Abstract
Deregulation of the IGF system observed in human tumors indicates a role in malignant cell transformation and in tumor cell proliferation. Although overexpression of the IGF2 and IGF1R genes was described in adrenocortical tumors (ACTs), few studies reported their profiles in pediatric ACTs. In this study, the IGF2 and IGF1R expression was evaluated by RT-qPCR according to the patient’s clinical/pathological features in 60 pediatric ACT samples, and IGF1R protein was investigated in 45 samples by immunohistochemistry (IHC). Whole transcriptome and functional assays were conducted after IGF1R inhibition with OSI-906 in NCI-H295A cell line. Significant IGF2 overexpression was found in tumor samples when compared with non-neoplastic samples (P<0.001), significantly higher levels of IGF1R in patients with relapse/metastasis (P=0.031) and moderate/strong IGF1R immunostaining in 62.2% of ACTs, but no other relationship with patient survival and clinical/pathological features was observed. OSI-906 treatment downregulated genes associated with MAPK activity, induced limited reduction of cell viability and increased the apoptosis rate. After 24h, the treatment also decreased the expression of genes related to the steroid biosynthetic process, the protein levels of the steroidogenic acute regulatory protein (STAR), and androgen secretion in cell medium, supporting the role of IGF1R in steroidogenesis of adrenocortical carcinoma cells. Our data showed that the IGF1R overexpression could be indicative of aggressive ACTs in children. However, in vitro treatments with high concentrations of OSI-906 (>1μM) showed limited reduction of cell viability, suggesting that OSI-906 alone could not be a suitable therapy to abolish carcinoma cell growth.
Introduction
Pediatric adrenocortical carcinoma (ACC) is a rare neoplasia with a worldwide incidence estimated at 0.2–0.3 cases/million children under 15 years per year (Else et al. 2014). The incidence in Southern of Brazil is 10–15 higher than the worldwide rate, which is related to the inherited germline TP53 mutation (p.R337H) (Ribeiro & Figueiredo 2004). Some of prognostic factors include older age, mitotic rate, tumor weight, tumor size, and presence of metastasis (Klein et al. 2011). Complete surgical resection remains the only treatment to achieve cure and long-term survival, which is less than 30% in patients with advanced stages of the disease (Fassnacht et al. 2011, Lorea et al. 2012). Adjuvant mitotane, chemotherapy and/or radiotherapy have been often recommended in order to reduce local recurrence. For palliative cases, the arterial chemoembolization, radiotherapy and radiofrequency ablation should also be considered (Berruti et al. 2012, Fassnacht et al. 2012, Else et al. 2014).
Among molecular markers, the overexpression of insulin growth factor 2 (IGF2) has been commonly found in pediatric tumors (Wilkin 2000, Lerario et al. 2014). Additionally, few studies have evaluated the insulin growth factor 1 receptor (IGF1R) gene expression (West et al. 2007, Almeida et al. 2008). The IGF signaling is activated by IGF1, IGF2, and/or insulin binding to the IGF1R, which autophosphorylates and begins downstream cascades such as PI3K and MAPK, promoting cell proliferation and differentiation and exerting anti-apoptosis effects and angiogenesis (Riedemann & Macaulay 2006, Pollak 2012). Although adrenal tumorigenesis involves several genetic abnormalities, the IGF2 overexpression seems to occur at an earlier stage of tumor formation, but it is unable to cause tumor formation alone (Assié et al. 2014, Pinto et al. 2015).
Evidences of the IGF pathway role in malignant cell transformation and proliferation have conducted to the study of IGF1R target-drugs. Among them, OSI-906 (Linsitinib) is a potent, oral, and selective inhibitor of the IGF1R and insulin receptor (IR) autophosphorylation, reducing cell proliferation in different types of cancer cell lines (Mulvihill et al. 2009). Recently, a large phase III trial in adults with advanced ACC demonstrated no differences in overall survival between patients treated with OSI-906 and the placebo group (Fassnacht et al. 2015, Kirschner 2015). Therefore, it remains to be established the real role of IGF1R in ACT and whether anti-IGF1R therapy alone or combined with other drugs is efficient as a preferred treatment.
In this study, we analyzed IGF1R and IGF2 gene expression profiles according to the clinical and pathological features of adrenocortical tumors in a large series of pediatric patients’ samples and investigated the effect of IGF1R inhibition by OSI-906 in the ACC cell line NCI-H295A as well as the altered downstream signaling pathways.
Subjects, material, and methods
Patients
A total of 60 pediatric adrenocortical tumor samples were obtained from the University Hospital, Ribeirao Preto Medical School, University of Sao Paulo, and Boldrini Children Center, Campinas. All patients underwent clinical and hormonal evaluation by biochemical and imaging investigation. Abdominal and chest CT and bone scintigraphy were conducted for metastasis detection at diagnosis and during follow-up. The disease stage at diagnosis was based on modified Sandrini’s classification of childhood ACTs (Michalkiewicz et al. 2004). Ten non-neoplastic adrenal samples were used as control, which were collected during nephrectomy due to Wilms’ tumor, before chemotherapy and from children without Beckwith–Wiedemann syndrome. The study was approved by the local Ethics Committees (protocol number: 8380/2010), and a signed statement of informed consent was obtained from the children’s parents.
The group of patients diagnosed with ACT consisted of 46 girls and 14 boys with a mean age at diagnosis of 40.5 months (range 5–187 months). Three patients had nonsecreting tumors, while 57 had hormone-secreting tumors (37 androgen, 18 mixed cortisol/androgen, and 2 cortisol). Thirty-five patients were classified as stage I, 10 as stage II, 8 as stage III, and 7 as stage IV. The germline TP53 p.R337H mutation was evaluated by direct genomic DNA sequencing and was detected in 52/60 (86.6%) patients. In most tumors, DNA was sequenced and the loss of heterozygosity (LOH) at the 17q locus was confirmed. The analysis of the entire coding and boundary regions of the TP53 gene revealed the absence of other mutations. Median follow-up was 68.3 months (range: 8–168 months). Twenty patients (33.3%) presented metastasis at diagnosis (n=7) or relapsed (n=13). The clinical and pathological features of these patients were described previously (Leal et al. 2011, Lorea et al. 2012, Gomes et al. 2014).
