. Salmena, A. Alimonti et al., Inhibition of mTORC1 leads to MAPK pathway activation through a PI3K-dependent feedback loop in human cancer. J. Clin. Investig. 118, 3065–74 (2008)PubMed A. Carracedo, L. Ma, J. Teruya-Feldstein, F. Rojo, L. Salmena, A. Alimonti et al., Inhibition of mTORC1 leads to MAPK pathway activation through a PI3K-dependent feedback loop in human cancer. J. Clin. Investig. 118, 3065–74 (2008)PubMed
25.
Zurück zum Zitat E. Monsalves, K. Juraschka, T. Tateno, S. Agnihotri, S.L. Asa, S. Ezzat et al., The PI3K/AKT/mTOR pathway in the pathophysiology and treatment of pituitary adenomas. Endocr. Relat. Cancer 21, R331–44 (2014)CrossRef E. Monsalves, K. Juraschka, T. Tateno, S. Agnihotri, S.L. Asa, S. Ezzat et al., The PI3K/AKT/mTOR pathway in the pathophysiology and treatment of pituitary adenomas. Endocr. Relat. Cancer 21, R331–44 (2014)CrossRef
26.
Zurück zum Zitat S. Jean, A.A. Kiger, Classes of phosphoinositide 3-kinases at a glance. J. Cell. Sci. 127, 923–8 (2014)CrossRef S. Jean, A.A. Kiger, Classes of phosphoinositide 3-kinases at a glance. J. Cell. Sci. 127, 923–8 (2014)CrossRef
27.
Zurück zum Zitat M. Chanal, P. Chevallier, V. Raverot, G. Fonteneau, K. Lucia, J.L. Monteserin Garcia et al., Differential effects of PI3K and dual PI3K/mTOR inhibition in rat prolactin-secreting pituitary tumors. Mol. Cancer Ther. 15, 1261–70 (2016)CrossRef M. Chanal, P. Chevallier, V. Raverot, G. Fonteneau, K. Lucia, J.L. Monteserin Garcia et al., Differential effects of PI3K and dual PI3K/mTOR inhibition in rat prolactin-secreting pituitary tumors. Mol. Cancer Ther. 15, 1261–70 (2016)CrossRef
28.
Zurück zum Zitat C.B. Murat, P.B. Braga, M.A. Fortes, M.D. Bronstein, M.L. Correa-Giannella, R.R. Giorgi, Mutation and genomic amplification of the PIK3CA proto-oncogene in pituitary adenomas. Braz. J. Med. Biol. Res. 45, 851–5 (2012)CrossRef C.B. Murat, P.B. Braga, M.A. Fortes, M.D. Bronstein, M.L. Correa-Giannella, R.R. Giorgi, Mutation and genomic amplification of the PIK3CA proto-oncogene in pituitary adenomas. Braz. J. Med. Biol. Res. 45, 851–5 (2012)CrossRef
29.
Zurück zum Zitat M. Cakir, A.B. Grossman, Targeting MAPK (Ras/ERK) and PI3K/Akt pathways in pituitary tumorigenesis. Expert. Opin. Ther. Targets 13, 1121–34 (2009)CrossRef M. Cakir, A.B. Grossman, Targeting MAPK (Ras/ERK) and PI3K/Akt pathways in pituitary tumorigenesis. Expert. Opin. Ther. Targets 13, 1121–34 (2009)CrossRef
30.
Zurück zum Zitat D. Dworakowska, E. Wlodek, C.A. Leontiou, S. Igreja, M. Cakir, M. Teng et al., Activation of RAF/MEK/ERK and PI3K/AKT/mTOR pathways in pituitary adenomas and their effects on downstream effectors. Endocr. Relat. Cancer 16, 1329–38 (2009)CrossRef D. Dworakowska, E. Wlodek, C.A. Leontiou, S. Igreja, M. Cakir, M. Teng et al., Activation of RAF/MEK/ERK and PI3K/AKT/mTOR pathways in pituitary adenomas and their effects on downstream effectors. Endocr. Relat. Cancer 16, 1329–38 (2009)CrossRef
31.
