MANAGEMENT OF ENDOCRINE DISEASE: Adrenocortical carcinoma: differentiating the good from the poor prognosis tumors

in European Journal of Endocrinology
Authors:
Anne JouinotInstitut Cochin, INSERM U1016, CNRS UMR8104, Paris Descartes University, Paris, France
Medical Oncology Reference Center for Rare Adrenal Diseases, Department of Endocrinology, Assistance Publique Hôpitaux de Paris, Hôpital Cochin, Paris, France

Search for other papers by Anne Jouinot in
Current site
Google Scholar
PubMed
Close
and
Jérôme BertheratInstitut Cochin, INSERM U1016, CNRS UMR8104, Paris Descartes University, Paris, France
Reference Center for Rare Adrenal Diseases, Department of Endocrinology, Assistance Publique Hôpitaux de Paris, Hôpital Cochin, Paris, France

Search for other papers by Jérôme Bertherat in
Current site
Google Scholar
PubMed
Close
View More View Less

Free access

Adrenocortical carcinoma (ACC) is a rare malignancy with a poor prognosis, the five-years overall survival being below 40%. However, there is great variability of outcomes and we have now a better view of the heterogeneity of tumor aggressiveness. The extent of the disease at the time of diagnosis, best assayed by the European Network for the Study of Adrenal Tumors (ENSAT) Staging Score, is a major determinant of survival. The tumor grade, including the mitotic count and the Ki67 proliferation index, also appears as a strong prognostic factor. The assessment of tumor grade, even by expert pathologists, still suffers from inter-observer reproducibility. The emergence of genomics in the last decade has revolutionized the knowledge of molecular biology and genetics of cancers. In ACC, genomic approaches – including pan-genomic studies of gene expression (transcriptome), recurrent mutations (exome or whole-genome sequencing), chromosome alterations, DNA methylation (methylome), miRNA expression (miRnome) – converge in a new classification of ACC, characterized by distinct molecular profiles and very different outcomes. Targeted measurements of a few discriminant molecular alterations have been developed in the perspective of clinical routine, and thus, may help defining therapeutic strategy. By individualizing patients’ prognosis and tumor biology, these recent progresses appear as an important step forward towards precision medicine.

Abstract

Adrenocortical carcinoma (ACC) is a rare malignancy with a poor prognosis, the five-years overall survival being below 40%. However, there is great variability of outcomes and we have now a better view of the heterogeneity of tumor aggressiveness. The extent of the disease at the time of diagnosis, best assayed by the European Network for the Study of Adrenal Tumors (ENSAT) Staging Score, is a major determinant of survival. The tumor grade, including the mitotic count and the Ki67 proliferation index, also appears as a strong prognostic factor. The assessment of tumor grade, even by expert pathologists, still suffers from inter-observer reproducibility. The emergence of genomics in the last decade has revolutionized the knowledge of molecular biology and genetics of cancers. In ACC, genomic approaches – including pan-genomic studies of gene expression (transcriptome), recurrent mutations (exome or whole-genome sequencing), chromosome alterations, DNA methylation (methylome), miRNA expression (miRnome) – converge in a new classification of ACC, characterized by distinct molecular profiles and very different outcomes. Targeted measurements of a few discriminant molecular alterations have been developed in the perspective of clinical routine, and thus, may help defining therapeutic strategy. By individualizing patients’ prognosis and tumor biology, these recent progresses appear as an important step forward towards precision medicine.

Invited Author’s profile

Jérôme Bertherat, MD-PhD, is professor of Endocrinology at Paris Descartes University, Chief of the Endocrinology Department of Cochin Hospital in Paris, head of the National Center for Rare Adrenal Diseases, one of the Health Care Provider of the European Reference Network for Rare Endocrine Conditions. His research team ‘Genomics and Signaling of Endocrine Tumors’ in the Cochin Institute (INSERM U1016 & CNRS UMR8104), works on the pathophysiology of Cushing’s syndrome, the molecular genetics of adrenocortical tumors and the genetics of familial adrenal tumors and Carney complex. His research on adrenal tumors is developed in the background of national (COMETE) and international (European Network for the Study of Adrenal Tumors: ENSAT) networks.

Introduction

Adrenocortical carcinoma (ACC) is a rare endocrine malignancy, developed from the adrenal cortex, with estimated incidence of one per million each year (1). The prognosis is generally poor with five-year overall survival (OS) below 40% in most series (1, 2, 3, 4, 5, 6, 7, 8, 9). However, the prognosis of ACC is also very heterogeneous and difficult to predict in clinical practice. Great variability in clinical presentations is observed, ranging from tumors with an indolent clinical course to aggressive tumors with fatal outcome.