Real-time PCR (qPCR)
Tumor fragments were collected during surgical resection, frozen in liquid nitrogen, microdissected, and revised by a pathologist. Total RNA was isolated using Trizol reagent (Invitrogen) according to manufacturer’s instructions. The cDNA was generated from 1µg total RNA using the High Capacity Kit (Applied Biosystems). The human genes IGF2 (Hs01005963_m1), IGF1R (Hs00609566_m1), MAPK1 (Hs01046830_m1), MAPK3 (Hs00385075_m1), and PIK3R5 (Hs01046353_m1) were amplified by qRT-PCR using TaqMan gene assays and the ABI 7500 Real Time PCR System (Applied Biosystems). All samples were analyzed in triplicate and normalized to the endogenous reference human genes ACTB and GUSB (Applied Biosystems) as described previously (Leal et al. 2011, Leite et al. 2014). The relative expression was determined by the 2−ΔΔCT method (Livak & Schmittgen 2001), and the median gene expression values of all non-neoplastic adrenal tissues was used as reference and defined as 1 for analysis of tumor samples. For in vitro assays, samples were normalized to the GUSB gene, and untreated cells were used as reference samples.
Immunohistochemistry
Immunohistochemistry for IGF1R was performed by the avidin–biotin peroxidase complex (ABC) method (Novocastra, Newcastle-upon-Tyne, UK) in 45 tumor samples from the 60 patients evaluated in this study. Fifteen samples were excluded because they presented few or no tumor representative area anymore. A small group of 21 tumors was also evaluated for IGF2. The primary antibody was applied for overnight incubation (mouse monoclonal anti-IGF-1R, 1:300; Biocare Medical – CM 414 A. C.; Concord, CA, USA; mouse monoclonal anti-IGF2, 1:50; Santa Cruz Biotechnology, sc-74119) and a biotinylated secondary antibody was incubated. The visualization was performed with streptavidin peroxidase followed by diaminobenzidine coloring (Gibco) and Harris’ hematoxylin counterstaining. The positive control was a breast carcinoma sample (IGF1R) and placenta (IGF2) and the negative control was obtained by replacing the primary antibody with PBS 1X.
The immunohistochemistry analysis classified the samples as negative (no/weak staining) or positive (moderate/strong staining) according to the intensity of IGF1R/IGF2 staining. Regarding the localization, the samples were grouped into cytoplasmic, cytoplasmic/membrane, or nuclear staining.
Cell line and reagents
The human adrenocortical carcinoma cell line NCI-H295A was cultivated in RPMI medium as described previously (Gomes et al. 2014). Cell line authentication was conducted by examining CSF1PO, D13S317, D16S539, D5S818, D7S820, THO1, TPOX, vWA, and AMEL polymorphic loci by short-tandem repeat (STR) profiling. To avoid genetic drift or selection of variant subclones, all experiments were performed under standard cell culture conditions in an incubator at 37°C in a humidified atmosphere of 5% CO2 and cells were used at low passages (<10).
The OSI-906 inhibitor was acquired from Selleck Chemicals (LLC, Houston, TX, USA), dissolved in dimethyl sulfoxide (DMSO) at stock concentration of 10mM, and stored at −20°C. Control groups were prepared for all experiments using cells grown in medium with DMSO only.
Whole-transcriptome analysis
After NCI-H295A treatment with OSI-906 (2μM) for 6 and 24h, total cellular RNA was extracted using Trizol Reagent (Invitrogen) and stored in DEPC-treated water at −80°C, and the quantity and quality of samples was evaluated with an ND-1000 NanoDrop spectrophotometer (NanoDrop Products, Wilmington, DE, USA). The mRNA library was constructed using 200ng total RNA and investigated with the Whole Human Genome Microarray Kit, 4x44K (Agilent Technologies) to determine the gene expression profiles. Spot images were processed by Feature Extraction Software v10.7.3.1 (Agilent Technologies). All steps of quality evaluation, normalization, background correction, and statistical analysis were performed using R statistical language (Gentleman et al. 2004, R Development Core Team 2014) and the Bioconductor package Agi4x44PreProcess: PreProcessing of Agilent 4x44 array data (Lopez-romero 2012). After background correction, the samples were quantile normalized and the expression of the genes was obtained using a linear model fitted to each gene, so that the fold change between conditions and its associated errors could be estimated. Differentially expressed genes were obtained from those with lowest P-value and extreme values of fold-change. To annotate the differentially expressed genes (DEGs), EnricheR tool was used for gene ontology (GO) and KEGG pathway analysis with criterion of P-value<0.05 and at least three genes per process (Chen et al. 2013).
Cell viability assay
The resazurin reduction method was used to investigate cellular metabolic activity after treatment with OSI-906 (O’Brien et al. 2000). For the assay, the cells were treated with OSI-906 inhibitor at different concentrations (0.125–3μM) for 24, 48, or 72h. After the treatment period, resazurin (Sigma-Aldrich) was added and the plates were incubated for 4h at 37°C, 5% CO2. Absorbance at 570nm wavelength was then read with a reference wavelength of 595nm using an iMark Microplate Absorbance Reader (Bio-Rad Laboratories). The effects of OSI-906 in the cell viability were reported as mean ± s.d. of at least three independent experiments performed in triplicate. The EC50 values were calculated using Calcusyn software (Biosoft, Ferguson, MO, USA) (Chou & Talalay 1984).
Apoptosis assay
Apoptotic cell death was determined by labeling with annexin V fluorescein isothiocyanate (BD Biosciences Pharmigen, San Jose, CA, USA) and propidium iodide (PI) staining. Briefly, after 72h of OSI-906 treatment, 1.5×105 cells were trypsinized and centrifuged at 112 g for 5 min at 4°C, washed with ice-cold PBS 1X, and then resuspended in 350µL annexin V binding buffer (BD Biosciences Pharmigen, San Jose, CA, USA). Cells were stained with 5µL annexin V and 50µL PI 50µmol/L and incubated at room temperature in the dark. The samples were analyzed using a BD FACSCalibur flow cytometer (BD Biosciences Pharmigen, San Jose, CA, USA).
Cell lysis and western blot analysis
After treatment with OSI-906 (2μM) for 3, 6, or 24h, cells were lysed in RIPA buffer (Sigma-Aldrich) in the presence of protease and phosphatase inhibitors. Equal amounts of whole-cell lysates were resolved by 12% SDS–polyacrylamide gel electrophoresis (PAGE) followed by transfer to nitrocellulose membranes (GE Healthcare), which were incubated in Tris-buffered saline and 0.1% Tween-20 containing 5% (w/v) dried nonfat milk for 1h at room temperature and probed with appropriately diluted primary antibodies overnight at 4°C. Blots were incubated with biotin-labeled horseradish peroxidase-conjugated species-specific secondary antibodies (1:10,000; GE Healthcare) followed by chemiluminescence detection using the ECL Western blotting Analysis System Kit (Amersham GE Healthcare) and ChemiDoc System (Bio-Rad Laboratories).