Zurück zum Zitat M. Musat, M. Korbonits, B. Kola, N. Borboli, M.R. Hanson, A.M. Nanzer et al., Enhanced protein kinase B/Akt signalling in pituitary tumours. Endocr. Relat. Cancer 12, 423–33 (2005)CrossRef M. Musat, M. Korbonits, B. Kola, N. Borboli, M.R. Hanson, A.M. Nanzer et al., Enhanced protein kinase B/Akt signalling in pituitary tumours. Endocr. Relat. Cancer 12, 423–33 (2005)CrossRef
32.
Zurück zum Zitat B. Svejda, M. Kidd, A. Kazberouk, B. Lawrence, R. Pfragner, I.M. Modlin, Limitations in small intestinal neuroendocrine tumor therapy by mTor kinase inhibition reflect growth factor-mediated PI3K feedback loop activation via ERK1/2 and AKT. Cancer 117, 4141–54 (2011)CrossRef B. Svejda, M. Kidd, A. Kazberouk, B. Lawrence, R. Pfragner, I.M. Modlin, Limitations in small intestinal neuroendocrine tumor therapy by mTor kinase inhibition reflect growth factor-mediated PI3K feedback loop activation via ERK1/2 and AKT. Cancer 117, 4141–54 (2011)CrossRef
33.
Zurück zum Zitat D.A. Cantrell, Phosphoinositide 3-kinase signalling pathways. J. Cell. Sci. 114, 1439–45 (2001)PubMed D.A. Cantrell, Phosphoinositide 3-kinase signalling pathways. J. Cell. Sci. 114, 1439–45 (2001)PubMed
34.
Zurück zum Zitat B.T. Hennessy, D.L. Smith, P.T. Ram, Y. Lu, G.B. Mills, Exploiting the PI3K/AKT pathway for cancer drug discovery. Nat. Rev. Drug. Discov. 4, 988–1004 (2005)CrossRef B.T. Hennessy, D.L. Smith, P.T. Ram, Y. Lu, G.B. Mills, Exploiting the PI3K/AKT pathway for cancer drug discovery. Nat. Rev. Drug. Discov. 4, 988–1004 (2005)CrossRef
35.
Zurück zum Zitat X. Bai, Y. Jiang, Key factors in mTOR regulation. Cell. Mol. Life Sci. 67, 239–53 (2010)CrossRef X. Bai, Y. Jiang, Key factors in mTOR regulation. Cell. Mol. Life Sci. 67, 239–53 (2010)CrossRef
36.
Zurück zum Zitat R. Chen, J. Duan, L. Li, Q. Ma, Q. Sun, J. Ma et al., mTOR promotes pituitary tumor development through activation of PTTG1. Oncogene 36, 979–88 (2017)CrossRef R. Chen, J. Duan, L. Li, Q. Ma, Q. Sun, J. Ma et al., mTOR promotes pituitary tumor development through activation of PTTG1. Oncogene 36, 979–88 (2017)CrossRef
37.
Zurück zum Zitat S.M. Maira, S. Pecchi, A. Huang, M. Burger, M. Knapp, D. Sterker et al., Identification and characterization of NVP-BKM120, an orally available pan-class I PI3-kinase inhibitor. Mol. Cancer Ther. 11, 317–28 (2012)CrossRef S.M. Maira, S. Pecchi, A. Huang, M. Burger, M. Knapp, D. Sterker et al., Identification and characterization of NVP-BKM120, an orally available pan-class I PI3-kinase inhibitor. Mol. Cancer Ther. 11, 317–28 (2012)CrossRef
38.
Zurück zum Zitat P. Furet, V. Guagnano, R.A. Fairhurst, P. Imbach-Weese, I. Bruce, M. Knapp et al., Discovery of NVP-BYL719 a potent and selective phosphatidylinositol-3 kinase alpha inhibitor selected for clinical evaluation. Bioorg. Med. Chem. Lett. 23, 3741–8 (2013)CrossRef P. Furet, V. Guagnano, R.A. Fairhurst, P. Imbach-Weese, I. Bruce, M. Knapp et al., Discovery of NVP-BYL719 a potent and selective phosphatidylinositol-3 kinase alpha inhibitor selected for clinical evaluation. Bioorg. Med. Chem. Lett. 23, 3741–8 (2013)CrossRef
39.
