Endocrine DOI 10.1007/s12020-014-0380-1

REVIEW

PI3K/Akt/mTOR signaling in medullary thyroid cancer: a promising molecular target for cancer therapy Gloria Irene Manfredi • Alessandra Dicitore Germano Gaudenzi • Michele Caraglia • Luca Persani • Giovanni Vitale



Received: 21 June 2014 / Accepted: 1 August 2014 Ó Springer Science+Business Media New York 2014

Abstract The phosphatidylinositol 3-kinase (PI3K)/Akt/ mammalian target of rapamycin (mTOR) pathway is a central hub for the regulation of cell proliferation, apoptosis, cell cycle, metabolism, and angiogenesis. Several studies have recently suggested that the PI3K/Akt/mTOR signaling pathway is implicated in the pathogenesis and progression of neuroendocrine tumors. Medullary thyroid cancer (MTC) is a neuroendocrine tumor developing from the C cells of the thyroid. Mutations in the RET protooncogene are involved in the pathogenesis of several forms of MTC. The deregulation of the PI3K/Akt/mTOR pathway seems to contribute to the tumorigenic activity of RET proto-oncogene mutations. Targeting this pathway through specific inhibitors at simple or multiple sites may represent an attractive potential therapeutic approach for patients with advanced MTCs. The aim of this review is to examine the role of the PI3K/Akt/mTOR pathway in the development and progression of MTC and the new therapeutic options that target this signaling pathway. Keywords Medullary thyroid cancer  mTOR  PI3K/ Akt  Targeted therapy

G. I. Manfredi  G. Gaudenzi  L. Persani  G. Vitale Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy A. Dicitore  L. Persani  G. Vitale (&) Laboratory of Endocrine and Metabolic Research, Istituto Auxologico Italiano IRCCS, University of Milan, Via Zucchi 18, Cusano Milanino (MI), 20095 Milan, Italy e-mail: [email protected] M. Caraglia Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy

Introduction Medullary thyroid cancer (MTC) is a neuroendocrine tumor (NET) developing from the C cells of the thyroid [1, 2]. This tumor accounts for about 5–10 % of all thyroid cancers, and it is characterized by the production of calcitonin [1]. MTC occurs in the sporadic form in about 70–80 % of cases, whereas the remaining 20–30 % is represented by three familial forms: multiple endocrine neoplasia type 2A (MEN 2A), multiple endocrine neoplasia type 2B (MEN 2B), and familial MTC not associated with MEN (FMTC) [3–7]. Surgery is the predominant treatment in MTC, while molecular targeted therapy is recently used in patients with advanced disease. Chemotherapy and radiotherapy play a marginal role in advanced MTC. This tumor is resistant to most of the common chemotherapy drugs. External radiation therapy is currently used only for palliation of locally recurrent tumors or symptomatic bone, central nervous system, and mediastinal metastases [8, 9]. Several studies have recently suggested that the phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) signaling pathway might be involved in NET tumorigenesis and progression, given its critical role in cell proliferation and angiogenesis [10–19]. In this context, it has been demonstrated that PI3K signaling pathway is constitutively activated by several aberrant receptor tyrosine kinases (RTKs), leading to high cell proliferation and vascularity [20–22]. NETs can also harbor different alterations of PI3K signaling, such as mutations in phosphatase and tensin homolog (PTEN), tuberous sclerosis complex 2 (TSC2) or PIK3CA genes, and other pathway abnormalities that lead to mTOR overexpression or Akt activation [20, 23–25]. For all these reasons, the inhibitors of PI3K/Akt/mTOR pathway are emerging as a new targeted therapeutic strategy for NETs [16, 21, 26–28].

123

Endocrine

Growth factors

FGF-3 GDNF VEGF PDGF

EGF

IGF-1

PIP3

PIP2

RTKs

PTEN

PI3K inhibitors -LY294002

3 mTORC1 inhibitors -Rapamycin -Everolimus -Temsirolimus

mTOR

Akt

2

RAF

R

TO

m mL

ST8

R

ap

4 Dual PI3K/mTOR inhibitors -BEZ235

4

Rictor

TSC-2

MEK

Pr

m LS T8

TSC-1

r PRAS40

2 AKT inhibitors -MK-2206 -Perifosine -Triciribine

mSIN1

4

mTORC2

4EBP1

mTORC1

to

1

1

ot or

PI3K

RAS

3

4

ERK

S6K1

eIF4E

Proliferation Cell cycle progression Differentiation Angiogenesis Inhibition of apoptosis Migration Protein synthesis

Fig. 1 Overview of the PI3K/Akt/mTOR signaling pathway in cancer cells. This pathway is commonly up-regulated in tumor cells through: 1 direct upstream stimulation (receptor tyrosine kinases: RTKs); 2 intrinsically via activating genetic alterations in PI3K, Akt or mTOR, or via loss of function in PTEN; 3 indirect activation via

cross-talk with Ras pathway. Specific inhibitors of PI3K, Akt, and mTOR could represent a potential therapeutic innovative strategy in tumors showing an overactivation of this pathway, such as medullary thyroid cancer

Indeed, in May 2011, the mTOR inhibitor everolimus was approved by FDA for the treatment of patients with progressive unresectable pancreatic NETs. It has recently been observed that MTC cell proliferation also depends on the activation of the PI3K/Akt/mTOR signaling cascade [29–32]. Therefore, inhibition of this pathway may be a novel therapeutic approach for MTC [33, 34]. The aim of this review is to examine the role of the PI3K/Akt/mTOR pathway in the development and treatment of MTC.