Complete surgical resection represents the only curative treatment for ACC, which can be achieved only in tumors with no extensive locoregional or distant tumor dissemination. For patients with localized tumors, up to 70% will develop recurrence in the 3 years following radical surgery and the vast majority of patients with tumor recurrence will die from tumor progression (5, 6, 9). However, more recent series suggest that patients with a localized tumor radically resected in expert centers might recur in no more than 30% (10).

In patients with metastatic disease, medical therapies, such as mitotane or platinum-based chemotherapy regimens, offer limited efficacy (11, 12). Local treatment of metastases by surgical or non-surgical approaches – such as trans-arterial chemoembolization, radiofrequency ablation or radiotherapy – are offered in selected cases, but their benefit in terms on OS has been poorly evaluated (13, 14). Median OS of metastatic ACC patients varies between 10 and 20 months (2, 3, 5), with 5-year survival around 10% (3, 6, 8). However, some long-term survivors are described, including a few cases with synchronous metastases at the time of diagnosis (15, 16, 17, 18, 19, 20). These observations suggest that some ACC have a slow natural history, in contrast with the usual poor prognosis of this malignancy. Such different clinical presentations could be explained by distinct biology. In fact, genomic studies have demonstrated the existence of subgroups of ACC with heterogeneous molecular background, independent of clinical and pathological features (21, 22).

Prognostic stratification is important for clinical management of ACC patients (23). In patients with localized disease that underwent radical surgery, the estimation of the recurrence risk determines the prescription of adjuvant therapies and individualizes patient follow-up. In patients with non-removable tumor, prognosis assessment is also a major determinant of the therapeutic strategy. Mitotane and local treatments on metastases can be proposed to patients with indolent tumors, whereas those presenting aggressive disease are better candidates for mitotane and chemotherapy combination.

This review describes and discusses the main clinical and pathological established prognostic factors in ACC, as well as the new molecular markers derived from genomics that provide new perspective to understand ACC heterogeneity and to develop new ACC prognostication strategies.

Methods

A systematic search on ACC prognosis was performed using the following search terms on PubMed until December 2017: ‘adrenocortical carcinoma’ (Mesh) and ‘prognosis’ (Mesh). A total of 380 records were screened. Original articles and reviews were considered, and their reference lists were screened for additional relevant publications.

Clinical prognostic factors

Tumor stage

Tumor stage represents the main prognostic factor of ACC. Different staging classifications have been proposed through years and tumor stage has been recognized as a consistent and independent predictor of survival in ACC (3, 4, 5, 9, 11, 24, 25, 26).

As for other cancer types, ACC staging is based on the tumor, node and metastasis (TNM) classification, which assesses the local extension of the primary tumor, the lymph node involvement and the presence of distant metastasis. The aim of this classification is to provide a robust prognostic tool for both disease-free (DFS) and disease-specific survival (DSS) in cancer patients. The first TNM classification in ACC was proposed in 2004 by the International Union Against Cancer (UICC) and the American Joint Committee on Cancer. This scheme was largely based on previous classification system proposed by Macfarlane (27) and modified by Sullivan (28). Tumors limited to the adrenal gland were classified as stage I (T1N0M0) if <5 centimeters (cm) or stage II (T2N0M0) if >5 cm. Advanced tumors were categorized as stage III in case of tumor infiltration into surrounding tissue (T3N0M0) or lymph node invasion (T1–2N1M0) and as stage IV in case of tumor infiltration into surrounding tissue and lymph node invasion (T3N1M0), tumor invasion into adjacent organs (T4N0–1M0) or metastatic disease (T1–4N0–1M1).

The European Network for the Study of Adrenal Tumors (ENSAT) evaluated the prognostic value of 2004 UICC classification, and showed that distant metastases were associated with decreased survival, compared to other advanced stages without metastases (2) (DSS hazard ratio, HR = 2.93). A modification of the 2004 UICC-staging system was proposed in consequence (Fig. 1): the 2008 ENSAT staging system reclassified stage III as all locally advanced tumors (T3–4N0M0 or T1–4N1M0). Conversely, the ENSAT stage IV included only tumors with distant metastases (T1–4N0–1M1) (2). The prognostic value of ENSAT-staging classification was validated by another team in North America population (29), which confirmed the better survival stratification of the ENSAT classification compared to the 2004 UICC classification. ENSAT-staging system is now considered as a standard prognostic factor for ACC management (23). A modified ENSAT (mENSAT) classification was then proposed in order to precise the prognosis of advanced ACC, with new definitions of stage III (invasion of surrounding tissues/organs or the vena renalis/cava) and stage IVa, IVb, IVc (2, 3 or >3 metastatic organs, including lymph node invasion, respectively) (30).