Rabbit monoclonal antibodies against phospho- and total-ERK1/2 were obtained from Cell Signaling (respectively, #9101 and #9102; both 1:800). A rabbit polyclonal antibody against STAR was purchased from Santa Cruz Biotechnology (1:200; sc-25806) and a mouse monoclonal antibody against GAPDH from Santa Cruz Biotechnology was used as loading control (1:1000; sc-47724).
Hormone measurements
After 24h of OSI-906 treatment (2μM), the supernatant medium was collected from each well before cell lysis for RNA or protein extraction and stored at −80°C until hormone quantification. Dehydroepiandrosterone sulfate (DHEAS), D4-androstenedione (D4), and testosterone concentrations were determined by radioimmunoassay (RIA) as described previously (Moreira & Elias 1992). All measurement of hormones from the NCI-H295A medium was conducted in triplicate and the mean was normalized to the cell viability effects in the same treatment conditions.
Evaluable data from circulating hormone concentrations (Cortisol, DHEAS, D4, Testosterone) of patient before tumor treatment was used for correlations analysis.
Statistical analyses
The expression of IGF2 and IGF1R genes according to following variables: age (<versus≥4 years), tumor weight (<versus≥100g), tumor size (<versus≥200cm3), TP53 p.R377H mutation (positive vs negative), and disease stage was compared by the Mann–Whitney test. Differences between the expression values are reported as fold change (FC) by dividing the median expression values for each variable analyzed. Event-free survival (EFS) analysis (with relapse and/or death due to any cause being considered as unfavorable events) was carried out based on Kaplan–Meier curves, using the gene expression median values from ACTs as the cut-off point for IGF2 and IGF1R. The curves for different groups were compared by the log-rank test. Correlations were determined using the Spearman correlation coefficient (ρ).
Data of the functional assays such as cell viability and apoptosis, as well as the gene expression after OSI-906 treatment, were evaluated by one-way ANOVA with the Bonferroni post-test. Moreover, differences of hormone concentrations in the cell medium were analyzed by the t-test for equality of means. All analyses were carried out using the IBM SPSS Statistics software version 20 (SPSS), with the level of significance set at 0.05.
Results
IGF2 gene is overexpressed in adrenocortical tumors
A significant overexpression of IGF2 was detected in ACT samples when compared with non-neoplastic adrenal tissue, (15.9-fold, P<0.001) (Fig. 1A); however, IGF2 overexpression was not related to relapse or metastasis in the patients (P=0.243), neither with clinical–pathological features nor with 5-year EFS (P=0.753).
IGF1R gene expression is higher in patients with relapse and metastasis
Although IGF1R gene expression was very similar for non-neoplastic adrenal samples and ACTs (P=0.822), we observed significant overexpression of IGF1R in patients with tumor relapse or metastasis when compared with the group of patients with complete remission (2.7-fold, P=0.031) (Fig. 1B). However, IGF1R overexpression (1× or 2×>median) was not indicative of significant differences in the EFS of patients with ACT (58.7±13.0% vs 80.6±6.2%, 2×>median; P=0.103). Interestingly, we found significant positive correlation between IGF1R gene expression and DHEAS concentrations in patient’s serum (ρ=0.663; P=0.02).
Transcripts do not correlate with protein expression
The IGF1R protein expression evaluated by IHC did not agree with the gene expression profiles observed by qRT-PCR. Positive immunoreactivity was found in 32/45 (71.1%) of the ACT samples, with 16 cases (35.5%) classified as strong, 12 (26.7%) as moderate, and 4 (8.9%) as weak. The remaining 13 samples (28.9%) as well as the non-neoplastic tissue adjacent to the tumor presented no IGF1R staining (Fig. 2). Moderate or strong immunostaining were considered as IGF1R positive, while samples with weak or absent staining were classified as IGF1R negative. The median of IGF1R mRNA expression was 0.58 for IHC-positive samples (mean: 1.46, range: 0.10–8.26) and 0.55 for IHC-negative samples (mean: 1.23, range: 0.17–4.88) (P=0.933). Cytoplasmic staining was observed in 26 tumor samples, cytoplasmic and focal membrane staining was present in 4 samples, and focal nuclear staining was detected in 2 cases. There was no significant difference in 5-year EFS between patients with moderate/strong and negative/weak IGF1R immunoreactivity (P=0.613) or with pure cytoplasmic versus membranous/nuclear staining (P=0.726).
Additionally, the IHC for IGF2 showed no correlation with transcripts, being 43% of negative cases, 43% with weak staining, 10% with moderate, and 5% with strong staining. The positive samples presented predominantly cytoplasmatic staining (data not shown), and no significant differences were found according to the clinical and pathological features.
Identification of differentially expressed genes after treatment of adrenocortical carcinoma cells with OSI-906
In order to investigate the role of IGF1R in adrenocortical carcinoma cells (NCI-H295A), the global gene expression by microarray analysis was performed after functional blockage of IGF1R with OSI-906. After normalization, the 2000 most differentially expressed genes (DEGs) at two time points during treatment (6 and 24h) were selected for further analysis. Biological processes (gene ontology (GO)) and pathway mappings (KEGG) were evaluated for the 200 most upregulated and the 200 most downregulated genes for each treatment time. The results demonstrated that upregulated DEGs were significantly enriched in the GO factors involved in biological processes such as cytoskeleton organization and NFKB cascade, while the downregulated DEGs were enriched in hormone metabolic/biosynthetic processes and MAPK activity.
We next evaluated whether treatment with OSI-906 could also impair time-dependent changes in the biological processes. At both times, the upregulated DEGs were enriched in GO terms such as activation/positive regulation of caspases and cell cycle regulation/arrest, while one KEGG pathway was identified only at 6h of treatment (HSA04115 – P53 signaling pathway; P=0.03). On the other hand, the group of downregulated DEGs were associated with positive regulation/activation of the MAPK pathway and cell motility at 6h (Fig. 3A) and, significantly enriched in lipid and hormone synthesis/metabolism, response to external stimuli and lipid metabolic processes (Fig. 3B) at 24h. The downregulated DEGs were also enriched in KEGG pathways related to hormone metabolism (Table 1), supporting the findings of the biological processes enrichment. Moreover, differential expression of specific genes such as higher expression of DEPTOR (FC=1.4) and IRS2 (FC=1.4) at 24h, as well as reduction in PPARGC1B (FC=−1.6) at 24h and PIK3C2G (FC=−1.4) at 6h suggests inactivation of mTOR signaling downstream IGF1R.