Zurück zum Zitat R.J. Motzer, B. Escudier, S. Oudard, T.E. Hutson, C. Porta, S. Bracarda et al., Efficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomised, placebo-controlled phase III trial. Lancet 372, 449–56 (2008)CrossRef R.J. Motzer, B. Escudier, S. Oudard, T.E. Hutson, C. Porta, S. Bracarda et al., Efficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomised, placebo-controlled phase III trial. Lancet 372, 449–56 (2008)CrossRef
40.
Zurück zum Zitat A.X. Zhu, M. Kudo, E. Assenat, S. Cattan, Y.K. Kang, H.Y. Lim et al., Effect of everolimus on survival in advanced hepatocellular carcinoma after failure of sorafenib: the EVOLVE-1 randomized clinical trial. JAMA 312, 57–67 (2014)CrossRef A.X. Zhu, M. Kudo, E. Assenat, S. Cattan, Y.K. Kang, H.Y. Lim et al., Effect of everolimus on survival in advanced hepatocellular carcinoma after failure of sorafenib: the EVOLVE-1 randomized clinical trial. JAMA 312, 57–67 (2014)CrossRef
41.
Zurück zum Zitat N. Wagle, B.C. Grabiner, E.M. Van Allen, A. Amin-Mansour, A. Taylor-Weiner, M. Rosenberg et al., Response and acquired resistance to everolimus in anaplastic thyroid cancer. N. Engl. J. Med. 371, 1426–33 (2014)CrossRef N. Wagle, B.C. Grabiner, E.M. Van Allen, A. Amin-Mansour, A. Taylor-Weiner, M. Rosenberg et al., Response and acquired resistance to everolimus in anaplastic thyroid cancer. N. Engl. J. Med. 371, 1426–33 (2014)CrossRef
42.
Zurück zum Zitat M.E. Pavel, J.D. Hainsworth, E. Baudin, M. Peeters, D. Horsch, R.E. Winkler et al., Everolimus plus octreotide long-acting repeatable for the treatment of advanced neuroendocrine tumours associated with carcinoid syndrome (RADIANT-2): a randomised, placebo-controlled, phase 3 study. Lancet 378, 2005–12 (2011)CrossRef M.E. Pavel, J.D. Hainsworth, E. Baudin, M. Peeters, D. Horsch, R.E. Winkler et al., Everolimus plus octreotide long-acting repeatable for the treatment of advanced neuroendocrine tumours associated with carcinoid syndrome (RADIANT-2): a randomised, placebo-controlled, phase 3 study. Lancet 378, 2005–12 (2011)CrossRef
43.
Zurück zum Zitat J.C. Yao, M.H. Shah, T. Ito, C.L. Bohas, E.M. Wolin, E. Van Cutsem et al., Everolimus for advanced pancreatic neuroendocrine tumors. N. Engl. J. Med. 364, 514–23 (2011)CrossRef J.C. Yao, M.H. Shah, T. Ito, C.L. Bohas, E.M. Wolin, E. Van Cutsem et al., Everolimus for advanced pancreatic neuroendocrine tumors. N. Engl. J. Med. 364, 514–23 (2011)CrossRef
44.
Zurück zum Zitat A. Gorshtein, H. Rubinfeld, E. Kendler, M. Theodoropoulou, V. Cerovac, G.K. Stalla et al., Mammalian target of rapamycin inhibitors rapamycin and RAD001 (everolimus) induce anti-proliferative effects in GH-secreting pituitary tumor cells in vitro. Endocr. Relat. Cancer 16, 1017–27 (2009)CrossRef A. Gorshtein, H. Rubinfeld, E. Kendler, M. Theodoropoulou, V. Cerovac, G.K. Stalla et al., Mammalian target of rapamycin inhibitors rapamycin and RAD001 (everolimus) induce anti-proliferative effects in GH-secreting pituitary tumor cells in vitro. Endocr. Relat. Cancer 16, 1017–27 (2009)CrossRef
45.
Zurück zum Zitat S. Sukumari-Ramesh, N. Singh, K.M. Dhandapani, J.R. Vender, mTOR inhibition reduces cellular proliferation and sensitizes pituitary adenoma cells to ionizing radiation. Surg. Neurol. Int. 2, 22 (2011)CrossRef S. Sukumari-Ramesh, N. Singh, K.M. Dhandapani, J.R. Vender, mTOR inhibition reduces cellular proliferation and sensitizes pituitary adenoma cells to ionizing radiation. Surg. Neurol. Int. 2, 22 (2011)CrossRef
46.