Figure 1
Figure 1

Disease-specific survival stratified on the 2008 ENSAT staging classification. From Fassnacht et al. (2). HR, hazard ratio with 95% confidence intervals; P, log-rank P-value.

Citation: European Journal of Endocrinology 178, 5; 10.1530/EJE-18-0027

One limit of the current staging system is the distinction between stage I and stage II. The cutoff of 5 cm in tumor size is rather arbitrary, and no significant difference in DSS was shown between stage I and stage II (2, 29). Of note, the number of patients with stage I tumors is small – only 5% of all ACC (2) – which limits substantially the survival analysis. However, tumor size alone is not consistently found as a prognostic factor in ACC (5, 26, 31) and the use of different cutoffs in tumor size does not improve prognosis stratification of patients with stage II compared with those with stage I disease (29).

Cortisol secretion

ACC cause steroid hormones excess in 50–75% of the cases (6, 26, 32), including androgens, estrogens, mineralocorticoids and corticosteroids. Cortisol-secreting tumors are the most common secreting tumors. However, the impact of cortisol secretion on survival in ACC remains uncertain. In patients that underwent surgery, cortisol secretion appeared as an adverse prognostic factor of recurrence and death in ACC (OS HR ≈ 1.5) (6, 26, 32, 33), although this effect was not always significant after adjustment on patient and disease characteristics (33). In addition, cortisol secretion failed to demonstrate prognostic value in other series involving localized (34) or metastatic ACC patients (3).

Although patients with non-functional tumors might be often diagnosed latter than those with steroid excess, several hypotheses can be raised about the poor prognostic value of cortisol-secreting tumors. First, patients with cortisol-secreting tumors commonly present with severe Cushing’s syndrome. Those patients are more likely to experience a postoperative complication compared to patients with non-functional tumors (33). Moreover, cortisol secretion may favor escape from immune surveillance by blunting the cellular immune response (35), which could result in tumor growth and recurrence. Independently of the hormonal consequences of cortisol excess, it is also possible that the biology of cortisol-secreting tumor is associated with specific growth characteristics.

Age

ACC can occur at any age of life. In some series, childhood ACC displays a better prognosis than adult’s ACC (36, 37). Younger patients presented lower tumor stages, whereas adults were more likely to show aggressive tumors with shorter prognosis. After complete tumor resection, 5-year DFS was above 80% for children under 3 years but only 40% for children over 13 years, similar to that of adults (37). However, pathology and molecular biology suggest that pediatric ACC are different tumors than adults ACC (38).

In adults, several studies have shown that older age is associated with decreased OS (5, 6, 8, 9, 26, 30, 32, 39). Asare et al. have even proposed a novel staging system, where stage I and stage II ACC were differentiated by age (≤55 years for stage I vs >55 years for stage II) and not by size, like it is done in the ENSAT-staging system (40). This new classification based on age showed significant differences in OS between stage I and stage II (40). However, the majority of studies including multivariate models did not found age as a predictor of recurrence (6, 32, 34) or cancer-specific mortality (29). These data suggest that older age could be associated with an increase in all-cause mortality due to an increase in treatment-related mortality or in non-specific mortality. Thus, in adult’s ACC, older age appears more as a confounding factor of frailty and comorbidities, than as an independent pejorative prognostic factor.

Pathological prognostic factors

Tumor grade: from mitoses to Ki67

Besides tumor stage, tumor grade represents another major prognostic factor for ACC management (23).

Mitotic count is a fundamental criterion in the diagnosis of ACC as part of the Weiss score, which gathers 9 histologic criteria associated with malignancy in adrenal tumors (41). These parameters include high nuclear grade, mitotic count >5 per 50 high-power fields (HPF), atypical mitoses, cytoplasm ≤25% clear cells, diffuse architecture, necrosis, venous invasion, invasion of sinusoidal invasion and capsular invasion. Adrenal tumors with a Weiss score >3 are considered malignant (ACC) and those with a score ≤2 as benign (adenomas). Moreover, Weiss score was developed by comparing metastatic and non-metastatic adrenal tumors and was also associated with prognostic value (41, 42). Among all features of the Weiss score, mitotic count was the only criterion consistently associated with patient prognosis (3, 42, 43, 44, 45, 46). A cut-off of 20 mitotic figures per 50 HPF was proposed to separate low from high-grade ACC (42, 44). Some limitations exist for using mitotic count as a prognostic factor in clinical routine: it is notably time consuming and subject to inter-observer variability, although inter-observer variability may decrease with trained pathologists (47).