KEGG Pathways enrichment of downregulated DEGs after 24h of OSI-906 treatment.
Term | Gene count | P-value | Z-score | Combined score | Genes |
---|---|---|---|---|---|
HSA00140 – C21 steroid hormone metabolism | 5 | 9.65×10−7 | −1.62 | 15.58 | HSD3B2; HSD3B1; CYP21A2; CYP11B1; CYP11B2 |
HSA00330 – arginine and proline metabolism | 4 | 0.001 | −1.88 | 5.84 | CKB; CKMT2; EPRS; OAT |
HSA00150 – androgen and estrogen metabolism | 4 | 0.006 | −2.00 | 4.10 | CYP11B1; CYP11B2; HSD3B2; HSD3B1 |
HSA03320 – PPAR signaling pathway | 4 | 0.013 | −1.72 | 2.92 | APOA1; ACSL6; FADS2; FABP6 |
HSA01510 – neurodegenerative diseases | 3 | 0.014 | −1.57 | 2.66 | UCHL1; NEFH; PRNP |
HSA01040 – polyunsaturated fatty acid biosynthesis | 2 | 0.016 | −0.50 | 0.86 | FADS2; ELOVL5 |
HSA00642 – ethylbenzene degradation | 2 | 0.018 | −0.61 | 1.04 | ESCO2; DHRS2 |
HSA00624 – 1 and 2 methylnaphthalene degradation | 2 | 0.044 | −1.00 | 0.96 | ESCO2; DHRS2 |
Confirming the microarray analysis by qRT-PCR, the cells treated with OSI-906 showed significant reduction in MAPK1 at 6h and PI3K at both times. MAPK3 expression was slightly lower at 6h and higher at 24h (not significant) (Fig. 4A and C). Moreover, the treated cells showed reduction in phosphorylated and total ERK1/2 proteins when compared with untreated cells (Fig. 4D).
OSI-906 reduces tumor cell viability and induces apoptosis
Treatments with 1, 2, and 3μM of OSI-906 for 24h decreased cell viability by 13, 15 and 18%, respectively, while at 48 and 72h, the reduction was significant with all doses of treatment (0.125–3μM) (P<0.001). However, limited reductions were observed at concentrations beyond 0.5μM, which were characterized by a plateau in the graph of cell viability (Fig. 4E). The most effective reduction was observed at 72h with 1μM (40%). The time-dependent effect was confirmed with distinct EC50 (>20±15.2μM for 24h; 2.64±0.23μM for 48h; 1.99±0.78μM for 72h). We also observed a significant dose-dependent increase in cell apoptosis rate at 72h, reaching 31% of apoptotic cells with 1μM of OSI-906 (CI: 62.4–79.3; P<0.001) (Fig. 4F).
Treatment with OSI-906 reduces hormone biosynthesis in adrenocortical carcinoma cells
To validate the microarray findings concerning the reduction in genes associated with hormone synthesis and lipid metabolism, we investigated the effects of OSI-906 on cellular hormone production after 24h. All steroid hormones were reduced, but it was significant only to testosterone (P=0.02) (Fig. 5A and C). Interestingly, STAR protein expression decreased 55% at 24h (Fig. 5D).
Discussion
The characteristics of our samples regarding clinical data such as age, sex, tumor stage, clinical symptoms, survival, and prognostic factors were similar to those described by the International Pediatric Adrenocortical Tumor Registry (IPACTR) (Michalkiewicz et al. 2004).
One hallmark of pediatric ACT in Brazil is the TP53 p.R337H germline mutation, which disrupts protein tetramer formation and reduces the p53 activity in higher pH levels (Wasserman et al. 2012). Interestingly, in response to cellular stress such as oncogene activation, the p53 can shutdown the IGF pathway, reducing IGF2 and IGF1R expression and the receptor tyrosine phosphorylation (Sampaoli et al. 2012). Similar to other Brazilian childhood ACT series (Sandrini et al. 2005, Custódio et al. 2013), we observed high frequency of TP53 p.R337H mutation in tumor samples, but no significant differences in IGF2 or IGF1R expression profiles between samples with or without the mutation.
In vitro studies have shown that apoptosis and inhibition of ACC cells growth after ionizing radiation is dependent of p53 protein stabilization (Sampaoli et al. 2012), and TP53 somatic mutations confer resistance for anti-IGF1R therapy in colorectal carcinoma cells (Wang et al. 2013). Interestingly, NCI-H295 lacks TP53 p.R337H, but presents other mutations affecting p53 functions (Sampaoli et al. 2012, Leal et al. 2015), which also could be related to reduced effectiveness of therapies, such as anti-IGF1R.
The role of the IGF system in the development and growth of adrenal cortex is well known, with high levels of IGF2 detected in the adrenal glands and serum during the fetal stage, followed by a strong decline during the postnatal period. However, overexpression of IGF2 has been associated with a higher risk of ACT recurrence in adults (Boulle et al. 1998, Fottner et al. 2004). In children, we observed that IGF2 overexpression was not associated with tumor recurrence or disease poor outcome, which is in agreement with a few studies reporting IGF2 higher expression in pediatric ACT (Wilkin 2000, West et al. 2007, Almeida et al. 2008).
Among the four receptors of the IGF pathway described in mammals, the tyrosine kinase receptor type 1 (IGF1R) originates signals that facilitate cell transformation by other agents in different types of tumors (Wang & Sun 2002). In agreement with its pivotal role in cell growth and homeostasis, the overexpression of IGF1R in several human cancers is not surprising and suggests the involvement of IGF1R in tumor growth and progression (Maki 2010). In this study, IGF1R expression was quite similar for non-neoplastic and tumor samples, but patients with metastasis or relapse showed significantly higher IGF1R expression levels. Similar to our findings, Almeida and coworkers (2008) observed IGF1R overexpression in adrenocortical carcinomas and significant association with higher risk of metastasis in a study with 23 pediatric ACT samples. Herein, all control samples consisted of non-neoplastic adrenal tissue with both cortical and medullary cells, which somehow could be a potential bias for the analysis.
