Abstract
Adrenocortical carcinoma (ACC) is a very rare and aggressive tumor with dismal outcomes. Best current treatments include complete surgical resection for localized resectable disease and systemic therapy with mitotane alone or in combination for advanced ACC. Advances in molecular genetic profiling of ACC have created multiple new targets for potential treatment options in ACC. This article reviews the current treatment options available for ACC and discusses the potential new targets identified through molecular profiling.
Keywords: adrenocortical carcinoma, genomics, molecular profiling, systemic treatment
Adrenocortical carcinoma (ACC) is a very rare tumor with an annual incidence of 1–2 cases per million. The long-term outcomes for ACC are poor with a 5-year survival rate of 35% in most series [1-4]. While stages III and IV have a dismal prognosis, localized disease has potential for cure with complete surgical resection [5]. Yet, even with an R0 resection, 50–80% of patients develop recurrent or metastatic disease [6,7]. Although complete surgical resection with negative margins remains the treatment of choice in localized ACC, the role of surgery in patients with recurrent and metastatic disease remains controversial. Most of the data regarding surgery in patients with recurrent or metastatic ACC are derived from retrospective reviews of single institutions. A recent study by Erdogan and colleagues compared clinical outcomes in ACC recurrent patients who underwent surgery with recurrent patients who did not have surgery [8]. They concluded that progression-free survival (PFS) was improved if the time to first recurrence was greater than 12 months and if patients were able to undergo an R0 resection for their recurrence [8]. Although a debulking surgery (R2 resection) did not significantly improve PFS, R2 resection did provide a modest benefit in overall survival compared with patients who did not undergo surgery (22 month vs 11 month) [8]. Other studies have also shown a survival benefit to surgical resection if the disease-free interval was greater than 1 year [9]. Metastasectomy in advanced ACC has provided some benefit for patients. Datrice et al. reported a 41% 5-year survival among 57 patients with metastatic ACC who underwent metastasectomy. The median survival for the cohort was 2.5 years months–12 years) [9]. Gaujoux and colleagues found similar results. In their cohort of 28 patients with metastatic ACC to the liver who underwent resection, they observed a median overall survival of 31.5 months and a 39% 5-year survival rate [10]. Several retrospective studies also identified a similar survival benefit with pulmonary resection for metastatic ACC [11,12]. An aggressive surgical approach for advanced ACC has been advocated in selected patients with more indolent disease since systemic treatment options are very limited. However, the interpretation of these retrospective studies is often difficult owing to a lack of comparable control group and patient selection. The majority of patients in these cohorts who underwent surgery often had less aggressive tumor compared with those in the ‘control group’ with unresectable tumor. This emphasizes that patient selection for surgery should be based on tumor biology. Mitotane has been the backbone of systemic treatment for advanced ACC until recently when a combination therapy of mitotane with etoposide, doxorubicin and cisplatin showed an improved response rate [13]. The objective of this review is to highlight the current systemic treatment for ACC and discuss the advances in molecular profiling that have led to new trials focusing on targeted therapies and the discoveries of potential new therapeutic targets for ACC.
Systemic chemotherapy
Chemotherapy options for ACC are limited. Mitotane, the only US FDA-approved agent for ACC, is an adrenolytic agent derived from the insecticide dichlorodiphenyltrichloroethane. Mitotane alone or in combination has been the standard treatment offered to patients with advanced stage ACC for decades. However, the response rates are dismal at only 30% and the toxic side effects make it difficult for patients to tolerate [14-16]. Discontinuation of mitotane-based therapy because of the toxicities is common. Haak et al. showed an objective tumor response in 15 of 27 patients treated with mitotane who maintained serum levels above 14 mg l−1. They found no tumor response in patients with serum levels of mitotane less than 14 mg l−1 [14]. The ability to reach serum levels of 14 mg l−1 is difficult, especially in the setting of combination therapy. Some advocate for a high-dose approach to mitotane monotherapy and a low-dose approach when mitotane is combined with other cytotoxic drugs [17]. The use of mitotane in combination with standard cytotoxic agents has been investigated with the most common regimens being mitotane + streptozocin (M-Sz) or mitotane + etoposide/doxorubicin/cisplatin (M-EDP). A Phase II trial by Khan et al. exhibited a 36% response rate in patients with advanced ACC receiving M-Sz [18]. Berruti and colleagues reported a 53% response rate in patients receiving M-EDP [13]. Recently the FIRM-ACT trial comparing M-Sz and M-EDP was released. This was a landmark randomized controlled trial in 304 patients with advanced ACC [19]. The trial stated that M-EDP was superior to M-Sz with respect to objective tumor response, PFS and proportion of patients without progression at 1 year. In total, 23.2% of patients with M-EDP had tumor response compared with 9.2% of patients with M-Sz. The PFS was 5.0 months for M-EDP and 2.1 months for M-Sz. There was no significant difference in overall survival between the two groups at the study’s conclusion [19]. Despite an improvement in response rates with M-EDP, it was a toxic regime with 58.1% of patients experiencing serious adverse events [19].
Systemic therapy has a clear role in advanced or metastatic ACC. However, the role of mitotane as adjuvant therapy is less clear and has led to several studies investigating the use of mitotane as postoperative adjuvant therapy in ACC. Terzolo et al. reported a significantly prolonged recurrence-free survival in patients receiving mitotane compared with the two control groups without treatment (42 months vs 10 months and 25 months, respectively) [20]. A retrospective review from MD Anderson Cancer Center (TX, USA) reported that a “lack of adjuvant mitotane” treatment was a predictor of increased risk of recurrence on multivariate analysis [21]. Fassnacht and colleagues reported an improved 5-year survival among patients treated with adjuvant mitotane compared with patients not treated with mitotane (87% vs 53%, p = 0.04) [7]. The 5-year disease-specific survival was 64.1% and overall survival was 52% among patients receiving adjuvant mitotane after surgical resection [22]. Recently, Else and colleagues retrospectively analyzed a large cohort (n = 391) and found that patients who received adjuvant mitotane (n = 105) had a significantly improved recurrence-free survival with no impact on overall survival [23]. Bertherat et al. initially reported an improved survival in 202 patients receiving adjuvant mitotane within 3 months of surgical resection [24]. However, upon further analysis of the 166 patients in that cohort who underwent complete surgical resection, only 52% were treated with adjuvant mitotane. Univariate and multivariate analysis failed to indicate a benefit in disease-free survival among patients receiving adjuvant mitotane [25]. The difference in outcome among these studies is likely due to multiple factors, such as heterogeneous population and selection bias. The controversy over the true benefit of adjuvant mitotane in resected ACC will likely continue until the results of the ADIUVO trial are available. This study aims to assess the efficacy of postoperative adjuvant mitotane in reducing recurrence rates among low to intermediate risk patients with ACC [26].