factors (vascular endothelial growth factor, insulin-like growth factor-1/2, fibroblast growth factor-3, plateletderived growth factor, glial cell line-derived neurotrophic factor, etc.), amino acids, and hypoxia [20, 35]. The main pathway that controls mTOR functions is the PI3K/Akt signaling. Activation of PI3K/Akt is driven by several RTKs, stimulated by specific growth factors (Fig. 1). When these receptors are activated, PI3K is recruited and catalyzes the conversion of phosphatidylinositol 4,5-bisphosphate (PIP2) in phosphatidylinositol3,4,5-trisphosphate (PIP3) and, thus, activates Akt. The latter, through the inactivation of mTOR’s negative regulators ‘‘TSC2’’ and ‘‘proline-rich Akt substrate 40 (PRAS40),’’ enables mTOR to carry out its multiple functions [26]. On the other hand, the main mTOR negative regulator is PTEN, a tumor suppressor which inhibits Akt, converting PIP3 back to PIP2 [36]. mTOR exists in two multiprotein complexes: mTOR complexes 1 and 2 (mTORC1 and mTORC2) (Fig. 1). The first consists of mTOR, mammalian LST8 (mLST8),

The PI3K/Akt/mTOR pathway and its role in carcinogenesis mTOR is a serine/threonine kinase involved in the regulation of cell proliferation, apoptosis, cell cycle, angiogenesis, metabolism, and protein synthesis [26]. mTOR signaling is regulated by numerous stimuli, such as growth

123

Endocrine

PRAS40, and raptor [10]. Its activation results in the phosphorylation of 4E-binding protein (4EBP1) and ribosomal protein S6 kinases (S6K1), both of which regulate transcription and translation of critical growth genes [12, 37, 38]. 4EBP1 prevents the initiation of mRNA translation by binding to eukaryotic initiation factor 4E (eiF4E). When phosphorylated by mTORC1, 4EBP1 reduces its affinity for eiF4E, allowing eiF4E to stimulate translation initiation. Similarly, the phosphorylation of S6K1 promotes mRNA translation by phosphorylating or binding multiple proteins, which collectively affects translation initiation and elongation [39]. Interestingly, the activation of mTORC1 and then the phosphorylation of these downstream targets (4EBP1 and S6K1) preferentially lead to the translation of mRNAs for pro-tumorigenic genes implicated in cell cycle progression, cell differentiation, cell death, angiogenesis, and migration [12, 39]. mTORC2 consists of mTOR, mLST8, mammalian stress-activated protein kinase-interacting protein (mSIN1), protor, and rictor [40, 41]. mTORC2 phosphorylates Akt at Ser473 leading to full Akt activation and then to mTORC1 stimulation, and regulates the phosphorylation of PKCa. It has also been supposed that it controls cytoskeleton organization, cell cycle progression, and cell survival [40–44]. As highlighted before, given that the PI3K/Akt/mTOR signaling controls cell proliferation, cell death, and angiogenesis, an inappropriate activation of this pathway can result in cancer [13–16, 45]. Several genetic alterations are known to induce an overactivation of the PI3K/Akt/mTOR signaling, but the most common mechanism observed in human cancers is the aberrant activation of RTKs [46–49].

Relevance of the PI3K/Akt/mTOR pathway in MTC Activating mutations of the RET proto-oncogene are implicated in the pathogenesis of several forms of MTC [50]. Germline mutations of this gene have been detected in about 88–98 % of familial forms [5, 50], while somatic mutations of RET gene have been reported in about 23–70 % of sporadic MTCs [31]. The RET proto-oncogene encodes a RTK expressed in cells derived from the neural crest, including C cells [2]. The ligands of the RET receptor are growth factors belonging to the glial cell line-derived neurotrophic factor (GDNF) family [2, 3]. The activation of this receptor regulates growth, survival, differentiation, and migration of cells through the stimulation of several intracellular pathways, including RAS/RAF/MEK/ERK and PI3K/Akt/ mTOR (Fig. 1) [8, 51, 52]. Segouffin-Cariou [53] reported that rat fibroblasts expressing a MEN 2A mutant form of RET showed a constitutive activation of PI3K and Akt. Interestingly, the

overactivation of PI3K/Akt/mTOR pathway results to be a limiting factor for RET-MEN 2A oncogenic potential. Indeed, Drosten et al. [54] constructed adenoviral vectors expressing a truncated form of RET, which eliminates its oncogenic functions, and studied its effects on TT cells, in terms of cell cycle, proliferation, apoptosis, viability, and RET downstream signaling. The expression of the truncated RET was associated to a suppression of Akt activity and to a reduction of cell proliferation and viability. Moreover, they detected elevated levels of cyclin D1 mRNA, a cell cycle regulator which is known to be controlled by PI3K/Akt pathway. In summary, these results suggest that oncogenic RET activity in MTC is partially modulated by the overactivation of the PI3K/Akt/mTOR pathway [54]. Tamburrino et al. [31] reported that mTOR pathway has a role in the primary phases of MTC tumorigenesis. In fact, mTOR activation appeared to be an early event in C-cell transformation. They also showed that the activation of mTOR was higher in lymph-node metastasis than in primary MTC, suggesting a role of this pathway in tumor progression as well as initiation. In support of this hypothesis, Kouvaraki et al. [55] reported a strong correlation between eiF4E expression and tumor stage in thyroid cancer. Interestingly, a strong expression of the most critical downstream targets of mTOR signaling (4EBP1 and eiF4E) was commonly observed in MTC [55]. In order to deepen the possible role of mTOR in the development of familial MTC, Rapa et al. [30] screened 65 cases of MTC (11 of which occurring as familial tumors in MEN 2 syndrome) for PI3K mutations. Although they did not find any mutations, PI3K/Akt/mTOR pathway was highly activated in MTC and correlated to the germline RET mutation status. In fact, the vast majority of cases with RET germline mutations were highly positive for phospho-mTOR and phospho-S6K1. On the other hand, MTC with sporadic RET mutations or wild-type RET showed heterogeneous activation of mTOR pathway. Therefore, the association between RET status and mTOR activity appears to be independent on the type of mutation, but dependent on its hereditary nature. Although, it is difficult to elucidate the molecular mechanism at the basis of such phenomenon, it has been recently demonstrated that mTOR activity was exclusively RET-dependent in MTC cancer cell lines with endogenous mutations of the proto-oncogene [56]. In fact, the treatment of two different RET mutant MTC cell lines (TT and MZ-CRC-1) with AST487, a RET kinase inhibitor, inhibited cell proliferation and showed a profound suppression of the mTORC1 signaling in a dose-dependent manner. On the other hand, mTOR effectors were not suppressed in the FTC133 cell line, which harbors a homozygous PTEN loss of function, but it is wild type for RET [56].