The correlation between correspondent IGF1R and IGF2 protein expression and their transcripts were not significant in the evaluated ACT samples. Other studies on cancer have revealed that the lack of correlation between mRNA and protein levels can be attributed to mRNA translational silencing, cleavage, or alternative splicing (Dziadziuszko et al. 2010, Mountzios et al. 2013). In the IGF1R context, alternative mRNA splicing can lead to distinct protein degradation rates in cancer cells (Mountzios et al. 2013) while the frequently expressed isoform (alpha) of IGF1R is not directly associated with global IGF1R gene expression (Pollak 2012). Interestingly, reports have demonstrated the impact of IGF1R protein localization on the biological behavior and prognosis of human breast cancer and in clear renal cancer cells (Aleksic et al. 2010, Tamimi et al. 2011). The ACT samples presented more IGF1R pure cytoplasmic than mixed cytoplasmic/membranous/nucleus staining, but no significant differences were observed regarding patients’ survival, suggesting that IGF1R localization has no prognostic relevance for pediatric ACTs.
Evidence of IGF signaling in malignant cell transformation and tumor cell proliferation has encouraged the development of many IGF1R target-drugs. Among them, OSI-906 is known to reduce tumor cells proliferation because of its selective effect on both IGF1R and IR (Mulvihill et al. 2009). According to the global gene expression analysis of NCI-H295A cells, the inhibition of IGF1R with OSI-906 seems to upregulate caspases activity and to induce cell cycle arrest. Gene array analysis conducted on a broad panel of colorectal cancer cell lines revealed that OSI-906-sensitive cells present upregulation of the P53 pathway, while resistant cells present MAPK pathway upregulation (Pitts et al. 2010). At 6h of OSI-906 treatment, we observed the upregulation of P53 signaling pathway genes and reduction of genes associated with MAPK pathway activation, suggesting a sensitive profile of the NCI-H295A cell line. In fact, it was obtained a significant dose-dependent increasing of apoptosis rate (31%), but cell viability was limited to 40% even after high doses of OSI-906. Since resistant colorectal cancer cells present upregulation of the WNT pathway (Pitts et al. 2010), the mild decrease of cell viability in NCI-H295A cells could be related to the constitutive activation of Wnt/β-catenin signaling due to S45P mutation in this cell line (Tadjine et al. 2008).
Reduction of cell viability after treatment with OSI-906 has been frequently reported in different types of cancer cells. In this study, we used doses between 0.125 and 3μM, which are comparable with other preclinical studies and are also within the range of human maximal plasma concentrations (Cmax from 1.705 to 3.110μM) after oral administration of OSI-906 (150mg of OSI-906 twice daily) (Fassnacht et al. 2015, Puzanov et al. 2015). Sensitive cells, including the ACC cell line NCI-H295R, usually present EC50<1µM at 72h (Buck et al. 2010, Zhao et al. 2012, Janku et al. 2013). Surprisingly, we found higher EC50 values (>20μM at 24h, 2.6µM at 48h and 2.0µM at 72h) which, together with the restoration of ERK-1/2 expression after 24h of treatment, suggest a resistant profile for NCI-H295A after long periods of treatment (Zinn et al. 2013). Since OSI-906 inhibits kinase activities from both IR and IGF1R, we ruled out a resistance mechanism through compensatory IR signaling (Buck et al. 2010). However, new functions for IGF1R have been described (Boucher et al. 2010, Janku et al. 2013), which can explain the significant increase in apoptosis with only a mild decrease in NCI-H295A viability observed after OSI-906 treatment. Janku and coworkers (2013) showed that IGF1R is able to keep intracellular glucose levels, supporting tumor cell survival independent of their kinase activity. Moreover, IGF1R interacts with other receptor tyrosine kinases such as epidermal growth factor receptors (EGFRs), vascular endothelial growth factor receptor (VEGFR), mesenchymal–epithelial transition factor (MET), platelet-derived growth factor receptor (PDGFR), estrogen receptors (ER), and others, which are frequently found upregulated in cancer cells and may play a role in resistance to therapies anti-IGF1R. The activation of common downstream effectors through other receptors has provided new approaches regarding cotargeting strategies for anticancer therapies (Singh et al. 2014, Brahmkhatri et al. 2015).
In addition, OSI-906 reduced PIK3C2G (catalytic subunit type 2 Gamma of PI3K) gene expression in the microarray analysis and the PI3K gene by qPCR. The PI3 kinases family transduces signals from various growth factors such as IGF-IGF1R, which activates downstream mTOR (mammalian target of rapamycin) signals (Liu et al. 2009). Activation of mTOR is induced by two complexes (TSC1 and TSC2), driving cancer cells growth and proliferation (Advani 2010). After OSI-906 treatment, the cells also expressed higher levels of DEPTOR gene, a negative regulator of mTORC1 and mTORC2, as well as lower expression of PPARGC1B and increased expression of IRS2, which, together with PI3K reduction, suggest the inactivation of mTOR pathway (Laplante & Sabatini 2009, Brouwer-Visser & Huang 2015).
Adrenocortical tumors are frequently characterized by hormonal secretion, inducing clinical symptoms of the disease or revealing tumor recurrence during follow-up (Gönç et al. 2014). As expected, most of the children evaluated presented pure androgen or mixed (androgen and cortisol) secreting tumors, while only two patients presented cortisol secretion alone. In vitro studies have shown that IGF1/IGF1R stimulates hormone synthesis in different steroidogenic cells by inducing the expression of steroidogenic genes and the steroidogenic acute regulatory (STAR) protein through MAPK/ERK signaling (Ramanjaneya et al. 2011). In agreement, we found significant positive correlation between IGF1R gene expression and DHEAS levels in diagnosis patient’s serum. Moreover, cells treated with OSI-906 presented lower expression of genes related to hormone synthesis/metabolism and STAR protein impairment of 55%, which is considered the first key mediator of steroidogenesis (Samandari et al. 2007).
To a lesser extent, only testosterone concentration was significantly reduced after OSI-906 treatment, suggesting that steroidogenesis impairment STARted with lower STAR protein expression could not be sufficient to exert downstream effects in the synthesis of all steroid hormones at 24h.
In summary, IGF2 gene overexpression in a relatively large series of children diagnosed with ACT was not related to any clinical or biological features analyzed here, while IGF1R gene expression was significantly higher in children who presented tumor relapse and metastasis, which was not true for the IGF1R protein expression analyzed by IHC. In vitro blockage of IGF1R signaling downregulated MAPK activity, causing reduction in cellular viability and increase in apoptosis rate in a dose-dependent manner. In addition, OSI-906 decreased the expression of genes related to the steroid biosynthetic process and impaired the expression of STAR, a key steroidogenic enzyme, events that were followed by the reduction in testosterone production. These findings suggest that IGF1R could have a role in adrenocortical cancer; however, its inhibition by OSI-906 in adrenocortical tumor cells seems to promote only a mild antitumoral effect in a similar way as observed in clinical trials. Thus, new studies which elucidate the mechanisms of resistance to OSI-906 as well as new therapy schedules, such as drugs combination, would be important for a clinical application of IGF1R as a therapeutic target in childhood adrenocortical cancer.