Molecular oncogenesis of ACC & targeted therapy
Research into the oncogenesis of ACC has increased our understanding of the molecular mechanisms from genomic studies involved in ACC tumor initiation and progression. Potential biomarkers have been identified to aid early diagnosis, improve prognostication and may serve as targets for new drug development [27]. These advances indicate the importance of genetic dysregulation in ACC development [28,29] Pan-genomic analysis of genetic mutations, chromosomal aberrations, DNA methylation, gene expression or microRNA dysregulation can provide essential data that are key to improving the application of personalized treatment for patients with ACC and other cancers [27]. Work by De Martino et al. using hot spot gene sequencing and comparative genomic hybridization identified copy number alterations or genetic mutations in 47.5% of ACC patients [30]. In total, 40% of patients had abnormal regulatory mechanisms in the G1→S-phase of cell cycle progression suggesting that the cell cycle may be a possible therapeutic target to investigate [30]. Gene expression profiling of adrenocortical tumors has helped to differentiate between benign and malignant tumors. Velazquez-Fernandez et al. found clearly different molecular signatures between adrenocortical adenomas and carcinomas [31]. Over 500 genes were significantly differentially expressed between ACC and adrenocortical adenomas (p < 0.01) [31]. Ubiquitin-related genes and several insulin-like growth factor-related genes were significantly upregulated in ACC. Several genes that regulate cell metabolism, (RARRES2, ALDH1A1, CYBRD1, and GSTA4), a cytokine (CXCL10) and cadherin 2 (CDH2) were all significantly downregulated in ACC [31]. Gene expression analysis has been used to identify molecular diagnostic and prognostic markers in ACC [1,32]. de Reynies et al. showed that benign adrenocortical tumors could be distinguished from malignant tumors using microarray technology [1]. In addition, they also showed that malignant tumors could be further divided into two different groups with different outcomes based on their molecular profile. Tumors with the combined expression of DLG7 and PINK1 had better disease-free survival and tumors with combined expression of BUB1B and PINK1 had the best overall survival [1]. Giordano and colleagues displayed gene expression differences between early and late-stage ACC. On a multivariate Cox regression model gene expression remained a significant prognostic indicator of survival that was independent of tumor stage [32]. Recently, alterations in the retinoblastoma gene (RB1) have been described in ACC and may play a role in determining the aggressiveness of the tumor biology [33]. Immunohistochemistry displayed a loss of the retinoblastoma protein in seven of the 26 poor outcome ACCs and in none of the 20 ACCs with better outcomes [33]. Predicting outcomes and prognosis for patients with ACC is crucial for patient care and a current investigation into microRNAs in ACC has identified new potential biomarkers that are predictive of ACC prognosis. Chabre and colleagues reported significant overexpression of miR-483-5p and lower expression of miR-195 in ACC compared with adrenocortical adenoma, consistent with work published by Patterson et al. [34]. Patterson and colleagues used microarray profiling of benign and malignant adrenal tumors. They found that miR-483-5p was significantly upregulated in ACC compared with benign adrenal tumors and that the expression of miR-483-5p can accurately predict which adrenal tumors are malignant [35]. In addition, increased circulating levels of miR-483-5p and decreased circulating levels of miR-195 were associated with poor survival outcomes in patients with ACC [34,36]. The information obtained from gene expression profiling and analysis of micro-RNA has provided new markers of malignancy and increased our understanding of aberrant signaling pathways allowing for potentially new targeted therapies [1,27,31,35,37-40].
IGF-1R antagonists
Molecular profiling demonstrates that the IGF-2 gene is significantly upregulated in approximately 90% of ACC compared with normal adrenals [1,29,31,37,41]. ACC tumors have also been shown to have high expression levels of the IGF-1R protein. [42] Upregulation of IGF-2 and overexpression of IGF-1R suggest an important role in the activation of the IGF pathway and tumorgenesis of ACC. Barlaskar et al. identified a 60-fold increase in IGF-2 expression of ACC compared with adenomas [43]. On tissue microarray analysis the group demonstrated a marked increase in signal intensity of phospho-IGF-1R and phospho-Akt in ACC compared with adenomas confirming overexpression of IGF-2 and IGF-1R and activation of the downstream effector Akt [43].
Understanding the role of the IGF pathway in ACC pathogenesis has led to preclinical and clinical studies with IGF1R antagonists. In vitro and in vivo studies demonstrated decreased ACC cell proliferation and tumor growth inhibition when treated with IGF-1R antagonists [43]. Haluska and colleagues performed a Phase I trial with the anti-IGF-1R monoclonal antibody figitumumab. In their study eight of 14 (57%) patients with advanced ACC treated with figitumumab had disease stability. Four patients had tumor shrinkage that did not meet clinical RECIST criteria for partial response [44]. The randomized double-blinded, placebo-controlled Phase III study of OSI-906 (a small molecule tyrosine kinase inhibitor directed against IGF-1R) in patients with locally advanced or metastatic ACC (GALACCTIC trial) has just been completed. Patients were randomized to placebo or treatment with OSI-906. The results of this trial are pending publication.
mTOR antagonists
The mammalian target of rapamycin (mTOR) is a protein kinase that is involved with cell growth and proliferation. It is activated in part by IGF-1R through the PI3K-Akt pathway and has been studied recently in cancer as a potential targeted therapy [45,46]. In vitro and in vivo mouse studies by Doghman et al. demonstrated that inhibition of mTOR signaling reduces adrenocortical tumor growth [47]. However, several studies have shown that mTOR inhibitors activate Akt through an IGF-1R-independent mechanism resulting in reduced antitumor effects of mTOR inhibitor and suggesting that IGF-1R inhibitors may reduce the Akt phosphorylation caused by mTOR inhibitors [48,49]. These findings support the rationale of combining mTOR inhibitor with an IGF-1R inhibitor to achieve additive antitumor effects in ACC [50]. A Phase I trial of cixutumumab (anti-IGF-1R antibody) in combination with temsirolimus (mTOR inhibitor) for patients with advanced cancer found tumor reduction in four of 10 patients with ACC [51]. Combination therapy of cixutumumab and temsirolimus resulted in a greater than 6 month stability of ACC in 42% of patients receiving treatment [52]. One-third of ACC patients treated with combination temsirolimus and lenalidomide (immunomodulatory drug with antiangiogenic properties) had disease stability greater than 6 months in a Phase I trial for patients with advanced cancer [53]. Further investigation into combination therapy of an mTOR inhibitor and an IGF-1R inhibitor is warranted based on early Phase I data.
Receptor tyrosine kinase inhibitors
Tyrosine kinase inhibitors have become important targeted therapies for cancer. The molecular profiling of ACC has revealed upregulation or overexpression of several receptor tyrosine kinases [54-59]. EGFR is expressed in more than 75% of ACC tumors [54,55,57]. Bernini et al. found a significantly higher expression of VEGF in ACC than in adrenal adenomas [59]. The overexpression of EGFR and VEGF has made receptor tyrosine kinase inhibition an attractive targeted strategy for the treatment of ACC. However, the results to date have been disappointing. In a Phase II trial, single agent Gefitinib, an EGFR antagonist, failed to demonstrate activity in 19 patients with advanced ACC [60]. Combination therapy of erlotinib and gemcitabine was also unsuccessful at preventing tumor progression in eight of 10 patients with advanced ACC [61]. Anti-VEGF antibody bevacizumab in combination with capecitabine provided no objective response or stability of disease in 10 patients with advanced ACC [62].