Zurück zum Zitat E. Rozengurt, H.P. Soares, J. Sinnet-Smith, Suppression of feedback loops mediated by PI3K/mTOR induces multiple overactivation of compensatory pathways: an unintended consequence leading to drug resistance. Mol. Cancer Ther. 13, 2477–88 (2014)CrossRef E. Rozengurt, H.P. Soares, J. Sinnet-Smith, Suppression of feedback loops mediated by PI3K/mTOR induces multiple overactivation of compensatory pathways: an unintended consequence leading to drug resistance. Mol. Cancer Ther. 13, 2477–88 (2014)CrossRef
47.
Zurück zum Zitat G. Brabant, A. von zur Muhlen, C. Wuster, M.B. Ranke, J. Kratzsch, W. Kiess et al., Serum insulin-like growth factor I reference values for an automated chemiluminescence immunoassay system: results from a multicenter study. Horm. Res. 60, 53–60 (2003)PubMed G. Brabant, A. von zur Muhlen, C. Wuster, M.B. Ranke, J. Kratzsch, W. Kiess et al., Serum insulin-like growth factor I reference values for an automated chemiluminescence immunoassay system: results from a multicenter study. Horm. Res. 60, 53–60 (2003)PubMed
48.
Zurück zum Zitat C. Pivonello, P. Rousaki, M. Negri, M. Sarnataro, M. Napolitano, F.Z. Marino et al., Effects of the single and combined treatment with dopamine agonist, somatostatin analog and mTOR inhibitors in a human lung carcinoid cell line: an in vitro study. Endocrine 56, 603–20 (2017)CrossRef C. Pivonello, P. Rousaki, M. Negri, M. Sarnataro, M. Napolitano, F.Z. Marino et al., Effects of the single and combined treatment with dopamine agonist, somatostatin analog and mTOR inhibitors in a human lung carcinoid cell line: an in vitro study. Endocrine 56, 603–20 (2017)CrossRef
49.
Zurück zum Zitat C. Desbois-Mouthon, A. Baron, M.J. Blivet-Van Eggelpoel, L. Fartoux, C. Venot, F. Bladt et al., Insulin-like growth factor-1 receptor inhibition induces a resistance mechanism via the epidermal growth factor receptor/HER3/AKT signaling pathway: rational basis for cotargeting insulin-like growth factor-1 receptor and epidermal growth factor receptor in hepatocellular carcinoma. Clin. Cancer Res. 15, 5445–56 (2009)CrossRef C. Desbois-Mouthon, A. Baron, M.J. Blivet-Van Eggelpoel, L. Fartoux, C. Venot, F. Bladt et al., Insulin-like growth factor-1 receptor inhibition induces a resistance mechanism via the epidermal growth factor receptor/HER3/AKT signaling pathway: rational basis for cotargeting insulin-like growth factor-1 receptor and epidermal growth factor receptor in hepatocellular carcinoma. Clin. Cancer Res. 15, 5445–56 (2009)CrossRef
50.
Zurück zum Zitat D.A. Donoho, N. Bose, G. Zada, J.D. Carmichael, Management of aggressive growth hormone secreting pituitary adenomas. Pituitary 20, 169–78 (2017)CrossRef D.A. Donoho, N. Bose, G. Zada, J.D. Carmichael, Management of aggressive growth hormone secreting pituitary adenomas. Pituitary 20, 169–78 (2017)CrossRef
51.
Zurück zum Zitat E.A. Sajjad, G. Zielinski, M. Maksymowicz, L. Hutnik, T. Bednarczuk, P. Wlodarski, mTOR is frequently active in GH-secreting pituitary adenomas without influencing their morphopathological features. Endocr. Pathol. 24, 11–9 (2013)CrossRef E.A. Sajjad, G. Zielinski, M. Maksymowicz, L. Hutnik, T. Bednarczuk, P. Wlodarski, mTOR is frequently active in GH-secreting pituitary adenomas without influencing their morphopathological features. Endocr. Pathol. 24, 11–9 (2013)CrossRef
52.