Declaration of interest
All authors declare that they had no conflict of interest that could be perceived to impair the impartiality of the research reported.
Funding
This work was supported by the Public Research Agencies: Fundação de Amparo à Pesquisa do Estado de São Paulo (FAPESP), Grant number: 2010/07020-9; Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq) and Fundação de Apoio ao Ensino, Pesquisa e Assistência do Hospital das Clínica – FMRP/USP (FAEPA), Brazil.
Acknowledgments
The authors thank Dr Aguinaldo Luiz Simões for providing the authentication of the cell line.
References
Advani SH 2010 Targeting mTOR pathway: a new concept in cancer therapy. Indian Journal of Medical and Paediatric Oncology 31 132–136. (doi:10.4103/0971-5851.76197)
Aleksic T, Chitnis MM, Perestenko OV, Gao S, Thomas PH, Turner GD, Protheroe AS, Howarth M & Macaulay VM 2010 Type 1 insulin-like growth factor receptor translocates to the nucleus of human tumor cells. Cancer Research 70 6412–6419. (doi:10.1158/0008-5472.CAN-10-0052)
Almeida MQ, Fragoso MCBV, Lotfi CFP, Santos MG, Nishi MY, Costa MHS, Lerario AM, Maciel CC, Mattos GE & Jorge AAL et al. 2008 Expression of insulin-like growth factor-II and its receptor in pediatric and adult adrenocortical tumors. Journal of Clinical Endocrinology & Metabolism 93 3524–3531. (doi:10.1210/jc.2008-0065)
Assié G, Letouzé E, Fassnacht M, Jouinot A, Luscap W, Barreau O, Omeiri H, Rodriguez S, Perlemoine K & René-Corail F et al. 2014 Integrated genomic characterization of adrenocortical carcinoma. Nature Genetics 46 607–612. (doi:10.1038/ng.2953)
Berruti A, Baudin E, Gelderblom H, Haak HR, Porpiglia F, Fassnacht M & Pentheroudakis G 2012 Adrenal cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Annals of Oncology 23 (Supplement 7) vii131–vii138.(doi:10.1093/annonc/mds231)
Boucher J, Macotela Y, Bezy O, Mori MA, Kriauciunas K & Kahn CR 2010 A kinase-independent role for unoccupied insulin and IGF-1 receptors in the control of apoptosis. Science Signaling 3 ra87. (doi:10.1126/scisignal.2001173)
Boulle N, Logié A, Gicquel C, Perin L & Le Bouc Y 1998 Increased levels of insulin-like growth factor II (IGF-II) and IGF-binding protein-2 are associated with malignancy in sporadic adrenocortical tumors. Journal of Clinical Endocrinology & Metabolism 83 1713–1720. (doi:10.1210/jc.83.5.1713)
Brahmkhatri VP, Prasanna C & Atreya HS 2015 Insulin-like growth factor system in cancer: novel targeted therapies. BioMed Research International 2015 article ID 538019. (doi:10.1155/2015/538019)
Brouwer-Visser J & Huang GS 2015 IGF2 signaling and regulation in cancer. Cytokine & Growth Factor Reviews 26 371–377. (doi:10.1016/j.cytogfr.2015.01.002)
Buck E, Gokhale PC, Koujak S, Brown E, Eyzaguirre A, Tao N, Rosenfeld-Franklin M, Lerner L, Chiu MI & Wild R et al. 2010 Compensatory insulin receptor (IR) activation on inhibition of insulin-like growth factor-1 receptor (IGF-1R): rationale for cotargeting IGF-1R and IR in cancer. Molecular Cancer Therapeutics 9 2652–2664. (doi:10.1158/1535-7163.MCT-10-0318)
Chen EY, Tan CM, Kou Y, Duan Q, Wang Z, Meirelles GV, Clark NR & Ma’ayan A 2013 Enrichr: interactive and collaborative HTML5 gene list enrichment analysis tool. BMC Bioinformatics 14 128. (doi:10.1186/1471-2105-14-128)
Chou TC & Talalay P 1984 Quantitative analysis of dose-effect relationships: the combined effects of multiple drugs or enzyme inhibitors. Advances in Enzyme Regulation 22 27–55. (doi:10.1016/0065-2571(84)90007-4)
Custódio G, Parise GA, Kiesel Filho N, Komechen H, Sabbaga CC, Rosati R, Grisa L, Parise IZS, Pianovski MAD & Fiori CMCM et al. 2013 Impact of neonatal screening and surveillance for the TP53 R337H mutation on early detection of childhood adrenocortical tumors. Journal of Clinical Oncology 31 2619–2626. (doi:10.1200/JCO.2012.46.3711)
Dziadziuszko R, Merrick DT, Witta SE, Mendoza AD, Szostakiewicz B, Szymanowska A, Rzyman W, Dziadziuszko K, Jassem J & Bunn PA et al. 2010 Insulin-like growth factor receptor 1 (IGF1R) gene copy number is associated with survival in operable non-small-cell lung cancer: a comparison between IGF1R fluorescent in situ hybridization, protein expression, and mRNA expression. Journal of Clinical Oncology 28 2174–2180. (doi:10.1200/JCO.2009.24.6611)
Else T, Kim AC, Sabolch A, Raymond VM, Kandathil A, Caoili EM, Jolly S, Miller BS, Giordano TJ & Hammer GD 2014 Adrenocortical carcinoma. Endocrine Reviews 35 282–326. (doi:10.1210/er.2013-1029)
Fassnacht M, Libé R, Kroiss M & Allolio B 2011 Adrenocortical carcinoma: a clinician’s update. Nature Reviews. Endocrinology 7 323–335. (doi:10.1038/nrendo.2010.235)
Fassnacht M, Terzolo M, Allolio B, Baudin E, Haak H, Berruti A, Welin S, Schade-Brittinger C, Lacroix A & Jarzab B et al. 2012 Combination chemotherapy in advanced adrenocortical carcinoma. New England Journal of Medicine 366 2189–2197. (doi:10.1056/NEJMoa1200966)
Fassnacht M, Berruti A, Baudin E, Demeure MJ, Gilbert J, Haak H, Kroiss M, Quinn DI, Hesseltine E & Ronchi CL et al. 2015 Linsitinib (OSI-906) versus placebo for patients with locally advanced or metastatic adrenocortical carcinoma: a double-blind, randomised, phase 3 study. Lancet. Oncology 16 426–435. (doi:10.1016/S1470-2045(15)70081-1)