Multikinase inhibitors have also been evaluated as potential new therapies in ACC. A Phase I study reported disease stability in two patients with advanced ACC treated with sorafenib and tipifarnib (farnesyltransferase inhibitor) and a case report described a 28 month sustained response to sorafenib in a patient with metastatic ACC [63,64]. Despite encouraging early data, a recent Phase II study of sorafenib with paclitaxel was terminated early owing to disease progression in nine of 25 patients with advanced ACC at the first assessment after starting treatment [65]. Sunitinib also inhibits multiple receptor tyrosine kinases. A partial response was noted in a patient with metastatic ACC who was treated with sunitinib after failing cytotoxic chemotherapy. The response was achieved in 4 months, however after 7.5 months the patient experienced tumor progression [66]. A Phase II trial of sunitinib in refractory ACC demonstrated disease stability in five of 35 patients for a 15.4% response rate and median PFS of 2.8 months. In the patients who responded to sunitinib the median PFS was between 5.6 and 11.2 months with an overall survival between 14 and 35.5 months [67]. Even though preclinical data for tyrosine kinase inhibitors were promising, clinical studies have failed to demonstrate durable, long-term response in patients with advanced ACC. The lack of clinical response to tyrosine kinase inhibitors may be partially due to increased CYP3A4 activity from mitotane. Several studies have shown that mitotane induces the CYP3A4 system making it difficult to achieve appropriate serum drug levels. Tyrosine kinase inhibitors are also metabolized by the CYP3A4 system, suggesting that the serum concentrations of tyrosine kinase inhibitors may have been below adequate serum drug levels. The previous studies using tyrosine kinase inhibitors did not monitor drug concentration levels and future clinical trials that may incorporate or use tyrosine kinase inhibitors should consider serum drug concentration monitoring and dose modification as needed [68,69].
Future targets for novel therapy
Wnt/β-catenin pathway
Microarray analysis shows upregulation of the Wnt/β-catenin pathway [70,71]. The CTNNB1 gene, which encodes β-catenin, is mutated in adrenal tumors [71,72]. The main event in pathogenesis of ACC may be related to activation of β-catenin and its nuclear localization. Moreover, the nuclear localization of β-catenin has been identified as a predictive factor associated with poor prognosis [73,74]. Preclinical studies using a transgenic mouse model have linked β-catenin activation to adrenal cortical dysplasia [75]. Furthermore, PKF115–584, a small molecule inhibitor of T-cell factor/β-catenin complex, has inhibited cell proliferation and induced apoptosis in NCI-H295R ACC cell lines [76]. Further studies of targeted therapies directed at this pathway are required.
Steroidogenic factor-1
Steroidogenic Factor-1 (SF-1) is a nuclear transcription factor involved with adrenocortical cell proliferation and steroidogenesis [77]. An increased overexpression of SF-1 in pediatric adrenal tumors has peaked interest in the role of SF-1 in tumor development [78]. Doghman and colleagues demonstrated that SF-1 regulates cellular proliferation, apoptosis, angiogenesis, adhesion to the extracellular matrix, cytoskeleton dynamics, transcriptional and post-transcriptional regulation of gene expression in adrenocortical cells [79]. Patients with increased expression of SF-1 had a worse prognosis compared with patients with lower expression of SF-1, suggesting its prognostic implication [80]. Recently SF-1 inverse agonists have been shown to selectively inhibit proliferation in the H295R ACC cell line with increased expression of SF-1 [77]. Targeting SF-1 can reduce cortisol production and may provide symptom relief for patients with ACC-associated hypercortisolism, in addition to anti-tumor effect.
PPARγ antagonists
PPARγ is a nuclear transcription factor expressed in the adrenal cortex of normal tissue and in adrenal tumors [81,82]. Thiazolidinedione is a class of drugs that are ligands for PPARγ. Rosiglitazone is a thiazolidinedione that has been shown to have antiproliferative effects on several different cancers [83]. In vitro and in vivo data demonstrate that thiazolidinediones have an antiproliferative effect on ACC cells [81,82,84-86]. Luconi et al. showed that treatment with rosiglitazone resulted in a significant reduction in tumor growth in the treated mice compared with the control group. In contrast to aggressive and more invasive tumors with abundant vascular network and high mitotic figures found in the control group, the tumors of the rosiglitazone-treated mice displayed a noninfiltrating margin, smaller vessels and apoptotic cells [86]. Despite the promising results from preclinical work, the true mechanism responsible for decreased proliferation by rosiglitazone is not completely understood. Preclinical data suggest that rosiglitazone works through both PPARγ-dependent and PPARγ-independent pathways to cause growth arrest, cell death and decreased neovascularization [81,84,85]. Rosiglitazone inhibits ACC cell proliferation by interfering with Akt and ERK1/2 phosphorylation mediated by IGF-1 [84]. PPARγ antagonists may hold potential as a future treatment option for ACC.
Selective estrogen receptor modulators
Normal adrenal cortical tissue and adrenocortical tumors express aromatase, estrogen receptors alpha and beta (ERα, ERβ) and androgens [87]. Aromatase is essential for peripheral conversion of androgens to estrogens. Barzon et al. demonstrated overexpression of aromatase in adrenocortical tumors. They also showed, through immunohistochemistry, that ERβ has a twofold greater expression than ERα in normal adrenal tissue. However, in ACC tumors the ratio of ERα/ERβ was increased indicating an overexpression of ERα in adrenocortical carcinoma [87]. An in vitro study of NCI-H295R ACC cell line demonstrated that ACC cells are capable of converting androgens to estrogens via aromatase. The newly converted estrogens may regulate cell proliferation through an autocrine mechanism mediated by the ER [88]. Montanaro et al. found that ER antagonists cause upregulation of ERβ and a dose-dependent reduction in NCI-H295R cell proliferation by increasing FasL, a proapoptotic figure [88]. Recent work by Sirianni and colleagues identified that ERα plays a critical role in IGF-II- and 17β-estradiol-dependent ACC cell proliferation. They showed that a selective estrogen antagonist reduced IGFR1 protein levels and decreased IGF-II and 17β-estradiol stimulated cell proliferation [89]. They also revealed significant tumor volume reduction with estrogen antagonist treatment in a mouse xenograft model. Tumor volume decreased by 46.7% compared with tumor size at the beginning of treatment and by 56.7% compared with control mice tumors [89]. These preclinical data provide an exciting new potential treatment pathway for ACC.