123

Endocrine

However, the aberrant activation of RET seems to be not the only factor implicated in the overactivation of PI3K/ Akt/mTOR pathway. In fact, while activation of mTOR signaling has been reported in 96–100 % of MTC [31, 55], the rate of activating RET mutations, particularly for sporadic MTC, is lower [57]. Therefore, several other factors, that need to be explored, may be involved in the overactivation of mTOR signaling in MTC.

Utility of PI3K/Akt/mTOR inhibitors in MTC: preclinical studies RAD001 (everolimus), a synthetic derivative of rapamycin, is a potent selective mTORC1 inhibitor (Fig. 1), which exerts its activity binding with an intracellular protein, FKBP12. This complex interacts with mTORC1 and inhibits signaling downstream [16]. In 2010, GrozinskyGlasberg et al. [58] investigated the in vitro effects of everolimus in a human MTC cell line (TT) and in two primary cultures from human MTC. Everolimus significantly inhibited cell viability and decreased cell number in a dose-dependent manner and increased the number of TT cells in the G0/G1 phase of the cell cycle, but no effect of this drug on the modulation of apoptosis has been reported. Calcitonin and CEA levels appeared not to be affected by RAD001 in MTC cells, suggesting that this drug does not modify hormone secretion. Most of these data were confirmed in other MTC models. We recently showed a potent anti-proliferative effect of everolimus in both TT and MZ-CRC-1 cell lines. This antitumor activity was mediated by the induction of cell cycle arrest in G0/G1 phase, but not apoptosis. Interestingly, this study revealed that everolimus can also induce cellular senescence; indeed, the percentage of TT senescent cells, positive at the staining for b-galactosidase activity, doubled after the treatment with everolimus. In addition, incubation of TT and MZ-CRC-1 cell lines with everolimus did not influence calcitonin secretion [59]. The cell cycle arrest and the induction of senescence obtained specifically with mTOR inhibition could be mediated by cyclin D downregulation and/or p21 upregulation, which are known cell cycle regulators and participate in the launch of the senescence program [55, 60, 61]. Although everolimus has demonstrated significant growth-inhibitory effects on a variety of human malignancies, it is increasingly recognized that its mechanism of action may not be sufficient for achieving a potent anticancer effect due to its inability to inhibit mTORC2 activity. Indeed, the specific inhibition of mTORC1 by everolimus seems to be associated with a paradoxical Akt activation mediated by mTORC2, and by the inhibition of a negative feedback loop via S6K1 leading to the attenuation

123

of its therapeutic effects [26]. Therefore, an upstream inhibition or targeting the PI3K/AKT/mTOR pathway at multiple levels may represent innovative therapeutic strategies to maximize blockade of the mTOR pathway and related antitumor activity. In fact, an upstream block of the PI3K/Akt/mTOR pathway through PI3K inhibitors seems to have additional effects on apoptosis (Fig. 1) [62]. Treatment of TT cells with the PI3K inhibitor LY294002 significantly suppressed cell proliferation through the inhibition of the phosphorylation of Akt and the induction of apoptosis. Moreover, LY294002 induced a progressive decrease in the expression of Chromogranin A and ASCL1, both proteins critical for the neuroendocrine cell differentiation and hormone production [62]. Unfortunately, LY294002 is too insoluble, making it ineffective in cancer clinical trials [63]. However, MK-2206, a novel orally bioavailable Akt-specific inhibitor, showed in vitro antitumor effects comparable to that observed after LY294002: 1) suppression of TT cell proliferation, 2) induction of apoptosis, and 3) decrease in expression of chromogranin A and ASCL1 [57]. Interestingly, the antitumor effect of MK-2206 resulted to be independent of RET inhibition, as RET activity was not blocked by MK-2206 incubation [57]. Further studies analyzed the possible effects of combined targeted therapies, which act at different sites of the PI3K/Akt/mTOR pathway (Fig. 1). The efficacy of BEZ235, a dual PI3K/mTOR inhibitor, which reduces PI3K, mTORC1, and mTORC2 kinase activity by competitive binding to the ATP-binding cleft of these enzymes, has been evaluated in vitro treating several thyroid cancer cell lines of four major pathological types [64]. BEZ235 was found to effectively inhibit cell proliferation in thyroid cancer cell lines. Anaplastic thyroid cancer cells were the most sensitive, followed by follicular undifferentiated, medullary, and well-differentiated thyroid cancer cell lines. Cancer cells harboring a PI3K gain-offunction mutation or a PTEN deletion demonstrated higher PI3K/mTOR pathway activity and greater sensitivity to BEZ235. In addition, the expression of p-S6 ribosomal protein and p27 correlated with the sensitivity of BEZ235 in thyroid cancer [64]. The concomitant targeting of RET and mTOR may represent another innovative therapeutic strategy in MTC. In fact at least in vitro, combined treatment with AST487 (a RET kinase inhibitor) and INK128 (an ATP-competitor inhibitor of mTORC1 and mTORC2) at low concentrations synergistically suppressed proliferation of RET mutant MTC cell lines through the induction of apoptosis [65]. In this regard, several natural compounds derived from plants have been tested for antitumor and cytotoxic properties. Withanolides and flavonoids are known to inhibit PI3K/ Akt/mTOR pathway. Several novel withanolides extracted