Fottner C, Hoeflich A, Wolf E & Weber MM 2004 Role of the insulin-like growth factor system in adrenocortical growth control and carcinogenesis. Hormone and Metabolic Research=Hormon- Und Stoffwechselforschung=Hormones et Métabolisme 36 397–405. (doi:10.1055/s-2004-814563)
Gentleman RC, Carey VJ, Bates DM, Bolstad B, Dettling M, Dudoit S, Ellis B, Gautier L, Ge Y & Gentry J et al. 2004 Bioconductor: open software development for computational biology and bioinformatics. Genome Biology 5 R80. (doi:10.1186/gb-2004-5-10-r80)
Gomes DC, Leal LF, Mermejo LM, Scrideli CA, Martinelli CE, Fragoso MCBV, Latronico AC, Tone LG, Tucci S & Yunes JA et al. 2014 Sonic hedgehog signaling is active in human adrenal cortex development and deregulated in adrenocortical tumors. Journal of Clinical Endocrinology & Metabolism 99 E1209–E1216. (doi:10.1210/jc.2013-4098)
Gönç EN, Özön ZA, Çakır MD, Alikaşifoğlu A & Kandemir N 2014 Need for comprehensive hormonal workup in the management of adrenocortical tumors in children. Journal of Clinical Research in Pediatric Endocrinology 6 68–73. (doi:10.4274/Jcrpe.1351)
Janku F, Huang HJ, Angelo LS & Kurzrock R 2013 A kinase-independent biological activity for insulin growth factor-1 receptor (IGF-1R) : implications for inhibition of the IGF-1R signal. Oncotarget 4 463–473. (doi:10.18632/oncotarget)
Kirschner LS 2015 Inhibition of IGF-1R in adrenocortical carcinoma. Lancet. Oncology 16 356–357. (doi:10.1016/S1470-2045(15)70130-0)
Klein JD, Turner CG, Gray FL, Yu DC, Kozakewich HP, Perez-Atayde AR, Voss SD, Zurakowski D, Shamberger RC & Weldon CB 2011 Adrenal cortical tumors in children: factors associated with poor outcome. Journal of Pediatric Surgery 46 1201–1207. (doi:10.1016/j.jpedsurg.2011.03.052)
Laplante M & Sabatini DM 2009 mTOR signaling at a glance. Journal of Cell Science 122 3589–3594. (doi:10.1242/jcs.051011)
Leal LF, Mermejo LM, Ramalho LZ, Martinelli CE, Yunes JA, Seidinger AL, Mastellaro MJ, Cardinalli IA, Brandalise SR & Moreira AC et al. 2011 Wnt/beta-catenin pathway deregulation in childhood adrenocortical tumors. Journal of Clinical Endocrinology & Metabolism 96 3106–3114. (doi:10.1210/jc.2011-0363)
Leal LF, Bueno AC, Gomes DC, Abduch R, de Castro M & Antonini SR 2015 Inhibition of the Tcf/beta-catenin complex increases apoptosis and impairs adrenocortical tumor cell proliferation and adrenal steroidogenesis. Oncotarget 6 43016–43032. (doi:10.18632/oncotarget.5513)
Leite FA, Lira RCP, Fedatto PF, Antonini SRR, Martinelli CE, de Castro M, Neder L, Ramalho LNZ, Tucci S & Mastelaro MJ et al. 2014 Low expression of HLA-DRA, HLA-DPA1, and HLA-DPB1 is associated with poor prognosis in pediatric adrenocortical tumors (ACT). Pediatric Blood and Cancer 61 1940–1948. (doi:10.1002/pbc.25118)
Lerario AM, Moraitis A & Hammer GD 2014 Genetics and epigenetics of adrenocortical tumors. Molecular and Cellular Endocrinology 386 67–84. (doi:10.1016/j.mce.2013.10.028)
Liu P, Cheng H, Roberts TM & Zhao JJ 2009 Targeting the phosphoinositide 3-kinase pathway in cancer. Nature Reviews. Drug Discovery 8 627–644. (doi:10.1038/nrd2926)
Livak KJ & Schmittgen TD 2001 Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods 25 402–408. (doi:10.1006/meth.2001.1262)
Lorea CF, Moreno DA, Borges KS, Martinelli CE, Antonini SRR, de Castro M, Tucci S, Neder L, Ramalho LNZ & Cardinalli I et al. 2012 Expression profile of apoptosis-related genes in childhood adrenocortical tumors: low level of expression of BCL2 and TNF genes suggests a poor prognosis. European Journal of Endocrinology 167 199–208. (doi:10.1530/EJE-12-0183)
Maki RG 2010 Small is beautiful: insulin-like growth factors and their role in growth, development, and cancer. Journal of Clinical Oncology 28 4985–4995. (doi:10.1200/JCO.2009.27.5040)
Michalkiewicz E, Sandrini R, Figueiredo B, Miranda ECM, Caran E, Oliveira-Filho AG, Marques R, Pianovski MAD, Lacerda L & Cristofani LM et al. 2004 Clinical and outcome characteristics of children with adrenocortical tumors: a report from the International Pediatric Adrenocortical Tumor Registry. Journal of Clinical Oncology 22 838–845. (doi:10.1200/JCO.2004.08.085)
Moreira AC & Elias LL 1992 Pituitary-adrenal responses to corticotropin-releasing hormone in different degrees of adrenal 21-hydroxylase deficiency. Journal of Clinical Endocrinology & Metabolism 74 198–203. (doi:10.1210/jcem.74.1.1309366)
Mountzios G, Kostopoulos I, Kotoula V, Sfakianaki I, Fountzilas E, Markou K, Karasmanis I, Leva S, Angouridakis N & Vlachtsis K et al. 2013 Insulin-like growth factor 1 receptor (IGF1R) expression and survival in operable squamous-cell laryngeal cancer. PLoS ONE 8 e54048. (doi:10.1371/journal.pone.0054048)
Mulvihill MJ, Cooke A, Rosenfeld-Franklin M, Buck E, Foreman K, Landfair D, O’Connor M, Pirritt C, Sun Y & Yao Y et al. 2009 Discovery of OSI-906: a selective and orally efficacious dual inhibitor of the IGF-1 receptor and insulin receptor. Future Medicinal Chemistry 1 1153–1171. (doi:10.4155/fmc.09.89)
O’Brien J, Wilson I, Orton T & Pognan F 2000 Investigation of the Alamar Blue (resazurin) fluorescent dye for the assessment of mammalian cell cytotoxicity. European Journal of Biochemistry 267 5421–5426. (doi:10.1046/j.1432-1327.2000.01606.x)