1α,25-dihydroxyvitamin D3
Several studies have shown that 1α,25-dihydroxyvitamin D3(1α,25[OH]2D3) can inhibit cell growth and affect tumor invasion and angiogenesis in several different cell lines [90-92]. Pilon et al. looked at the ACC cell line NCI-H295R and the effect 1α,25(OH)2 D3 had on cell proliferation and steroid secretion [93]. They found a 20% reduction in cell proliferation caused by cell arrest in G1 and identified a decrease in steroid production secondary to the antiproliferative effect of 1α,25(OH)2 D3 on the cells [93]. This early preclinical study may indicate further investigation of 1α,25(OH)2 D3 as a potential new treatment option for ACC.
Current ongoing trials
The advances in molecular profiling have led to an improved understanding of ACC pathogenesis and have resulted in new clinical trials for patients with ACC. Table 1 displays the current available trials that are actively recruiting patients at the time of writing this review. Updated statuses of clinical trials for patients with ACC are available at www.clinicaltrials.gov.
Table 1.
Clinical trial | NCT number | Primary outcome |
---|---|---|
Phase II Trial of Surgical Resection and Heated Intraperitoneal Peritoneal Chemotherapy (HIPEC) for Adrenocortical Carcinoma |
NCT01833832 | Intraperitoneal progressionfree survival |
Phase I Study of the Safety and Tolerability of ATR-101 in Adrenocortical Carcinoma |
NCT01898715 | Determine dose-limiting toxicity and maximum-tolerated dose |
A Phase I/II Trial of IL-13-PE in Patients With Treatment Refractory Malignancies With a Focus on Metastatic and Locally Advanced Adrenocortical Carcinoma |
NCT01832974 | Maximum-tolerated dose and objective response in Phase I, progression free survival in Phase II |
A Phase I/II Dose Escalation Study to Determine the Safety, Pharmacokinetics, and Pharmacodynamics of Intravenous TKM-080301 in Patients With Advanced Solid Tumors |
NCT01262235 | Determine dose-limiting toxicity, maximum-tolerated dose and antitumor activity |
Efficacy of Adjuvant Mitotane Treatment in Prolonging Recurrence-free Survival in Patients With Adrenocortical Carcinoma at Low-intermediate Risk of Recurrence (ADIUVO trial)† |
NCT00777244 | Disease-free survival |
European trial – no sites available in the USA.
Conclusion
Adrenocortical carcinoma is an aggressive tumor with limited treatment options resulting in a less than favorable outcome. Standard systemic therapies provide benefit to a small fraction of patients and the toxicities make these treatments less appealing to patients. Recent advances in the molecular biology of ACC have provided options for potential new targeted therapies. However, to date the clinical data are underwhelming. As we continue to discover the molecular mechanisms involved with ACC pathogenesis, new therapies and pathways are actively being investigated to provide hope for a new era of treatment in ACC.
Future perspective
The aggressiveness of ACC, limited therapeutic options and poor patient outcomes have compelled the medical research community to investigate the molecular biology of ACC to allow for better insight into the tumorigenesis of this disease. As our knowledge and understanding of ACC molecular pathways increase so will the number of potential therapeutic targets that are specific to the tumors, thus leading to more clinical trials and potentially a personalized approach to each individual tumor biology. Such an approach can result in higher efficacy and reduced toxicity. In addition, there is an urgent need to understand the role of adjuvant therapy in localized resected ACC. The results of the ADUIVO trial will hopefully provide guidance on this topic. We are hopeful that the drive for better understanding of ACC pathogenesis will lead to the development of new effective therapeutic treatments, particularly targeted therapy, better diagnostic and prognostic markers, and an overall improvement in patient outcomes.
Executive summary.
Mitotane, as a single agent or in combination with other cytotoxic drugs, is the current standard treatment for advanced adrenocortical carcinoma (ACC). Response rates are dismal at approximately 30%. The FIRM-ACT trial revealed that mitotane plus etoposide, doxorubicin and cisplatin had an improvement in progression-free survival (PFS) compared with mitotane plus streptozosin.
The role of mitotane in the adjuvant setting is controversial but the pending results of the ADIUVO trial may help clarify the role of mitotane in earlier stage disease.
Gene expression profiling has improved our understanding of the oncogenesis of ACC and helped identify potential new targets for treatment.
Several pathways have been identified in the tumorigenesis of ACC. IGF-2, mTOR, EGFR and VEGF are overexpressed in ACC. In vitro and in vivo studies have been preformed to identify potential targeted therapies for ACC.
β-catenin, steroidogenic factor-1, PPARγ and estrogen receptors have all been identified as potential markers for ACC tumorgenesis. Preclinical studies are currently ongoing to identify and validate new targeted drug treatments for ACC.
Footnotes
Financial & competing interests disclosure
The research activities performed in this manuscript were supported by the Intramural Research Program of the Center for Cancer Research, National Cancer Institute, National Institutes of Health. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.
No writing assistance was utilized in the production of this manuscript.
References
Papers of special interest have been highlighted as:
• of interest
- 1.De Reynies A, Assie G, Rickman DS, et al. Gene expression profiling reveals a new classification of adrenocortical tumors and identifies molecular predictors of malignancy and survival. J. Clin. Oncol. 2009;27(7):1108–1115. doi: 10.1200/JCO.2008.18.5678. [DOI] [PubMed] [Google Scholar]
- • Landmark study of gene-expression profiling for adrenocortical carcinoma (ACC).
- 2.Allolio B, Fassnacht M. Clinical review: Adrenocortical carcinoma: clinical update. J. Clin. Endocrinol. Metab. 2006;91(6):2027–2037. doi: 10.1210/jc.2005-2639. [DOI] [PubMed] [Google Scholar]
- 3.Luton JP, Cerdas S, Billaud L, et al. Clinical features of adrenocortical carcinoma, prognostic factors, and the effect of mitotane therapy. N. Engl. J. Med. 1990;322(17):1195–1201. doi: 10.1056/NEJM199004263221705. [DOI] [PubMed] [Google Scholar]
- 4.Fassnacht M, Kroiss M, Allolio B. Update in adrenocortical carcinoma. J. Clin. Endocrinol. Metab. 2013;98(12):4551–4564. doi: 10.1210/jc.2013-3020. [DOI] [PubMed] [Google Scholar]
- 5.Fassnacht M, Johanssen S, Quinkler M, et al. Limited prognostic value of the 2004 International Union Against Cancer staging classification for adrenocortical carcinoma: proposal for a Revised TNM Classification. Cancer. 2009;115(2):243–250. doi: 10.1002/cncr.24030. [DOI] [PubMed] [Google Scholar]
- 6.Schulick RD, Brennan MF. Long-term survival after complete resection and repeat resection in patients with adrenocortical carcinoma. Ann. Surg. Oncol. 1999;6(8):719–726. doi: 10.1007/s10434-999-0719-7. [DOI] [PubMed] [Google Scholar]
- 7.Fassnacht M, Johanssen S, Fenske W, et al. Improved survival in patients with stage II adrenocortical carcinoma followed up prospectively by specialized centers. J. Clin. Endocrinol. Metab. 2010;95(11):4925–4932. doi: 10.1210/jc.2010-0803. [DOI] [PubMed] [Google Scholar]
- 8.Erdogan I, Deutschbein T, Jurowich C, et al. The role of surgery in the management of recurrent adrenocortical carcinoma. J. Clin. Endocrinol. Metab. 2013;98(1):181–191. doi: 10.1210/jc.2012-2559. [DOI] [PubMed] [Google Scholar]
- • Recent study that confirms the importance of time to first recurrence and complete microscopic resection in outcome of recurrent ACC.