The Ki67 immunohistochemistry represents an alternative technique for assessing proliferation in ACC. Several studies have reported the prognostic value of Ki67 index in ACC (30, 34, 48, 49, 50, 51, 52). A large study in localized ACC identified Ki67 as the most powerful prognostic factor of DFS after complete surgery. This study suggested a tumor grading based on three levels of Ki67 index: grade 1 tumors with Ki67 <10%, grade 2 with Ki67 10–19% (DFS HR = 1.94) and grade 3 tumors with Ki67 ≥20% (DFR HR = 2.58). Likewise, the Ki67 index was also an independent prognostic factor of OS (34). In recent series, evaluating both mitotic count and Ki67, the Ki67 proliferation index proved to be the most powerful tool to predict patient’s survival (34, 51) (Fig. 2). However, Ki67 index was not consistently associated with OS in stages III and IV ACC (30).

Figure 2
Figure 2

Recurrence-free (A) and overall (B) survival stratified on the Ki67 index in stages I–III ACC after R0 surgery. From Beuschlein et al. (34).

Citation: European Journal of Endocrinology 178, 5; 10.1530/EJE-18-0027

Compared to mitotic count, Ki67 proliferation index is less time consuming but also confronted to limited reproducibility. First, preanalytical protocol – including time to fixation, type of fixative, time in fixative and method for long-term storage – is not standardized and might affect Ki67 measurement (53). Ki67 results also depend on the assay that was used for immunostaining. Mouse anti-human Ki67 monoclonal antibody, MIB1 clone, is the most commonly used assay and is now considered as a gold standard for Ki67 immunostaining. However, even with the same preanalytical protocol and the same antibody, interpretation of Ki67 staining and scoring may be subjected to controversies: how many cells or high-power fields have to be included for evaluation? Do we need to score Ki67 into hot spot areas, or average score across the section, including or excluding hot spot area? A recent study has evaluated the inter-observer variability of Ki67 scoring assessment in ACC among 14 trained endocrine pathologists. Low levels of inter-observer and intra-observer concordance were observed (54), especially for intermediate Ki67 indexes (10–30%). This may be a concern for clinical practice since the clinically relevant cutoff values are in this range. However, the development of digital microscopy-enabled methods could increase reproducibility and improve reliability for clinical setting (54).

In 2014, Pennanen and coworkers have introduced a novel prognostic scoring system – the Helsinki score – that uses 2 Weiss criteria – mitotic index (3 points if >5/50 HPF) and necrosis (5 points) – plus Ki67 index in the most proliferative area of the tumor measured by computer-assisted image analysis. This score was developed by combining the most discriminative parameters for metastatic disease in a large cohort of adrenocortical tumors. For malignant tumors, a score >17 was associated with poor OS (45). The prognostic value of Helsinki score for ACC was validated in an independent cohort (46).

Resection status

The effect of margin status on outcome has been well established for several cancers but only few retrospective studies have evaluated the effect of the margin status in ACC prognosis (5, 8, 55).

Among patients that underwent surgical resection, 20–30% presented positive margins – i.e. R1 (microscopically positive resection margins) or R2 (macroscopic residual disease) resection. R1 and R2 resection are associated with higher risk of recurrence (8) and death (5, 55) (adjusted OS HR ≈ 2). These results highlight the critical role of surgical expertise to achieve R0 resection and to improve outcome.

P53 and β-catenin (CTNNB1) immunohistochemistry

The tumor stage and the resection status have long been the only widely accepted prognostic factor of ACC. However, these features do not capture the wide heterogeneity in outcome for a given stage, leading to the development of several prognostic markers based on immunohistochemistry through years (56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70). Most of these markers are derived from small retrospective series, mostly without adjustment to other prognostic factors and without external validation. Among those, CTNNB1 and P53 staining retain attention as they may be used as surrogates for molecular alterations in the two major signaling pathways of ACC (60). Indeed, activation of the Wnt/CTNNB1 pathway is associated with CTNNB1 nuclear staining and correlates with high mitotic rate (58, 59) and poor survival (56, 57, 58, 59, 60). Somatic mutations or loss of heterozygosity of TP53 result in aberrant P53 expression and are linked with aggressive phenotype (57, 61, 62, 71), with higher tumor stage and poorer DFS. However, these markers did not show independent prognostic value in multivariate analyses including tumor grade (59).

Genomic studies and molecular prognostic markers

The development of the genomics methods during the last decade has allowed to study gene expression, genetic and epigenetic alterations at the pan-genomic level in numerous cancer types. In ACC, genomic studies led to identifying subgroups of tumors with distinct biology and different outcome. These recent progresses also allow the development of prognostic molecular markers derived from genomics (Table 1).