Pinto EM, Chen X, Easton J, Finkelstein D, Liu Z, Pounds S, Rodriguez-Galindo C, Lund TC, Mardis ER & Wilson RK et al. 2015 Genomic landscape of paediatric adrenocortical tumours. Nature Communications 6 6302. (doi:10.1038/ncomms7302)
Pitts TM, Tan AC, Kulikowski GN, Tentler JJ, Brown AM, Flanigan SA, Leong S, Coldren CD, Hirsch FR & Varella-Garcia M et al. 2010 Development of an integrated genomic classifier for a novel agent in colorectal cancer: approach to individualized therapy in early development. Clinical Cancer Research 16 3193–3204. (doi:10.1158/1078-0432.CCR-09-3191)
Pollak M 2012 The insulin and insulin-like growth factor receptor family in neoplasia: an update. Nature Reviews. Cancer 12 159–169. (doi:10.1038/nrc3215)
Puzanov I, Lindsay CR, Goff L, Sosman J, Gilbert J, Berlin J, Poondru S, Simantov R, Gedrich R & Stephens A et al. 2015 A phase I study of continuous oral dosing of OSI-906, a dual inhibitor of insulin-like growth factor-1 and insulin receptors, in patients with advanced solid tumors. Clinical Cancer Research 21 701–711. (doi:10.1158/1078-0432.CCR-14-0303)
R Development Core Team 2014 R : A language and environment for statistical computing. R Foundation for Statistical Computing . (available at: https://www.r-project.org/)
Ramanjaneya M, Conner AC, Brown JEP, Chen J, Digby JE, Barber TM, Lehnert H & Randeva HS 2011 Adiponectin (15-36) stimulates steroidogenic acute regulatory (STAR) protein expression and cortisol production in human adrenocortical cells: role of AMPK and MAPK kinase pathways. Biochimica et Biophysica Acta 1813 802–809. (doi:10.1016/j.bbamcr.2011.02.010)
Ribeiro RC & Figueiredo B 2004 Childhood adrenocortical tumours. European Journal of Cancer 40 1117–1126. (doi:10.1016/j.ejca.2004.01.031)
Riedemann J & Macaulay VM 2006 IGF1R signalling and its inhibition. Endocrine-Related Cancer 13 (Supplement 1) S33–S43. (doi:10.1677/erc.1.01280)
Samandari E, Kempná P, Nuoffer J-M, Hofer G, Mullis PE & Flück CE 2007 Human adrenal corticocarcinoma NCI-H295R cells produce more androgens than NCI-H295A cells and differ in 3beta-hydroxysteroid dehydrogenase type 2 and 17,20 lyase activities. Journal of Endocrinology 195 459–472. (doi:10.1677/JOE-07-0166)
Sampaoli C, Cerquetti L, El Gawhary R, Bucci B, Amendola D, Marchese R, Misiti S, Novelli G, Toscano V & Stigliano A 2012 p53 Stabilization induces cell growth inhibition and affects IGF2 pathway in response to radiotherapy in adrenocortical cancer cells. PLoS ONE 7 e45129. (doi:10.1371/journal.pone.0045129)
Sandrini F, Villani DP, Tucci S, Moreira AC, de Castro M & Elias LLK 2005 Inheritance of R337H p53 gene mutation in children with sporadic adrenocortical tumor. Hormone and Metabolic Research 37 231–235. (doi:10.1055/s-2005-861373)
Singh P, Alex JM & Bast F 2014 Insulin receptor (IR) and insulin-like growth factor receptor 1 (IGF-1R) signaling systems: Novel treatment strategies for cancer. Medical Oncology 31 1–14. (doi:10.1007/s12032-013-0805-3)
Tadjine M, Lampron A, Ouadi L & Bourdeau I 2008 Frequent mutations of -catenin gene in sporadic secreting adrenocortical adenomas. Clinical Endocrinology 68 264–270. (doi:10.1111/j.1365-2265.2007.03033.x)
Tamimi RM, Colditz GA, Wang Y, Collins LC, Hu R, Rosner B, Irie HY, Connolly JL & Schnitt SJ 2011 Expression of IGF1R in normal breast tissue and subsequent risk of breast cancer. Breast Cancer Research and Treatment 128 243–250. (doi:10.1007/s10549-010-1313-1)
Wang Y & Sun Y 2002 Insulin-like growth factor receptor-1 as an anti-cancer target: blocking transformation and inducing apoptosis. Current Cancer Drug Targets 2 191–207. (doi:10.2174/1568009023333863)
Wang Q, Wei F, Lv G, Li C, Liu T, Hadjipanayis CG, Zhang G, Hao C & Bellail AC 2013 The association of TP53 mutations with the resistance of colorectal carcinoma to the insulin-like growth factor-1 receptor inhibitor picropodophyllin. BMC Cancer 13 521. (doi:10.1186/1471-2407-13-521)
Wasserman JD, Zambetti GP & Malkin D 2012 Towards an understanding of the role of p53 in adrenocortical carcinogenesis. Molecular and Cellular Endocrinology 351 101–110. (doi:10.1016/j.mce.2011.09.010)
West AN, Neale GA., Pounds S, Figueredo BC, Galindo CR, Pianovski MAD, Oliveira Filho AG, Malkin D, Lalli E & Ribeiro R et al. 2007 Gene expression profiling of childhood adrenocortical tumors. Cancer Research 67 600–608. (doi:10.1158/0008-5472.CAN-06-3767)
Wilkin F 2000 Pediatric adrenocortical tumors: molecular events leading to insulin-like growth factor II gene overexpression. Journal of Clinical Endocrinology & Metabolism 85 2048–2056. (doi:10.1210/jc.85.5.2048)
Zhao H, Desai V, Wang J, Epstein DM, Miglarese M & Buck E 2012 Epithelial-mesenchymal transition predicts sensitivity to the dual IGF-1R/IR inhibitor OSI-906 in hepatocellular carcinoma cell lines. Molecular Cancer Therapeutics 11 503–513. (doi:10.1158/1535-7163.MCT-11-0327)
Zinn RL, Gardner EE, Marchionni L, Murphy SC, Dobromilskaya I, Hann CL & Rudin CM 2013 ERK phosphorylation is predictive of resistance to IGF-1R inhibition in small cell lung cancer. Molecular Cancer Therapeutics 12 1131–1139. (doi:10.1158/1535-7163.MCT-12-0618)