- 9.Datrice NM, Langan RC, Ripley RT, et al. Operative management for recurrent and metastatic adrenocortical carcinoma. J. Surg. Oncol. 2012;105(7):709–713. doi: 10.1002/jso.23015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Gaujoux S, Al-Ahmadie H, Allen PJ, et al. Resection of adrenocortical carcinoma liver metastasis: is it justified? Ann. Surg. Oncol. 2012;19(8):2643–2651. doi: 10.1245/s10434-012-2358-7. [DOI] [PubMed] [Google Scholar]
- 11.Op Den Winkel J, Pfannschmidt J, Muley T, et al. Metastatic adrenocortical carcinoma: results of 56 pulmonary metastasectomies in 24 patients. Ann. Thorac. Surg. 2011;92(6):1965–1970. doi: 10.1016/j.athoracsur.2011.07.088. [DOI] [PubMed] [Google Scholar]
- 12.Kemp CD, Ripley RT, Mathur A, et al. Pulmonary resection for metastatic adrenocortical carcinoma: the National Cancer Institute experience. Ann. Thorac. Surg. 2011;92(4):1195–1200. doi: 10.1016/j.athoracsur.2011.05.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Berruti A, Terzolo M, Sperone P, et al. Etoposide, doxorubicin and cisplatin plus mitotane in the treatment of advanced adrenocortical carcinoma: a large prospective Phase II trial. Endocr. Relat. Cancer. 2005;12(3):657–666. doi: 10.1677/erc.1.01025. [DOI] [PubMed] [Google Scholar]
- 14.Haak HR, Hermans J, Van De Velde CJ, et al. Optimal treatment of adrenocortical carcinoma with mitotane: results in a consecutive series of 96 patients. Br. J. Cancer. 1994;69(5):947–951. doi: 10.1038/bjc.1994.183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Baudin E, Pellegriti G, Bonnay M, et al. Impact of monitoring plasma 1,1-dichlorodiphenildichloroethane (o, p’DDD) levels on the treatment of patients with adrenocortical carcinoma. Cancer. 2001;92(6):1385–1392. doi: 10.1002/1097-0142(20010915)92:6<1385::aid-cncr1461>3.0.co;2-2. [DOI] [PubMed] [Google Scholar]
- 16.Gonzalez RJ, Tamm EP, Ng C, et al. Response to mitotane predicts outcome in patients with recurrent adrenal cortical carcinoma. Surgery. 2007;142(6):867–875. doi: 10.1016/j.surg.2007.09.006. discussion 867–875. [DOI] [PubMed] [Google Scholar]
- 17.Kerkhofs TM, Baudin E, Terzolo M, et al. Comparison of two mitotane starting dose regimens in patients with advanced adrenocortical carcinoma. J. Clin. Endocrinol. Metab. 2013;98(12):4759–4767. doi: 10.1210/jc.2013-2281. [DOI] [PubMed] [Google Scholar]
- 18.Khan TS, Imam H, Juhlin C, et al. Streptozocin and o,p’DDD in the treatment of adrenocortical cancer patients: long-term survival in its adjuvant use. Ann. Oncol. 2000;11(10):1281–1287. doi: 10.1023/a:1008377915129. [DOI] [PubMed] [Google Scholar]
- 19.Fassnacht M, Terzolo M, Allolio B, et al. Combination chemotherapy in advanced adrenocortical carcinoma. N. Engl. J. Med. 2012;366(23):2189–2197. doi: 10.1056/NEJMoa1200966. [DOI] [PubMed] [Google Scholar]
- • The first randomized controlled trial for combination treatment in advanced ACC. It also serves as the evidence guiding modern day treatment of advanced ACC.
- 20.Terzolo M, Angeli A, Fassnacht M, et al. Adjuvant mitotane treatment for adrenocortical carcinoma. N. Engl. J. Med. 2007;356(23):2372–2380. doi: 10.1056/NEJMoa063360. [DOI] [PubMed] [Google Scholar]
- • Important study that supports the role of adjuvant mitotane in ACC.
- 21.Grubbs EG, Callender GG, Xing Y, et al. Recurrence of adrenal cortical carcinoma following resection: surgery alone can achieve results equal to surgery plus mitotane. Ann. Surg. Oncol. 2010;17(1):263–270. doi: 10.1245/s10434-009-0716-x. [DOI] [PubMed] [Google Scholar]
- 22.Wangberg B, Khorram-Manesh A, Jansson S, et al. The long-term survival in adrenocortical carcinoma with active surgical management and use of monitored mitotane. Endocr. Relat. Cancer. 2010;17(1):265–272. doi: 10.1677/ERC-09-0190. [DOI] [PubMed] [Google Scholar]
- 23.Else T, Williams AR, Sabolch A, Jolly S, Miller BS, Hammer GD. Adjuvant therapies and patient and tumor characteristics associated with survival of adult patients with adrenocortical carcinoma. J. Clin. Endocrinol. Metab. 2014;99(2):455–461. doi: 10.1210/jc.2013-2856. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Abiven G, Coste J, Groussin L, et al. Clinical and biological features in the prognosis of adrenocortical cancer: poor outcome of cortisol-secreting tumors in a series of 202 consecutive patients. J. Clin. Endocrinol. Metab. 2006;91(7):2650–2655. doi: 10.1210/jc.2005-2730. [DOI] [PubMed] [Google Scholar]
- 25.Bertherat J, Coste J, Bertagna X. Adjuvant mitotane in adrenocortical carcinoma. N. Engl. J. Med. 2007;357(12):1256–1257. doi: 10.1056/NEJMc076267. author reply 1259. [DOI] [PubMed] [Google Scholar]
- 26.Terzolo M, Ardito A, Zaggia B, et al. Management of adjuvant mitotane therapy following resection of adrenal cancer. Endocrine. 2012;42(3):521–525. doi: 10.1007/s12020-012-9719-7. [DOI] [PubMed] [Google Scholar]
- 27.Assie G, Jouinot A, Bertherat J. The ‘omics’ of adrenocortical tumours for personalized medicine. Nat. Rev. Endocrinol. 2014;10(4):215–228. doi: 10.1038/nrendo.2013.272. [DOI] [PubMed] [Google Scholar]
- 28.Gicquel C, Bertagna X, Le Bouc Y. Recent advances in the pathogenesis of adrenocortical tumours. Eur. J. Endocrinol. 1995;133(2):133–144. doi: 10.1530/eje.0.1330133. [DOI] [PubMed] [Google Scholar]
- 29.Gicquel C, Bertagna X, Gaston V, et al. Molecular markers and long-term recurrences in a large cohort of patients with sporadic adrenocortical tumors. Cancer Res. 2001;61(18):6762–6767. [PubMed] [Google Scholar]
- 30.De Martino MC, Al Ghuzlan A, Aubert S, et al. Molecular screening for a personalized treatment approach in advanced adrenocortical cancer. J. Clin. Endocrinol. Metab. 2013;98(10):4080–4088. doi: 10.1210/jc.2013-2165. [DOI] [PubMed] [Google Scholar]
- 31.Velazquez-Fernandez D, Laurell C, Geli J, et al. Expression profiling of adrenocortical neoplasms suggests a molecular signature of malignancy. Surgery. 2005;138(6):1087–1094. doi: 10.1016/j.surg.2005.09.031. [DOI] [PubMed] [Google Scholar]
- 32.Giordano TJ, Kuick R, Else T, et al. Molecular classification and prognostication of adrenocortical tumors by transcriptome profiling. Clin. Cancer Res. 2009;15(2):668–676. doi: 10.1158/1078-0432.CCR-08-1067. [DOI] [PMC free article] [PubMed] [Google Scholar]
- • Landmark study on molecular classification and predictors of prognosis in adrenal tumors.