Table 1

Main studies on prognostic molecular markers derived from genomics in ACC.

Reference Molecular marker Patients for prognostic study, n Targeted measurement Prognostic value – univariate analysis Prognostic value – multivariable analysis
mRNA markers
  (72) Clusters based on expression of 14 genes including ISGF3G and IL2RG 13 None Recurrence-free survival NA
 (65) SNAI1 26 RT-qPCR IHC Overall survival NA
 (75) Clusters based on expression of 60 genes 11 None Overall survival NA
 (73) BUB1B-PINK1 differential expression 35 RT-qPCR Disease-specific survival NA
  (63) SF-1 33 (genomics)167 (IHC) IHC Recurrence-free survivalOverall survival Independent of tumor stage, sex, age, and hormone secretion
  (76) BUB1B-PINK1 differential expression 14 RT-qPCR Overall survival NA
 . (77) PTTG1 35 RT-qPCR Overall survival NA
  (70) TOP2A and EZH2 61 IHC Overall survivalRecurrence-free survival TOP2A independent of age and histological invasion
 (78) EZH2 149 RT-qPCR Overall survival NA
 (79) VAV2 126 (genomics)171 (IHC) IHC Recurrence-free survivalOverall survival NA
 (80) VAV2 171 (IHC) IHC Recurrence-free survivalOverall survival Independent of age, sex, tumor stage and Ki67
Non-coding RNA markers
  (94) miR-195 and miR-483-5p 18 RT-qPCR Overall survival NA
 (97) miR-503, miR-1202 and miR-1275 25 RT-qPCR Overall survival NA
 (99) Serum miR-483-5p and miR-195 23 RT-qPCR Recurrence-free survivalOverall survival NA
  (69) miR-9 28 RT-qPCR Recurrence-free survivalOverall survival NA
 (100) miR-210 51 RT-qPCR Overall survival Not independent of mitotic count
 (107) PRINS lncRNA 20 RT-qPCR Recurrence-free survival NA
Chromosome alterations markers
  (83) Clusters based on aberrations in 10 chromosomes 25 None Overall survival NA
  (84) Clusters based on whole genome aberrations 46 None Overall survival Not independent of tumor stage
  (86) Clusters based on aberrations in 12 chromosomes 22 None Overall survival Not independent of tumor stage and Ki67
Methylation markers
  (88) Clusters based on CpG islands methylation 51 MS-MLPA Overall survival NA
 (91) PAX5, PAX6, PYCARD, GSTP1 203 MS-MLPA Recurrence-free survivalOverall survival Independent of tumor stage and Ki67

IHC, immunohistochemistry; MS-MLPA, methylation-specific multiplex ligation-dependent probe amplification; NA, not available; RT-qPCR, reverse transcription-quantitative polymerase chain reaction.

Transcriptome and mRNA prognostic markers

Unsupervised cluster analyses of transcriptome data have identified two major subgroups of ACC based on gene expression profiles. Four studies reported a strong prognostic value of the transcriptome considering DFS (72, 73) and OS (73, 74, 75). Our team has named the ACC transcriptome clusters ‘C1A’ and ‘C1B’, the latter being associated with better outcome (73). Giordano et al. also observed that genes differentially expressed in the two clusters of ACC are mostly involved in cell-cycle progression and proliferation and that transcriptome signatures are in agreement with tumor grade (74). However, the prognostic value of the transcriptome-based classification remained significant independently of tumor stage and tumor grade (73, 74), suggesting that transcriptome did not only reflect tumor grade.

Consequently, molecular prognostic markers were set by translating global gene expression profiles to focus assessments for clinical practice. Our team has developed a simple prognostic marker based on the differential expression of two genes (BUB1B-PINK1) measured by quantitative reverse transcription PCR (RT-qPCR) on tumor RNA. These two gene predictors of survival have been validated in an independent cohort in our institution (73) and by another team (76), and showed an excellent prognostic value – with 5 years OS <20% in the ‘poor prognosis’ subgroup vs >80% in the ‘good prognosis’ subgroup.

Other prognostic markers were proposed based on transcriptome studies (63, 65, 70, 77, 78, 79, 80). For instance, SF-1 expression – which is usually used as a diagnostic tool for the adrenocortical origin of a tumor – was investigated as a prognostic marker in ACC and demonstrated prognostic value on DFS and OS, independently of tumor stage, sex, age and hormone secretion (63). A recent analysis showed that VAV2 expression, which is a SF-1 dosage-dependent target, was also an independent prognostic factor in multivariate analysis including age, sex, tumor stage and Ki67 (79, 80). Finally, a recent meta-analysis of transcriptome data identified the histone methyltransferase EZH2 as overexpressed in ACC. High EZH2 expression was also associated with proliferation and poor prognosis (78). Of note, another team demonstrated that EZH2 staining also carries prognostic value in stages I–II ACC (70). The major limit of these studies is that EZH2 expression was not confronted to other prognostic factors of ACC, such as tumor stage.