Endocrine

from Physalis plant resulted to suppress activation of both RET and PI3K/Akt/mTOR signaling, leading to the inhibition of cell growth through cell cycle arrest and the induction of apoptosis in MTC cell lines [34]. The Ras/Raf/MEK/ERK signaling represents another crucial pathway involved in the pathogenesis and progression of MTC, particularly in RET-negative MTC [65]. Interestingly, Ras is capable of signaling through both Ras/ Raf/MEK/ERK and PI3K/Akt/mTOR pathways. In fact, both pathways are known to interact at multiple sites, and this can explain the occurrence of resistance to anticancer agents that target a single signaling [66]. Jin et al. [67] evaluated the antitumor activity of RAF265/CHIR-265, an ATP-competitive pan-RAF inhibitor, in combination with the dual PI3K/mTOR inhibitor BEZ235 in MTC preclinical models. This treatment strongly suppressed cell proliferation, both in vitro and in vivo, by inhibiting ERK and PI3K signaling and inducing cell cycle arrest in G0/G1 phase.

Everolimus as a possible therapy for MTC: clinical studies Few preliminary clinical studies suggest that mTOR inhibitors such as everolimus could be a possible treatment option for patients affected by MTC. Druce et al. [68] reported a case of a 57-year-old lady with MEN 2A and progressive metastatic MTC treated with everolimus (10 mg daily). The treatment was well tolerated and the disease, that was previously progressive, initially stabilized. Interestingly, the calcitonin doublingtime increased from 5.5–62 months during the treatment over the first 10 months. Subsequently, over a further 12 months, a decrease in the calcitonin doubling-time (27.9 months) and some minor size increase in the mediastinal lymph nodes and a new liver lesion were observed. Faggiano et al. [59] recently treated two patients affected by progressive metastatic MTC with everolimus in combination with somatostatin analog (octreotide). Patient # 1 was treated with everolimus initially at the dose of 10 mg a day, and then reduced to 5 mg a day because of hematological toxicity (grade 3 thrombocytopenia and grade 2 anemia). Serum calcitonin rapidly decreased one month after the start of everolimus, but it moderately rose again after the change of the dosage. Serum carcinoembryonic antigen (CEA) variations paralleled calcitonin variations, though being lower. Neurological symptoms secondary to vertebral metastasis, such as paraplegia, pain, and bladder dysfunction, improved during the treatment. Patient # 2 was initially treated with everolimus at the dose of 5 mg a day, because of a preexisting mild anemia, then, after a two months treatment, it was increased to 10 mg daily because no change in the serum markers occurred. The higher

dosage determined a marked decrease of serum calcitonin and CEA. Bone metastases were substantially unchanged, while a dramatic decrease in tumor vascularization and size was observed in laterocervical lymph node metastases. A multicenter phase II trial [69] recently investigated the efficacy and tolerability of everolimus in patients with progressive locally advanced or metastatic thyroid cancer of all histological subtypes. Forty patients with progressive, locally advanced, or I131 refractory thyroid cancers (9/40 MTC) were enrolled in this study, and they self-administered everolimus 10 mg orally once a day, until severe toxicity or disease progression. They achieved a confirmed objective response in 2/40 (5 %) patients, and a stable disease was described in 29/40 (76 %) patients. Clinical benefits were reported in 50 % patients. They did not observe any association between efficacy and histology, but it is reported a high proportion of disease control (stable disease [ 12 months) in patients with differentiated thyroid cancer and MTC. The progression free survival according to histology was not reached in MTC, but all patients enrolled with this kind of neoplasm showed at least a stable disease (2/ 9 experienced tumor shrinkage) at the end of the treatment, despite the initial condition of progressive, locally advanced, or metastatic cancer. Serum calcitonin and CEA levels were measured for 9 MTC patients. Calcitonin and CEA levels were C 50 % lower than baseline in 3 (30 %) and 4 (44 %) patients with MTC, respectively. Adverse effects registered during the therapy were predominantly grade 1 or 2; the most common effects reported were mucositis (84 %), anorexia (44 %), liver enzymes elevation (26 %), and rash (21 %). Some ongoing clinical trials are currently evaluating the antitumor activity of everolimus, alone or in combination with other drugs, in MTCs. A phase II trial is studying the efficacy of everolimus in patients with progressive or recurrent, unresectable, or metastatic MTC (Identifier: NCT01118065) [70]. Two studies are evaluating the efficacy of everolimus associated with the ‘‘pan-receptor’’ somatostatin ligand pasireotide (SOM230) in patients with progressive and advanced MTC (NCT01625520 and NCT01270321) [70]. Another trial is studying the effects of everolimus associated with cixutumumab, a monoclonal antibody against IGF-I receptor, in advanced neuroendocrine tumors, including MTC (Identifier: NCT01204476) [70]. Moreover, the effects of temsirolimus, another mTOR inhibitor, in combination with chemotherapy (vinorelbine ditartrate) are currently being studied in several neoplasms, including MTC (Identifier: NCT01155258) [70].

Conclusions Recent findings show that the overactivation of the PI3K/ Akt/mTOR pathway plays a crucial role in the

123

Endocrine

pathogenesis of MTC. Indeed, most of the tumorigenic effects of RET mutations seem to be mediated by the activation of PI3K/Akt/mTOR cascade. Few preliminary clinical studies show the potential beneficial effects of everolimus in the treatment of MTC, but further studies are needed to confirm this hypothesis. In addition, the development of novel therapeutic strategies targeting RET/ PI3K/Akt/mTOR pathway at multiple sites and other survival pathways for cancer cells, such as Ras/Raf/MEK/ ERK signaling, may open a new scenario in the treatment of MTC. Acknowledgments This work was partially supported by the Italian Ministry of Education, Research and University (FIRB RBAP11884 M). Conflict of interest No potential conflict of interest relevant to this article was reported.