Zurück zum Zitat M. Lee, T. Wiedemann, C. Gross, I. Leinhauser, F. Roncaroli, R. Braren et al., Targeting PI3K/mTOR signaling displays potent antitumor efficacy against nonfunctioning pituitary adenomas. Clin. Cancer Res. 21, 3204–15 (2015)CrossRef M. Lee, T. Wiedemann, C. Gross, I. Leinhauser, F. Roncaroli, R. Braren et al., Targeting PI3K/mTOR signaling displays potent antitumor efficacy against nonfunctioning pituitary adenomas. Clin. Cancer Res. 21, 3204–15 (2015)CrossRef
53.
Zurück zum Zitat M. Lee, M. Theodoropoulou, J. Graw, F. Roncaroli, M.C. Zatelli, N.S. Pellegata, Levels of p27 sensitize to dual PI3K/mTOR inhibition. Mol. Cancer Ther. 10, 1450–9 (2011)CrossRef M. Lee, M. Theodoropoulou, J. Graw, F. Roncaroli, M.C. Zatelli, N.S. Pellegata, Levels of p27 sensitize to dual PI3K/mTOR inhibition. Mol. Cancer Ther. 10, 1450–9 (2011)CrossRef
54.
Zurück zum Zitat H. Rubinfeld, I. Shimon, PI3K/Akt/mTOR and Raf/MEK/ERK signaling pathways perturbations in non-functioning pituitary adenomas. Endocrine 42, 285–91 (2012)CrossRef H. Rubinfeld, I. Shimon, PI3K/Akt/mTOR and Raf/MEK/ERK signaling pathways perturbations in non-functioning pituitary adenomas. Endocrine 42, 285–91 (2012)CrossRef
55.
Zurück zum Zitat J.D. Valentino, J. Li, Y.Y. Zaytseva, W.C. Mustain, V.A. Elliott, J.T. Kim et al., Cotargeting the PI3K and RAS pathways for the treatment of neuroendocrine tumors. Clin. Cancer Res. 20, 1212–22 (2014)CrossRef J.D. Valentino, J. Li, Y.Y. Zaytseva, W.C. Mustain, V.A. Elliott, J.T. Kim et al., Cotargeting the PI3K and RAS pathways for the treatment of neuroendocrine tumors. Clin. Cancer Res. 20, 1212–22 (2014)CrossRef
56.
Zurück zum Zitat S. Nolting, J. Rentsch, H. Freitag, K. Detjen, F. Briest, M. Mobs et al., The selective PI3Kalpha inhibitor BYL719 as a novel therapeutic option for neuroendocrine tumors: results from multiple cell line models. PLoS ONE 12, e0182852 (2017)CrossRef S. Nolting, J. Rentsch, H. Freitag, K. Detjen, F. Briest, M. Mobs et al., The selective PI3Kalpha inhibitor BYL719 as a novel therapeutic option for neuroendocrine tumors: results from multiple cell line models. PLoS ONE 12, e0182852 (2017)CrossRef
57.
Zurück zum Zitat Y. Hu, R. Guo, J. Wei, Y. Zhou, W. Ji, J. Liu et al., Effects of PI3K inhibitor NVP-BKM120 on overcoming drug resistance and eliminating cancer stem cells in human breast cancer cells. Cell Death Dis. 6, e2020 (2015)CrossRef Y. Hu, R. Guo, J. Wei, Y. Zhou, W. Ji, J. Liu et al., Effects of PI3K inhibitor NVP-BKM120 on overcoming drug resistance and eliminating cancer stem cells in human breast cancer cells. Cell Death Dis. 6, e2020 (2015)CrossRef
58.
Zurück zum Zitat Y. Zheng, J. Yang, J. Qian, L. Zhang, Y. Lu, H. Li et al., Novel phosphatidylinositol 3-kinase inhibitor NVP-BKM120 induces apoptosis in myeloma cells and shows synergistic anti-myeloma activity with dexamethasone. J. Mol. Med. 90, 695–706 (2012)CrossRef Y. Zheng, J. Yang, J. Qian, L. Zhang, Y. Lu, H. Li et al., Novel phosphatidylinositol 3-kinase inhibitor NVP-BKM120 induces apoptosis in myeloma cells and shows synergistic anti-myeloma activity with dexamethasone. J. Mol. Med. 90, 695–706 (2012)CrossRef
59.