- 33.Ragazzon B, Libe R, Assie G, et al. Mass-array screening of frequent mutations in cancers reveals RB1 alterations in aggressive adrenocortical carcinomas. Eur. J. Endocrinol. 2014;170(3):385–391. doi: 10.1530/EJE-13-0778. [DOI] [PubMed] [Google Scholar]
- 34.Chabre O, Libe R, Assie G, et al. Serum miR-483-5p and miR-195 are predictive of recurrence risk in adrenocortical cancer patients. Endocr. Relat. Cancer. 2013;20(4):579–594. doi: 10.1530/ERC-13-0051. [DOI] [PubMed] [Google Scholar]
- 35.Patterson EE, Holloway AK, Weng J, Fojo T, Kebebew E. MicroRNA profiling of adrenocortical tumors reveals miR-483 as a marker of malignancy. Cancer. 2011;117(8):1630–1639. doi: 10.1002/cncr.25724. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Soon PS, Tacon LJ, Gill AJ, et al. miR-195 and miR-483-5p identified as predictors of poor prognosis in adrenocortical cancer. Clin. Cancer Res. 2009;15(24):7684–7692. doi: 10.1158/1078-0432.CCR-09-1587. [DOI] [PubMed] [Google Scholar]
- 37.Giordano TJ, Thomas DG, Kuick R, et al. Distinct transcriptional profiles of adrenocortical tumors uncovered by DNA microarray analysis. Am. J. Pathol. 2003;162(2):521–531. doi: 10.1016/S0002-9440(10)63846-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Slater EP, Diehl SM, Langer P, et al. Analysis by cDNA microarrays of gene expression patterns of human adrenocortical tumors. Eur. J. Endocrinol. 2006;154(4):587–598. doi: 10.1530/eje.1.02116. [DOI] [PubMed] [Google Scholar]
- 39.Ozata DM, Caramuta S, Velazquez-Fernandez D, et al. The role of microRNA deregulation in the pathogenesis of adrenocortical carcinoma. Endocr. Relat. Cancer. 2011;18(6):643–655. doi: 10.1530/ERC-11-0082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Tombol Z, Szabo PM, Molnar V, et al. Integrative molecular bioinformatics study of human adrenocortical tumors: microRNA, tissue-specific target prediction, and pathway analysis. Endocr. Relat. Cancer. 2009;16(3):895–906. doi: 10.1677/ERC-09-0096. [DOI] [PubMed] [Google Scholar]
- 41.West AN, Neale GA, Pounds S, et al. Gene expression profiling of childhood adrenocortical tumors. Cancer Res. 2007;67(2):600–608. doi: 10.1158/0008-5472.CAN-06-3767. [DOI] [PubMed] [Google Scholar]
- 42.Weber MM, Auernhammer CJ, Kiess W, Engelhardt D. Insulin-like growth factor receptors in normal and tumorous adult human adrenocortical glands. Eur. J. Endocrinol. 1997;136(3):296–303. doi: 10.1530/eje.0.1360296. [DOI] [PubMed] [Google Scholar]
- 43.Barlaskar FM, Spalding AC, Heaton JH, et al. Preclinical targeting of the type I insulin-like growth factor receptor in adrenocortical carcinoma. J. Clin. Endocrinol. Metab. 2009;94(1):204–212. doi: 10.1210/jc.2008-1456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Haluska P, Worden F, Olmos D, et al. Safety, tolerability, and pharmacokinetics of the anti-IGF-1R monoclonal antibody figitumumab in patients with refractory adrenocortical carcinoma. Cancer Chemother. Pharmacol. 2010;65(4):765–773. doi: 10.1007/s00280-009-1083-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Liu P, Cheng H, Roberts TM, Zhao JJ. Targeting the phosphoinositide 3-kinase pathway in cancer. Nat. Rev. Drug Discov. 2009;8(8):627–644. doi: 10.1038/nrd2926. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Hay N, Sonenberg N. Upstream and downstream of mTOR. Genes Dev. 2004;18(16):1926–1945. doi: 10.1101/gad.1212704. [DOI] [PubMed] [Google Scholar]
- 47.Doghman M, El Wakil A, Cardinaud B, et al. Regulation of insulin-like growth factor-mammalian target of rapamycin signaling by microRNA in childhood adrenocortical tumors. Cancer Res. 2010;70(11):4666–4675. doi: 10.1158/0008-5472.CAN-09-3970. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Wan X, Harkavy B, Shen N, Grohar P, Helman LJ. Rapamycin induces feedback activation of Akt signaling through an IGF-1R-dependent mechanism. Oncogene. 2007;26(13):1932–1940. doi: 10.1038/sj.onc.1209990. [DOI] [PubMed] [Google Scholar]