References 1. C. Eng, D. Clayton, I. Schuffenecker, G. Lenoir, G. Cote, R.F. Gagel, H.K. van Amstel, C.J. Lips, I. Nishisho, S.I. Takai, D.J. Marsh, B.G. Robinson, K. Frank-Raue, F. Raue, F. Xue, W.W. Noll, C. Romei, F. Pacini, M. Fink, B. Niederle, J. Zedenius, M. Nordenskjo¨ld, P. Komminoth, G.N. Hendy, L.M. Mulligan et al., The relationship between specific RET proto-oncogene mutations and disease phenotype in multiple endocrine neoplasia type 2. International RET mutation consortium analysis. JAMA 276(19), 1575–1579 (1996) 2. C. Eng, L.M. Mulligan, Mutations of the RET protooncogene in multiple endocrine neoplasia type 2, related sporadic tumours and Hirschsprung diseases. Hum. Mutat. 9, 97–109 (1997) 3. B. Pasini, M.G. Borrello, A. Greco, I. Bongarzone, Y. Luo, P. Mondellini, L. Alberti, C. Miranda, E. Arighi, R. Bocciardi et al., Loss of function effect of RET mutations causing Hirschsprung disease. Nature Genet. 10, 35–40 (1995) 4. L. Mulligan, C. Eng, C.S. Healey, D. Clayton, J.B. Kwok, E. Gardner, M.A. Ponder, A. Frilling, C.E. Jackson, H. Lehnert et al., Specific mutations of the RET proto-oncogene are related to disease phenotype in MEN 2A and FMTC. Nature Genet. 6, 70–74 (1994) 5. M.G. Borrello, D.P. Smith, B. Pasini, I. Bongarzone, A. Greco, M.J. Lorenzo, E. Arighi, C. Miranda, C. Eng, L. Alberti et al., RET activation by germline MEN2A and MEN2B mutations. Oncogene 11, 2419–2427 (1995) 6. G. Vitale, M. Caraglia, A. Ciccarelli, G. Lupoli, A. Abbruzzese, P. Tagliaferri, G. Lupoli, Current approaches and perspectives in the therapy of medullary thyroid carcinoma. Cancer 91(9), 1797–1808 (2001) 7. C. Puppin, C. Durante, M. Sponziello, A. Verrienti, V. Pecce, E. Lavarone, F. Baldan, A.F. Campese, A. Boichard, L. Lacroix, D. Russo, S. Filetti, G. Damante, Overexpression of genes involved in miRNA biogenesis in medullary thyroid carcinomas with RET mutation. Endocrine (2014). doi:10.1007/s12020-014-0204-3 8. A. Cerrato, V. De Falco, M. Santoro, Molecular genetics of medullary thyroid carcinoma: the quest for novel therapeutic targets. J. Mol. Endocrinol. 43, 143–155 (2009) 9. C. Durante, A. Paciaroni, K. Plasmati, F. Trulli, S. Filetti, Vandetanib: opening a new treatment practice in advanced medullary thyroid carcinoma. Endocrine 44(2), 334–342 (2013) 10. S. Wullschleger, R. Loewith, M.N. Hall, TOR signaling in growth and metabolism. Cell 124, 471–484 (2006)