Zurück zum Zitat E. Musi, G. Ambrosini, E. de Stanchina, G.K. Schwartz, The phosphoinositide 3-kinase alpha selective inhibitor BYL719 enhances the effect of the protein kinase C inhibitor AEB071 in GNAQ/GNA11-mutant uveal melanoma cells. Mol. Cancer Ther. 13, 1044–53 (2014)CrossRef E. Musi, G. Ambrosini, E. de Stanchina, G.K. Schwartz, The phosphoinositide 3-kinase alpha selective inhibitor BYL719 enhances the effect of the protein kinase C inhibitor AEB071 in GNAQ/GNA11-mutant uveal melanoma cells. Mol. Cancer Ther. 13, 1044–53 (2014)CrossRef
60.
Zurück zum Zitat H. Ren, H. Guo, A. Thakur, S. Zhang, T. Wang, Y. Liang et al., Blockade efficacy of MEK/ERK-dependent autophagy enhances PI3K/Akt inhibitor NVP-BKM120’s therapeutic effectiveness in lung cancer cells. Oncotarget 7, 67277–87 (2016)PubMedPubMedCentral H. Ren, H. Guo, A. Thakur, S. Zhang, T. Wang, Y. Liang et al., Blockade efficacy of MEK/ERK-dependent autophagy enhances PI3K/Akt inhibitor NVP-BKM120’s therapeutic effectiveness in lung cancer cells. Oncotarget 7, 67277–87 (2016)PubMedPubMedCentral
61.
Zurück zum Zitat J.C. Bendell, J. Rodon, H.A. Burris, M. de Jonge, J. Verweij, D. Birle et al., Phase I, dose-escalation study of BKM120, an oral pan-Class I PI3K inhibitor, in patients with advanced solid tumors. J. Clin. Oncol. 30, 282–90 (2012)CrossRef J.C. Bendell, J. Rodon, H.A. Burris, M. de Jonge, J. Verweij, D. Birle et al., Phase I, dose-escalation study of BKM120, an oral pan-Class I PI3K inhibitor, in patients with advanced solid tumors. J. Clin. Oncol. 30, 282–90 (2012)CrossRef
62.
Zurück zum Zitat S.S. De Buck, A. Jakab, M. Boehm, D. Bootle, D. Juric, C. Quadt et al., Population pharmacokinetics and pharmacodynamics of BYL719, a phosphoinositide 3-kinase antagonist, in adult patients with advanced solid malignancies. Br. J. Clin. Pharmacol. 78, 543–55 (2014)CrossRef S.S. De Buck, A. Jakab, M. Boehm, D. Bootle, D. Juric, C. Quadt et al., Population pharmacokinetics and pharmacodynamics of BYL719, a phosphoinositide 3-kinase antagonist, in adult patients with advanced solid malignancies. Br. J. Clin. Pharmacol. 78, 543–55 (2014)CrossRef
63.
Zurück zum Zitat M.C. Zatelli, M. Minoia, C. Filieri, F. Tagliati, M. Buratto, M.R. Ambrosio et al., Effect of everolimus on cell viability in nonfunctioning pituitary adenomas. J. Clin. Endocrinol. Metab. 95, 968–76 (2010)CrossRef M.C. Zatelli, M. Minoia, C. Filieri, F. Tagliati, M. Buratto, M.R. Ambrosio et al., Effect of everolimus on cell viability in nonfunctioning pituitary adenomas. J. Clin. Endocrinol. Metab. 95, 968–76 (2010)CrossRef
64.
Zurück zum Zitat V. Cerovac, J. Monteserin-Garcia, H. Rubinfeld, M. Buchfelder, M. Losa, T. Florio et al., The somatostatin analogue octreotide confers sensitivity to rapamycin treatment on pituitary tumor cells. Cancer Res. 70, 666–74 (2010)CrossRef V. Cerovac, J. Monteserin-Garcia, H. Rubinfeld, M. Buchfelder, M. Losa, T. Florio et al., The somatostatin analogue octreotide confers sensitivity to rapamycin treatment on pituitary tumor cells. Cancer Res. 70, 666–74 (2010)CrossRef
65.