- 49.O’Reilly KE, Rojo F, She QB, et al. mTOR inhibition induces upstream receptor tyrosine kinase signaling and activates Akt. Cancer Res. 2006;66(3):1500–1508. doi: 10.1158/0008-5472.CAN-05-2925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Kurmasheva RT, Dudkin L, Billups C, Debelenko LV, Morton CL, Houghton PJ. The insulin-like growth factor-1 receptor-targeting antibody, CP-751,871, suppresses tumor-derived VEGF and synergizes with rapamycin in models of childhood sarcoma. Cancer Res. 2009;69(19):7662–7671. doi: 10.1158/0008-5472.CAN-09-1693. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Naing A, Kurzrock R, Burger A, et al. Phase I trial of cixutumumab combined with temsirolimus in patients with advanced cancer. Clin. Cancer Res. 2011;17(18):6052–6060. doi: 10.1158/1078-0432.CCR-10-2979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Naing A, Lorusso P, Fu S, et al. Insulin growth factor receptor (IGF-1R) antibody cixutumumab combined with the mTOR inhibitor temsirolimus in patients with metastatic adrenocortical carcinoma. Br. J. Cancer. 2013;108(4):826–830. doi: 10.1038/bjc.2013.46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Ganesan P, Piha-Paul S, Naing A, et al. Phase I clinical trial of lenalidomide in combination with temsirolimus in patients with advanced cancer. Invest. New Drugs. 2013;31(6):1505–1513. doi: 10.1007/s10637-013-0013-1. [DOI] [PubMed] [Google Scholar]
- 54.Kamio T, Shigematsu K, Sou H, Kawai K, Tsuchiyama H. Immunohistochemical expression of epidermal growth factor receptors in human adrenocortical carcinoma. Hum. Pathol. 1990;21(3):277–282. doi: 10.1016/0046-8177(90)90227-v. [DOI] [PubMed] [Google Scholar]
- 55.Sasano H, Suzuki T, Shizawa S, Kato K, Nagura H. Transforming growth factor alpha, epidermal growth factor, and epidermal growth factor receptor expression in normal and diseased human adrenal cortex by immunohistochemistry and in situ hybridization. Mod. Pathol. 1994;7(7):741–746. [PubMed] [Google Scholar]
- 56.Nakamura M, Miki Y, Akahira J, et al. An analysis of potential surrogate markers of target-specific therapy in archival materials of adrenocortical carcinoma. Endocr. Pathol. 2009;20(1):17–23. doi: 10.1007/s12022-009-9058-2. [DOI] [PubMed] [Google Scholar]
- 57.Adam P, Hahner S, Hartmann M, et al. Epidermal growth factor receptor in adrenocortical tumors: analysis of gene sequence, protein expression and correlation with clinical outcome. Mod. Pathol. 2010;23(12):1596–1604. doi: 10.1038/modpathol.2010.153. [DOI] [PubMed] [Google Scholar]
- 58.De Fraipont F, El Atifi M, Gicquel C, Bertagna X, Chambaz EM, Feige JJ. Expression of the angiogenesis markers vascular endothelial growth factor-A, thrombospondin-1, and platelet-derived endothelial cell growth factor in human sporadic adrenocortical tumors: correlation with genotypic alterations. J. Clin. Endocrinol. Metab. 2000;85(12):4734–4741. doi: 10.1210/jcem.85.12.7012. [DOI] [PubMed] [Google Scholar]
- 59.Bernini GP, Moretti A, Bonadio AG, et al. Angiogenesis in human normal and pathologic adrenal cortex. J. Clin. Endocrinol. Metab. 2002;87(11):4961–4965. doi: 10.1210/jc.2001-011799. [DOI] [PubMed] [Google Scholar]
- 60.Samnotra V, Vassilopoulou-Sellin R, Fojo AT, et al. A Phase II trial of gefitinib monotherapy in patients with unresectable adrenocortical carcinoma. J. Clin. Oncol. 2007;25(18 Suppl.):15527. [Google Scholar]
- 61.Quinkler M, Hahner S, Wortmann S, et al. Treatment of advanced adrenocortical carcinoma with erlotinib plus gemcitabine. J. Clin. Endocrinol. Metab. 2008;93(6):2057–2062. doi: 10.1210/jc.2007-2564. [DOI] [PubMed] [Google Scholar]
- 62.Wortmann S, Quinkler M, Ritter C, et al. Bevacizumab plus capecitabine as a salvage therapy in advanced adrenocortical carcinoma. Eur. J. Endocrinol. 2010;162(2):349–356. doi: 10.1530/EJE-09-0804. [DOI] [PubMed] [Google Scholar]
- 63.Hong DS, Sebti SM, Newman RA, et al. Phase I trial of a combination of the multikinase inhibitor sorafenib and the farnesyltransferase inhibitor tipifarnib in advanced malignancies. Clin. Cancer Res. 2009;15(22):7061–7068. doi: 10.1158/1078-0432.CCR-09-1241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Butler C, Butler WM, Rizvi AA. Sustained remission with the kinase inhibitor sorafenib in stage IV metastatic adrenocortical carcinoma. Endocr. Pract. 2010;16(3):441–445. doi: 10.4158/EP09295.CR. [DOI] [PubMed] [Google Scholar]
- 65.Berruti A, Sperone P, Ferrero A, et al. Phase II study of weekly paclitaxel and sorafenib as second/third-line therapy in patients with adrenocortical carcinoma. Eur. J. Endocrinol. 2012;166(3):451–458. doi: 10.1530/EJE-11-0918. [DOI] [PubMed] [Google Scholar]
- 66.Lee JO, Lee KW, Kim CJ, et al. Metastatic adrenocortical carcinoma treated with sunitinib: a case report. Jpn J. Clin. Oncol. 2009;39(3):183–185. doi: 10.1093/jjco/hyn146. [DOI] [PubMed] [Google Scholar]
- 67.Kroiss M, Quinkler M, Johanssen S, et al. Sunitinib in refractory adrenocortical carcinoma: a Phase II, single-arm, open-label trial. J. Clin. Endocrinol. Metab. 2012;97(10):3495–3503. doi: 10.1210/jc.2012-1419. [DOI] [PubMed] [Google Scholar]
- • Important Phase II trial that showed some tumor stability/response to tyrosine kinase inhibitors.