123

11. D.M. Sabatini, MTOR and cancer: insights into a complex relationship. Nat. Rev. Cancer 6, 729–734 (2006) 12. T. Schmeizle, M.N. Hall, TOR, a central controller of cell growth. Cell 103, 253–262 (2000) 13. Y. Samuels, Z. Wang, A. Bardelli, N. Silliman, J. Ptak, S. Szabo, H. Yan, A. Gazdar, S.M. Powell, G.J. Riggins, J.K. Willson, S. Markowitz, K.W. Kinzler, B. Vogelstein, V.E. Velculescu, High frequency of mutations of the PIK3CA gene in human cancers. Science 304, 554 (2004) 14. I. Vivanco, C.L. Sawyers, The phosphatidylinositol 3-Kinase AKT pathway in human cancer. Nat. Rev. Cancer 2, 489–501 (2002) 15. A. De Benedetti, J.R. Graff, EIF-4E expression and its role in malignancies and metastases. Oncogene 23, 3189–3199 (2004) 16. J.A. Engelman, Targeting PI3K signaling in cancer: opportunities, challenges and limitations. Nat. Rev. Cancer 9, 550–562 (2009) 17. J. Li, C. Yen, D. Liaw, K. Podsypanina, S. Bose, S.I. Wang, J. Puc, C. Miliaresis, L. Rodgers, R. McCombie, S.H. Bigner, B.C. Giovanella, M. Ittmann, B. Tycko, H. Hibshoosh, M.H. Wigler, R. Parsons, PTEN, a putative protein tyrosine phosphatase gene mutated in human brain, breast, and prostate cancer. Science 275, 1943–1947 (1997) 18. T. Gagliano, M. Bellio, E. Gentilin, D. Mole`, F. Tagliati, M. Schiavon, N.G. Cavallesco, L.G. Andriolo, M.R. Ambrosio, F. Rea, E. Degli Uberti, M.C. Zatelli, mTOR, p70S6K, AKT, and ERK1/2 levels predict sensitivity to mTOR and PI3K/mTOR inhibitors in human bronchial carcinoids. Endocr. Relat. Cancer 20(4), 463–475 (2013) 19. K.L. Yim, Role of biological targeted therapies in gastroenteropancreatic neuroendocrine tumours. Endocrine 40(2), 181–186 (2011) 20. J. Capdevila, J. Tabernero, A shining light in the darkness for the treatment of pancreatic neuroendocrine tumors. Cancer Discov. 1, 213–221 (2011) 21. M.C. Zatelli, M. Minoia, C. Martini, F. Tagliati, M.R. Ambrosio, M. Schiavon, M. Buratto, F. Calabrese, E. Gentilin, G. Cavallesco, L. Berdondini, F. Rea, E.C. degli Uberti, Everolimus as a new potential antiproliferative agent in aggressive human bronchial carcinoids. Endocr. Relat. Cancer. 17(3), 719–729 (2010) 22. K.I. Alexandraki, G. Kaltsas, Gastroenteropancreatic neuroendocrine tumors: new insights in the diagnosis and therapy. Endocrine 41(1), 40–52 (2012) 23. E. Missaglia, I. Dalai, S. Barbi, S. Beghelli, M. Falconi, M. della Peruta, L. Piemonti, G. Capurso, A. Di Florio, G. delle Fave, P. Pederzoli, C.M. Croce, A. Scarpa, Pancreatic endocrine tumors: expression profiling evidences a role for Akt-mTOR pathway. J. Clin. Oncol. 28, 245–255 (2010) 24. Y. Jiao, C. Shi, B.H. Edil, R.F. de Wilde, D.S. Klimstra, A. Maitra, R.D. Schulick, L.H. Tang, C.L. Wolfgang, M.A. Choti, V.E. Velculescu, L.A. Diaz Jr, B. Vogelstein, K.W. Kinzler, R.H. Hruban, N. Papadopoulos, DAXX/ATRX, MEN1 and mTOR pathway genes are frequently altered in pancreatic neuroendocrine tumors. Science 331, 1199–1203 (2011) 25. T. Shida, T. Kishimoto, M. Furuya, T. Nikaido, K. Koda, S. Takano, F. Kimura, H. Shimizu, H. Yoshidome, M. Ohtsuka, T. Tanizawa, Y. Nakatani, M. Miyazaki, Expression of an activated mammalian target of rapamycin (mTOR) in gastroenteropancreatic neuroendocrine tumors. Cancer Chemother. Pharmacol. 65, 889–893 (2010) 26. F. Meric-Bernstam, A.M. Gonzalez-Angulo, Targeting the mTOR signaling network for cancer therapy. J. Clin. Oncol. 27, 2278–2287 (2009) 27. K.G. Foster, D.C. Fingar, Mammalian target of rapamycin (mTOR): conducting the cellular signaling symphony. J. Biol. Chem. 285, 14071–14077 (2010)

Endocrine 28. J.C. Yao, M.H. Shah, T. Ito, C.L. Bohas, E.M. Wolin, E. Van Cutsem, T.J. Hobday, T. Okusaka, J. Capdevila, E.G. de Vries, P. Tomassetti, M.E. Pavel, S. Hoosen, T. Haas, J. Lincy, D. Leb¨ berg, RAD001 in Advanced Neuroendocrine Tumors, wohl, K. O Third Trial (RADIANT-3) Study Group: everolimus for advanced pancreatic neuroendocrine tumors. N. Engl. J. Med. 364(6), 514–523 (2011) 29. R.M. Melillo, F. Carlomagno, G. De Vita, P. Formisano, G. Vecchio, A. Fusco, M. Billaud, M. Santoro, The insulin receptor substrate (IRS)-1 recruits phosphatidylinositol 3-kinase to Ret: evidence for a competition between Shc and IRS-1 for the binding to Ret. Oncogene 20, 209–218 (2001) 30. I. Rapa, E. Saggiorato, D. Giachino, N. Palestini, F. Orlandi, M. Papotti, M. Volante, Mammalian target of rapamycin pathway activation is associated to RET mutation status in medullary thyroid carcinoma. J. Clin. Endocrinol. Metab. 96, 2146–2153 (2011) 31. A. Tamburrino, A.A. Molinolo, P. Salerno, R.D. Chernock, M. Raffeld, L. Xi, J.S. Gutkind, J.F. Moley, S.A. Wells Jr, M. Santoro, Activation of the mTOR pathway in primary medullary thyroid carcinoma and lymph node metastases. Clin. Cancer Res. 18, 3532–3540 (2012) 32. S.A. Wells, M. Santoro, Targeting the RET pathway in thyroid cancer. Clin. Cancer Res. 15, 7119–7123 (2009) 33. M. Mazumdar, A. Adhikary, S. Chakraborty, S. Mukherjee, A. Manna, S. Saha, S. Mohanty, A. Dutta, P. Bhattacharjee, P. Ray, S. Chattopadhyay, S. Banerjee, J. Chakraborty, A.K. Ray, G. Sa, T. Das, Targeting RET to induce medullary thyroid cancer cell apoptosis: an antagonistic interplay between PI3K/Akt and p38MAPK/caspase-8 pathways. Apoptosis 18, 589–604 (2013) 34. A. Samadi, J. Bazzill, X. Zhang, R. Gallagher, H. Zhang, R. Gollapudi, K. Kindscher, B. Timmermann, M.S. Cohen, Novel withanolides target medullary thyroid cancer through inhibition of both RET phosphorylation and the mammalian target of rapamycin pathway. Surgery. 152(6), 1238–1247 (2012) 35. H.D. Skinner, J.Z. Zheng, J. Fang, F. Agani, B.H. Jiang, Vascular endothelial growth factor transcriptional activation is mediated by hypoxia inducible factor 1alpha, HDM2, and p70S6K1 in response to phosphatidylinositol 3kinase/AKT signaling. J. Biol. Chem. 279, 45643–45651 (2004) 36. A. Sekulic, C.C. Hudson, J.L. Homme, P. Yin, D.M. Otterness, L.M. Karnitz, R.T. Abraham, A direct linkage between the phosphoinositide 3-kinase-AKT signaling pathway and the mammalian target of rapamycin in mitogen-stimulated and transformed cells. Cancer Res. 60, 3504–3513 (2000) 37. G. Vitale, S. Zappavigna, M. Marra, A. Dicitore, S. Meschini, M. Condello, G. Arancia, S. Castiglioni, P. Maroni, P. Bendinelli, R. Piccoletti, P.M. van Koetsveld, F. Cavagnini, A. Budillon, A. Abbruzzese, L.J. Hofland, M. Caraglia, The PPAR-c agonist troglitazone antagonizes survival pathways induced by STAT-3 in recombinant interferon-b treated pancreatic cancer cells. Biotechnol. Adv. 30(1), 169–184 (2012) 38. S. Isotani, K. Hara, C. Tokunaga, H. Inoue, J. Avruch, K. Yonezawa, Immunopurified mammalian target of rapamycin phosphorylates and activates p70 S6 kinase alpha in vitro. J. Biol. Chem. 274(48), 34493–34498 (1999) 39. R. Zoncu, A. Efeyan, D.M. Sabatini, mTOR: from growth signal integration to cancer, diabetes and ageing. Nat. Rev. Mol. Cell Biol. 12(1), 21–35 (2011) 40. E. Jacinto, V. Facchinetti, D. Liu, N. Soto, S. Wei, S.Y. Jung, Q. Huang, J. Qin, B. Su, SIN1/MIP1 maintains rictor-mTOR complex integrity and regulates Akt phosphorylation and substrate specificity. Cell 127, 125–137 (2006) 41. J. Martin, J. Masri, A. Bernath, R.N. Nishimura, J. Gera, Hsp70 associates with rictor and is required for mTORC2 formation and activity. Biochem Biophys Res Commun. 372, 578–583 (2008)