Zurück zum Zitat R. Loewith, E. Jacinto, S. Wullschleger, A. Lorberg, J.L. Crespo, D. Bonenfant et al., Two TOR complexes, only one of which is rapamycin sensitive, have distinct roles in cell growth control. Mol. Cell 10, 457–68 (2002)CrossRef R. Loewith, E. Jacinto, S. Wullschleger, A. Lorberg, J.L. Crespo, D. Bonenfant et al., Two TOR complexes, only one of which is rapamycin sensitive, have distinct roles in cell growth control. Mol. Cell 10, 457–68 (2002)CrossRef
66.
Zurück zum Zitat S.C. Hanna, S.A. Heathcote, W.Y. Kim, mTOR pathway in renal cell carcinoma. Expert. Rev. AntiCancer Ther. 8, 283–92 (2008)CrossRef S.C. Hanna, S.A. Heathcote, W.Y. Kim, mTOR pathway in renal cell carcinoma. Expert. Rev. AntiCancer Ther. 8, 283–92 (2008)CrossRef
67.
Zurück zum Zitat A. O’Donnell, S. Faivre, H.A. Burris 3rd, D. Rea, V. Papadimitrakopoulou, N. Shand et al., Phase I pharmacokinetic and pharmacodynamic study of the oral mammalian target of rapamycin inhibitor everolimus in patients with advanced solid tumors. J. Clin. Oncol.: Off. J. Am. Soc. Clin. Oncol. 26, 1588–95 (2008)CrossRef A. O’Donnell, S. Faivre, H.A. Burris 3rd, D. Rea, V. Papadimitrakopoulou, N. Shand et al., Phase I pharmacokinetic and pharmacodynamic study of the oral mammalian target of rapamycin inhibitor everolimus in patients with advanced solid tumors. J. Clin. Oncol.: Off. J. Am. Soc. Clin. Oncol. 26, 1588–95 (2008)CrossRef
68.
Zurück zum Zitat C. Di Pasquale, E. Gentilin, S. Falletta, M. Bellio, M. Buratto, E. Degli Uberti et al. , PI3K/Akt/mTOR pathway involvement in regulating growth hormone secretion in a rat pituitary adenoma cell line. Endocrine 60, 308–316 (2018).CrossRef C. Di Pasquale, E. Gentilin, S. Falletta, M. Bellio, M. Buratto, E. Degli Uberti et al. , PI3K/Akt/mTOR pathway involvement in regulating growth hormone secretion in a rat pituitary adenoma cell line. Endocrine 60, 308–316 (2018).CrossRef
69.
Zurück zum Zitat R.J. Shaw, L.C. Cantley, Ras, PI(3)K and mTOR signalling controls tumour cell growth. Nature 441, 424–30 (2006)CrossRef R.J. Shaw, L.C. Cantley, Ras, PI(3)K and mTOR signalling controls tumour cell growth. Nature 441, 424–30 (2006)CrossRef
70.
Zurück zum Zitat M. Breuleux, M. Klopfenstein, C. Stephan, C.A. Doughty, L. Barys, S.M. Maira et al., Increased AKT S473 phosphorylation after mTORC1 inhibition is rictor dependent and does not predict tumor cell response to PI3K/mTOR inhibition. Mol. Cancer Ther. 8, 742–53 (2009)CrossRef M. Breuleux, M. Klopfenstein, C. Stephan, C.A. Doughty, L. Barys, S.M. Maira et al., Increased AKT S473 phosphorylation after mTORC1 inhibition is rictor dependent and does not predict tumor cell response to PI3K/mTOR inhibition. Mol. Cancer Ther. 8, 742–53 (2009)CrossRef
Metadaten
Titel
Effect of combined treatment with a pan-PI3K inhibitor or an isoform-specific PI3K inhibitor and everolimus on cell proliferation in GH-secreting pituitary tumour in an experimental setting
verfasst von
Claudia Pivonello
Roberta Patalano
Domenico Solari
Renata S. Auriemma
Federico Frio
Francesca Vitulli
Ludovica F. S. Grasso
Marialuisa Di Cera
Maria Cristina De Martino
Luigi M. Cavallo
Paolo Cappabianca
Annamaria Colao
Rosario Pivonello
Publikationsdatum
31.07.2018
Verlag
Springer US
Erschienen in
Endocrine / Ausgabe 3/2018
Print ISSN: 1355-008X
Elektronische ISSN: 1559-0100
DOI
https://doi.org/10.1007/s12020-018-1677-2

Weitere Artikel der Ausgabe 3/2018

Endocrine 3/2018 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.