- 68.Kroiss M, Quinkler M, Lutz WK, Allolio B, Fassnacht M. Drug interactions with mitotane by induction of CYP3A4 metabolism in the clinical management of adrenocortical carcinoma. Clin. Endocrinol. 2011;75(5):585–591. doi: 10.1111/j.1365-2265.2011.04214.x. [DOI] [PubMed] [Google Scholar]
- 69.Chortis V, Taylor AE, Schneider P, et al. Mitotane therapy in adrenocortical cancer induces CYP3A4 and inhibits 5alpha-reductase, explaining the need for personalized glucocorticoid and androgen replacement. J. Clin. Endocrinol. Metab. 2013;98(1):161–171. doi: 10.1210/jc.2012-2851. [DOI] [PubMed] [Google Scholar]
- 70.Assie G, Giordano TJ, Bertherat J. Gene expression profiling in adrenocortical neoplasia. Mol. Cell. Endocrinol. 2012;351(1):111–117. doi: 10.1016/j.mce.2011.09.044. [DOI] [PubMed] [Google Scholar]
- 71.Ragazzon B, Assie G, Bertherat J. Transcriptome analysis of adrenocortical cancers: from molecular classification to the identification of new treatments. Endocr. Relat. Cancer. 2011;18(2):R15–R27. doi: 10.1530/ERC-10-0220. [DOI] [PubMed] [Google Scholar]
- 72.Tissier F, Cavard C, Groussin L, et al. Mutations of beta-catenin in adrenocortical tumors: activation of the Wnt signaling pathway is a frequent event in both benign and malignant adrenocortical tumors. Cancer Res. 2005;65(17):7622–7627. doi: 10.1158/0008-5472.CAN-05-0593. [DOI] [PubMed] [Google Scholar]
- 73.Kim AC, Barlaskar FM, Heaton JH, et al. In search of adrenocortical stem and progenitor cells. Endocr. Rev. 2009;30(3):241–263. doi: 10.1210/er.2008-0039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Gaujoux S, Grabar S, Fassnacht M, et al. Beta-catenin activation is associated with specific clinical and pathologic characteristics and a poor outcome in adrenocortical carcinoma. Clin. Cancer Res. 2011;17(2):328–336. doi: 10.1158/1078-0432.CCR-10-2006. [DOI] [PubMed] [Google Scholar]
- 75.Berthon A, Sahut-Barnola I, Lambert-Langlais S, et al. Constitutive beta-catenin activation induces adrenal hyperplasia and promotes adrenal cancer development. Hum. Mol. Genet. 2010;19(8):1561–1576. doi: 10.1093/hmg/ddq029. [DOI] [PubMed] [Google Scholar]
- 76.Doghman M, Cazareth J, Lalli E. The T cell factor/beta-catenin antagonist PKF115–584 inhibits proliferation of adrenocortical carcinoma cells. J. Clin. Endocrinol. Metab. 2008;93(8):3222–3225. doi: 10.1210/jc.2008-0247. [DOI] [PubMed] [Google Scholar]
- 77.Doghman M, Cazareth J, Douguet D, Madoux F, Hodder P, Lalli E. Inhibition of adrenocortical carcinoma cell proliferation by steroidogenic factor-1 inverse agonists. J. Clin. Endocrinol. Metab. 2009;94(6):2178–2183. doi: 10.1210/jc.2008-2163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Pianovski MA, Cavalli LR, Figueiredo BC, et al. SF-1 overexpression in childhood adrenocortical tumours. Eur. J. Cancer. 2006;42(8):1040–1043. doi: 10.1016/j.ejca.2006.01.022. [DOI] [PubMed] [Google Scholar]
- 79.Doghman M, Karpova T, Rodrigues GA, et al. Increased steroidogenic factor-1 dosage triggers adrenocortical cell proliferation and cancer. Mol. Endocrinol. 2007;21(12):2968–2987. doi: 10.1210/me.2007-0120. [DOI] [PubMed] [Google Scholar]
- 80.Sbiera S, Schmull S, Assie G, et al. High diagnostic and prognostic value of steroidogenic factor-1 expression in adrenal tumors. J. Clin. Endocrinol. Metab. 2010;95(10):E161–E171. doi: 10.1210/jc.2010-0653. [DOI] [PubMed] [Google Scholar]
- 81.Betz MJ, Shapiro I, Fassnacht M, Hahner S, Reincke M, Beuschlein F. Peroxisome proliferator-activated receptor-gamma agonists suppress adrenocortical tumor cell proliferation and induce differentiation. J. Clin. Endocrinol. Metab. 2005;90(7):3886–3896. doi: 10.1210/jc.2004-1267. [DOI] [PubMed] [Google Scholar]
- 82.Ferruzzi P, Ceni E, Tarocchi M, et al. Thiazolidinediones inhibit growth and invasiveness of the human adrenocortical cancer cell line H295R. J. Clin. Endocrinol. Metab. 2005;90(3):1332–1339. doi: 10.1210/jc.2004-0978. [DOI] [PubMed] [Google Scholar]
- 83.Blanquicett C, Roman J, Hart CM. Thiazolidinediones as anti-cancer agents. Cancer Ther. 2008;6(A):25–34. [PMC free article] [PubMed] [Google Scholar]
- 84.Cantini G, Lombardi A, Piscitelli E, et al. Rosiglitazone inhibits adrenocortical cancer cell proliferation by interfering with the IGF-IR intracellular signaling. PPAR Res. 2008 doi: 10.1155/2008/904041. 2008, 904041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Cerquetti L, Sampaoli C, Amendola D, et al. Rosiglitazone induces autophagy in H295R and cell cycle deregulation in SW13 adrenocortical cancer cells. Exp. Cell Res. 2011;317(10):1397–1410. doi: 10.1016/j.yexcr.2011.02.014. [DOI] [PubMed] [Google Scholar]
- 86.Luconi M, Mangoni M, Gelmini S, et al. Rosiglitazone impairs proliferation of human adrenocortical cancer: preclinical study in a xenograft mouse model. Endocr. Relat. Cancer. 2010;17(1):169–177. doi: 10.1677/ERC-09-0170. [DOI] [PubMed] [Google Scholar]
- 87.Barzon L, Masi G, Pacenti M, et al. Expression of aromatase and estrogen receptors in human adrenocortical tumors. Virchows Arch. 2008;452(2):181–191. doi: 10.1007/s00428-007-0542-0. [DOI] [PubMed] [Google Scholar]
- 88.Montanaro D, Maggiolini M, Recchia AG, et al. Antiestrogens upregulate estrogen receptor beta expression and inhibit adrenocortical H295R cell proliferation. J. Mol. Endocrinol. 2005;35(2):245–256. doi: 10.1677/jme.1.01806. [DOI] [PubMed] [Google Scholar]
- 89.Sirianni R, Zolea F, Chimento A, et al. Targeting estrogen receptor-alpha reduces adrenocortical cancer (ACC) cell growth in vitro and in vivo: potential therapeutic role of selective estrogen receptor modulators (SERMs) for ACC treatment. J. Clin. Endocrinol. Metab. 2012;97(12):E2238–E2250. doi: 10.1210/jc.2012-2374. [DOI] [PubMed] [Google Scholar]
- 90.Eelen G, Gysemans C, Verlinden L, et al. Mechanism and potential of the growth-inhibitory actions of vitamin D and ana-logs. Curr. Med. Chem. 2007;14(17):1893–1910. doi: 10.2174/092986707781058823. [DOI] [PubMed] [Google Scholar]
- 91.Yang L, Ma J, Zhang X, Fan Y, Wang L. Protective role of the vitamin D receptor. Cell. Immunol. 2012;279(2):160–166. doi: 10.1016/j.cellimm.2012.10.002. [DOI] [PubMed] [Google Scholar]
- 92.Hossein-Nezhad A, Spira A, Holick MF. Influence of vitamin D status and vitamin D3 supplementation on genome wide expression of white blood cells: a randomized double-blind clinical trial. PLoS ONE. 2013;8(3):e58725. doi: 10.1371/journal.pone.0058725. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Pilon C, Urbanet R, Williams TA, et al. 1alpha,25-dihydroxyvitamin D3 inhibits the human H295R cell proliferation by cell cycle arrest: a model for a protective role of vitamin D receptor against adrenocortical cancer. J. Steroid Biochem. Mol. Biol. 2014;140:26–33. doi: 10.1016/j.jsbmb.2013.11.008. [DOI] [PubMed] [Google Scholar]