42. D.D. Sarbassov, S.M. Ali, D.H. Kim, D.A. Guertin, R.R. Latek, H. Erdjument-Bromage, P. Tempst, D.M. Sabatini, Rictor, a novel binding partner of mTOR, defines a rapamycin-insensitive and raptor-independent pathway that regulates the cytoskeleton. Curr. Biol. 14, 1296–1302 (2004) 43. P.C. McDonald, A. Oloumi, J. Mills, I. Dobreva, M. Maidan, V. Gray, E.D. Wederell, M.B. Bally, L.J. Foster, S. Dedhar, Rictor and integrin-linked kinase interact and regulate Akt phosphorylation and cancer cell survival. Cancer Res. 68, 1618–1624 (2008) 44. I. Hernandez-Negrete, J. Carretero-Ortega, H. Rosenfeldt, R. Herna´ndez-Garcı´a, J.V. Caldero´n-Salinas, G. Reyes-Cruz, J.S. Gutkind, J. Va´zquez-Prado, P-Rex1 links mammalian target of rapamycin signaling to Rac activation and cell migration. J. Biol. Chem. 282, 23708–23715 (2007) 45. Y. Samuels, V.E. Velculescu, Oncogenic mutations of PIK3CA in human cancers. Cell Cycle 3, 1221–1224 (2004) 46. R. Bianco, I. Shin, C.A. Ritter, F.M. Yakes, A. Basso, N. Rosen, J. Tsurutani, P.A. Dennis, G.B. Mills, C.L. Arteaga, Loss of PTEN/MMAC1/TEP in EGF receptor-expressing tumor cells counteracts the antitumor action of EGFR tyrosine kinase inhibitors. Oncogene 22, 2812–2822 (2003) 47. S.L. Moulder, F.M. Yakes, S.K. Muthuswamy, R. Bianco, J.F. Simpson, C.L. Arteaga, Epidermal growth factor receptor (HER1) tyrosine kinase inhibitor ZD1839 (Iressa) inhibits HER2/ neu (erbB2)-overexpressing breast cancer cells in vitro and in vivo. Cancer Res. 61, 8887–8895 (2001) 48. F.M. Yakes, W. Chinratanalab, C.A. Ritter, W. King, S. Seelig, C.L. Arteaga, Herceptin-induced inhibition of phosphatidylinositol-3 kinase and Akt is required for antibody-mediated effects on p27, cyclin D1, and antitumor action. Cancer Res. 62, 4132–4141 (2002) 49. I.K. Mellinghoff, M.Y. Wang, I. Vivanco, D.A. Haas-Kogan, S. Zhu, E.Q. Dia, K.V. Lu, K. Yoshimoto, J.H. Huang, D.J. Chute, B.L. Riggs, S. Horvath, L.M. Liau, W.K. Cavenee, P.N. Rao, R. Beroukhim, T.C. Peck, J.C. Lee, W.R. Sellers, D. Stokoe, M. Prados, T.F. Cloughesy, C.L. Sawyers, P.S. Mischel, Molecular determinants of the response of glioblastomas to EGFR kinase inhibitors. N. Engl. J. Med. 353, 2012–2024 (2005) 50. C. Eng, D. Clayton, I. Schuffenecker, G. Lenoir, G. Cote, R.F. Gagel, H.K. van Amstel, C.J. Lips, I. Nishisho, S.I. Takai, D.J. Marsh, B.G. Robinson, K. Frank-Raue, F. Raue, F. Xue, W.W. Noll, C. Romei, F. Pacini, M. Fink, B. Niederle, J. Zedenius, M. Nordenskjo¨ld, P. Komminoth, G.N. Hendy, L.M. Mulligan et al., The relationship between specific RET proto-oncogene mutations and disease phenotype in multiple endocrine neoplasia type 2. International RET mutation consortium analysis. JAMA. 276(19), 1575–1579 (1996) 51. N. Hay, N. Sonenberg, Upstream and downstream of mTOR. Genes Dev. 18, 1926–1945 (2004) 52. S. Schubbert, K. Shannon, G. Bollag, Hyperactive Ras in developmental disorders and cancer. Nat. Rev. Cancer 7, 295–308 (2007) 53. C. Segouffin-Cariou, M. Billaud, Transforming ability of MEN2ARET requires activation of the phosphatidylinositol 3-kinase/Akt signaling pathway. J. Biol. Chem. 275, 3568–3576 (2000) 54. M. Drosten, G. Hilken, M. Bo¨ckmann, F. Ro¨dicker, N. Mise, A.N. Cranston, U. Dahmen, B.A. Ponder, B.M. Pu¨tzer, Role of MEN2Aderived RET in maintenance and proliferation of medullary thyroid cancer. J. Natl Cancer Inst. 96, 1231–1239 (2004) 55. M.A. Kouvaraki, C. Liakou, A. Paraschi, K. Dimas, E. Patsouris, S. Tseleni-Balafouta, G.Z. Rassidakis, D. Moraitis, Activation of mTOR signaling in medullary and aggressive papillary thyroid carcinomas. Surgery. 150, 1258–1265 (2011) 56. M. Gild, I. Landa, M. Ryder, R.A. Ghossein, J.A. Knauf, J.A. Fagin, Targeting mTOR in RET mutant medullary and