Exome and genes with recurrent mutations

Before genomics studies, several publications focused on specific candidate genes in ACC, including TP53 and CTNNB1. Recently, three pangenomic studies have investigated instead exome sequencing and SNP arrays to identify somatic mutations, homozygous deletions and high-level amplifications in adult’s ACC (21, 22, 81). These studies converged on a short list of genes with recurrent alterations, occurring in about 50% of ACC. The most frequent alterations were CTNNB1 and TP53 mutations and ZNRF3 and CDKN2A homozygous deletions.

About 20% of ACC harbored gain-of-function mutations in CTNNB1 (21, 22, 81). Another 20% presented ZNRF3 deletions (21, 22) – a negative regulator of Wnt/CTNNB1 – which results in the activation of the Wnt/CTNNB1 pathway. Thus, CTNNB1 mutations correlated with nuclear CTNNB1 staining (57, 58, 60). Moreover, CTNNB1 mutations were associated with higher mitotic count (58), poor prognosis transcriptome cluster (21, 57) and poor survival (57, 58, 60). However, the prognostic value of CTNNB1 was not tested after adjustment on tumor grade. A trend toward OS was also noted for ZNFR3 deletions (81).

Loss-of-function mutations of TP53 occurred in about 20% of adult’s ACC (21, 22), often associated with loss of heterozygosity (LOH) of the 17p13 region, on which TP53 is located (61, 71). TP53 mutations were almost mutually exclusive from CTTNB1 mutations (21, 22, 81). TP53-mutated tumors were associated with aberrant P53 staining (57, 61, 62), advanced stage (61), poor prognosis transcriptome cluster (21, 57) and shorter DFS (61). No significant association with OS was reported (61, 62, 81). Germline TP53 mutations have also been linked with the development of ACC, as part of the Li-Fraumeni syndrome. In pediatric ACC, germline TP53 mutations were indeed very frequent, representing up to 70% of tumors and were also associated with advanced stage and shorter DFS (38).

Chromosomal alterations

Comparative genomic hybridization (CGH) and single-nucleotide polymorphism (SNP) arrays showed recurrent chromosomal alterations in ACC genome, including gains (chromosome 5, 7, 12, and 19) and losses (chromosome 1, 2, 13, 17, 22) (82, 83, 84, 85, 86), extended LOH (21, 22, 86) and whole-genome doubling (22).

Gains and losses profiles contained prognostic information on DFS (84) and OS (83, 84). Recently, Zheng et al. have determined three subgroups of ACC from unsupervised clustering based on arm-level alterations (22). ‘Chromosomal’ profiles showed the highest frequency of whole chromosome arm gains and losses, with extended LOH. ‘Noisy’ profiles were characterized by high number of chromosomal breaks. ‘Quiet’ profiles displayed a very limited number of large alterations. In terms of prognosis, noisy tumors showed decreased DFS, compared with chromosomal and quiet tumors. Moreover, in the subgroup of chromosomal tumors, whole genome doubling event was associated with worse prognosis.

The prognostic value of clusters based on chromosomal alterations has been validated in independent cohorts (22, 84, 86). However, some limitations need to be taken in account before using chromosomal alterations as a prognostic marker in clinical practice. On the one hand, single alterations cannot account for significant prognostic information, making DNA chips necessary to obtain a large combination of alterations for ACC prognostication (84). Our team has recently developed a new process, using NGS to determine chromosomal alterations profiles by sequencing a limited number of SNP on selected chromosome arms (87). On the other hand, the prognostic value of chromosomal alterations needs to be evaluated after adjustment on other prognostic factors in ACC, including tumor stage and tumor grade, and requires further prospective validations in independent centers.

Methylome and methylation prognostic marker

Three studies have investigated genome-wide DNA methylation profiles in ACC and showed that a subset of ACC presents hypermethylation of CpG islands that are located in gene promoter regions (88, 89, 90). The CpG islands methylator phenotype (CIMP) resulted in transcriptional inactivation of tumor suppressor genes (88, 89, 90) and was associated with poor prognosis in ACC (88).