123

Endocrine

57.

58.

59.

60.

61.

62.

63.

differentiated thyroid cancer cells. Endocr. Relat. Cancer 20, 659–667 (2013) J.F. Burke, L. Schlosser, A.D. Harrison, M. Kunnimalaiyaan, H. Chen, MK-2206 causes growth suppression and reduces neuroendocrine tumor marker production in medullary thyroid cancer through Akt inhibition. Ann. Surg. Oncol. 20(12), 3862–3868 (2013) S. Grozinsky-Glasberg, H. Rubinfeld, Y. Nordenberg, A. Gorshtein, M. Praiss, E. Kendler, R. Feinmesser, A.B. Grossman, I. Shimon, The rapamycin derivative RAD001 (everolimus) inhibits cell viability and interacts with the Akt-mTOR-p70S6K pathway in human medullary thyroid carcinoma cells. Mol. Cell. Endocrinol. 315, 87–94 (2010) A. Faggiano, V. Ramundo, A. Dicitore, S. Castiglioni, M.O. Borghi, R. Severino, P. Ferolla, L. Crino`, A. Abbruzzese, P. Sperlongano, M. Caraglia, D. Ferone, L. Hofland, A. Colao, G. Vitale, Everolimus is an active agent in medullary thyroid cancer: a clinical and in vitro study. J. Cell Mol. Med. 16, 1563–1572 (2012) S. Wang, R.V. Lloyd, M.J. Hutzler, M.S. Safran, N.A. Patwardhan, A. Khan, The role of cell cycle regulatory protein, cyclin D1, in the progression of thyroid cancer. Mod. Pathol. 13, 882–887 (2000) S.M. Jafarmejad, G. Li, Regulation of p53 by ING family members in suppression of tumor initiation and progression. Cancer Metastasis Rev. 31(1–2), 55–73 (2012) M. Kunnimayalaan, M. Ndiaye, H. Chen, Apoptosis-mediated medullary thyroid cancer growth suppression by the PI3K inhibitor LY294002. Surgery 140(6), 1009–1015 (2006) R. Fre´de´rick, C. Mawson, J.D. Kendall, C. Chaussade, G.W. Rewcastle, P.R. Shepherd, W.A. Denny, Phosphoinositide-3-

123

64.

65.

66.

67.

68.

69.

70.

kinase (PI3K) inhibitors: identification of new scaffolds using virtual screening. Bioorg. Med. Chem. Lett. 19(20), 5842–5847 (2009) S.F. Lin, Y.Y. Huang, J.D. Lin, T.C. Chou, C. Hsueh, R.J. Wong, Utility of a PI3K/mTOR Inhibitor (NVP-BEZ235) for Thyroid Cancer Therapy. PLoS One 7(10), e46726 (2012) M.L. Gild, I. Landa, M. Ryder, R.A. Ghossein, J.A. Knauf, J.A. Fagin, Targeting mTOR in RET mutant medullary and differentiated thyroid cancer cells. Endocr. Relat. Cancer 20(5), 659–667 (2013) C.D. Britten, PI3K and MEK inhibitor combinations: examining the evidence in selected tumor types. Cancer Chemother. Pharmacol. 71(6), 1395–1409 (2013) N. Jin, T. Jiang, D.M. Rosen, B.D. Nelkin, D.W. Ball, Synergistic Action of a RAF Inhibitor and a Dual PI3K/mTOR Inhibitor in Thyroid Cancer. Clin. Cancer Res. 17, 6482–6489 (2011) M. Druce, T.T. Chung, S. Grozinsky-Glasberg, D.J. Gross, A.B. Grossman, Preliminary report of the use of everolimus in a patient with progressive medullary thyroid carcinoma. Clin. Endocrinol. 77(1), 154–155 (2012) S.M. Lim, H. Chang, M.J. Yoon, Y.K. Hong, H. Kim, W.Y. Chung, C.S. Park, K.H. Nam, S.W. Kang, M.K. Kim, S.B. Kim, S.H. Lee, H.G. Kim, I.I. Na, Y.S. Kim, M.Y. Choi, J.G. Kim, K.U. Park, H.J. Yun, J.H. Kim, B.C. Cho, A multicenter, phase II trial of everolimus in locally advanced or metastatic thyroid cancer of all histologic subtypes. Ann. Oncol. 24(12), 3089–3094 (2013) http://www.clinicaltrials.gov/. Accessed 8 Aug 2014

mTOR signaling in medullary thyroid cancer: a promising molecular target for cancer therapy.

The phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) pathway is a central hub for the regulation of cell proliferation, a...
486KB Sizes 2 Downloads 9 Views