Our team has recently developed a targeted prognostic marker measuring methylation of 4 genes (PAX5, PAX6, PYCARD, GSTP1) (91). These genes were selected for their strong correlation to methylome profiles (CIMP or non-CIMP) and for association with survival. The methylation of selected genes was measured with a commercial kit by methylation-specific multiplex ligation-dependent probe amplification (MS-MLPA), a technique based on methylation-sensitive restriction enzyme and PCR. This methylation marker demonstrated strong prognostic value on DFS (HR = 2.8) and OS (HR = 2.4), independently of ENSAT tumor stage and Ki67 proliferation index (91).

In the perspective of moving to clinical practice, targeted measurements by MS-MLPA represent a fast and cost-effective approach compared to methylation arrays. In contrast to the previous RNA marker derived from the transcriptome study, the methylation marker is based on DNA analysis, which is easier to obtain in routine practice. In the future, the prognostic value of methylation has also to be confronted with other molecular markers, including those derived from transcriptome, chromosomal alterations and miRNome studies.

MiRNome and miRNA prognostic markers

MicroRNA (miRNA) are small non-coding RNA (18–25 nt), regulating the gene expression at the post-transcriptional level by RNA-interference (92). Several hundreds of miRNA have been identified, and some of them may be good biomarkers for cancer diagnosis and prognosis (93). MiRnome studies identified several miRNA differentially expressed between ACC and normal adrenal or benign tumors: among the most consensual ones, miR-483-3p, miR-483-5p, miR-210 and miR-503 were upregulated, while miR-195 and miR-335 were downregulated in ACC (21, 94, 95, 96, 97, 98, 99, 100, 101). Unsupervised classifications of ACC based on miR expression showed 2 or 3 subgroups of ACC, which are associated with different outcomes (21, 94, 97). MiRNA clusters were correlated with transcriptome subgroups, with rather miRNA overexpression in the transcriptome ‘C1A’ cluster.

MiRNA are more stable than mRNA, and therefore, offer an important advantage for clinical routine use. Indeed, several studies have proposed RT-qPCR prognostic markers based on miRNA expression, including miR-195 and miR-483-5p (94), miR-503 (97), miR-9 (69) and miR-210 (100). However, the prognostic value of selected miRNA was either not tested after adjustment on other prognostic factors or dropped in multivariate analysis for miR-210 (100). This could be explained by the fact that miR-210 expression was significantly associated with Ki67 index and brought no additional prognostic information to tumor grade.

MiRnome dysregulation in ACC has led to investigate the expression of the miRNA processing machinery, such as DICER1 enzyme and its cofactor TARBP2, as potential diagnostic and prognostic markers in adrenocortical tumors (102, 103). Thus, DICER1 immunochemistry was found as an independent predictor of DFS (HR = 2.6) but not of OS (103).

Chabre et al. also investigated the prognostic value of circulating miRNA (99). Low miR-195 and high miR-483-5p levels in the serum of ACC patients were associated with shorter DFS and OS. However, this result was obtained on 12 ACC patients and was not confirmed by another small series (104).

Long non-coding RNA

Long non-coding RNA (lncRNA) are RNA transcripts longer than 200 nt that may act in epigenetic silencing, transcriptional and splicing regulation (105). LncRNA dysregulation has been involved in the development of several neoplasms (106). SNP array analysis identified recurrent deletions in the locus of LINbib290, suggesting a role of lncRNA in ACC pathogenesis (21). One study focused on lncRNA expression profiling in ACC (107). LncRNA microarrays showed distinct patterns of expression between ACC and normal adrenal or benign tumors. Moreover, expression of the lncRNA PRINS was associated with DFS and its prognostic value was validated by RT-qPCR in an independent cohort of 20 ACC (107).

Molecular classification

Genomics studies showed major differences between aggressive and non-aggressive ACC. Further step was to generate global genomics classification by integration of transcriptome profiles with exome or whole-genome sequencing, SNP array analysis, methylome and miRnome. Integrated genomics has drawn a new classification of ACC, with distinct molecular subgroups that are associated with very different outcome. Omics-based classifications have been generated in two independent international cohorts: one from the ENSAT network (21) in Europe and one from The Cancer Genome Atlas (TCGA) (22) consortium in America, Europe and Australia. These two studies converged in a highly consensual global molecular classification. The strong agreement between the different omics allowed to define two main molecular subgroups, corresponding to ACC of ‘good’ and ‘bad’ prognosis.

Our team as part of the ENSAT network has proposed a classification based on the two transcriptome signatures ‘C1A’ and ‘C1B’ (Fig. 3A and B). The molecular group of ‘good’ prognosis ACC was characterized by ‘C1B’ cluster, low mutation rate, very rare mutations in ACC driver genes and no hypermethylation (non-CIMP). Conversely, the molecular group of aggressive ACC corresponded to ‘C1A’ transcriptome signature and harbored high mutation rate and frequent alterations on the recurrent ACC driver genes. A subset of aggressive tumors was also hypermethylated and associated with poorer outcome compared with non-hypermethylated tumors. Therefore, methylation pattern defined one subgroup of poorer prognosis (C1A and CIMP) and another subgroup of intermediate prognosis (C1A and non-CIMP) into this group of aggressive ACC (21).

Figure 3
Figure 3

Integrated genomics of ACC. From Assié et al. (A and B) and Zheng et al. (C and D). (A) Integrated genomics of the ENSAT cohort. Tumors were divided into 2 groups from mRNA expression. DNA methylation, miRNA expression clusters, mutation rate and alterations in key genes and pathways converge with transcriptome clusters. (B) Overall survival stratified on the ENSAT genomic classification. (C) Integrated genomics of the TCGA cohort. Tumors were divided into 3 groups by clustering the clusters from mRNA expression (red), chromosome alterations profiles (black), DNA methylation profiles (blue); miRNA expression (purple). (D) Event-free survival stratified on the TCGA genomic classification.

Citation: European Journal of Endocrinology 178, 5; 10.1530/EJE-18-0027

In the TCGA study, the unsupervised clustering of omics clusters – including transcriptome, miRnome, methylome and chromosomal alterations profiles – established a molecular classification into three groups (Fig. 3C and D). The first group corresponded to ‘good’ prognosis ACC and was characterized by transcriptome ‘C1B’, ‘chromosomal’ SNP profile and no hypermethylation. The two other groups corresponded to aggressive ACC and showed a ‘C1A’ transcriptome cluster profile. The second group showed intermediate levels of hypermethylation (CIMP-intermediate), and ‘chromosomal’ SNP profile, and was associated with intermediate outcome, whereas the third group of tumors was characterized by high levels of hypermethylation (CIMP-high), ‘noisy’ SNP profile and poorer prognosis (22).

A global molecular classification of ACC emerges from the studies described in this review (Fig. 4). As stated before for individual omics studies, the molecular classification could theoretically be summarized to a combination of targeted measurements for ACC prognostication in clinical setting. However, the combination of targeted molecular markers should both be as simple as possible for routine use and should demonstrate prognostic value independently of tumor stage and tumor grade.

Figure 4
Figure 4

Global molecular classification of ACC. Gene expression, chromosome alterations and DNA methylation profiles divided ACC into 3 prognostic groups. These groups are also associated with some other molecular events – including miRNA expression and somatic mutations – and clinical characteristics. Gene expression profiling defines poor/intermediate prognostic ‘C1A’ and better prognostic ‘C1B’ clusters. The ‘C1A’ group of poor prognosis is also characterized by ‘noisy’ chromosome profile (numerous chromosome losses and gains) and hypermethylation in CpG islands (CIMP). The ‘C1A’ group of intermediate prognosis displays ‘noisy’ profile or ‘CIMP’ but not both. The ‘C1B’ group shows a small number of molecular events – the most common alteration being a downregulation of the DLK1-MEG3 miR cluster (14q32.2) – and a better prognosis.

Citation: European Journal of Endocrinology 178, 5; 10.1530/EJE-18-0027

Summary and perspectives

Due to rarity of the disease, prognostic factors in ACC are established on small series, mostly in retrospective design. The tumor stage still represents the main prognostic factor in ACC. Among all classifications that were proposed, the ENSAT-staging system provides the best survival discrimination. This classification highlights the poor outcome of metastatic disease (stage IV), that are now individualized from locally advanced tumors (stage III) (2).

Beyond tumor stage, the tumor grade also shows a strong prognostic value. The mitotic count tends to be replaced by the Ki67 proliferation index (34). Tumor grade assessment still suffers from inter-observer reproducibility, although new computer-assisted techniques seem promising (54).

Molecular classification provides new insights in ACC biology. Distinct molecular subgroups have emerged from genomics, associated with different outcomes (21, 22). This clearly explains why prognosis of this aggressive cancer might nevertheless vary among tumors. On the short term, this could help to develop new molecular prognostic markers for patient management. On the long term, this will inevitably stimulate the development of specific targeted therapies based on the tumor biology driving these molecular classes.

To develop molecular markers, gene and miRNA expression can be assayed by targeted measurements using RT-qPCR. Focal measurements of methylation can be done with qPCR based techniques – such as MS-MLPA – or with targeted NGS after bisulfite treatment (87). Alterations of driver genes and chromosomal profiles can both be determined by targeted NGS (87). However, additional studies are needed to precise the best set of molecular markers for prognosis and to confront their prognostic value to the already validated prognostic markers, especially ENSAT stage and Ki67 index. Moreover, the reproducibility of the discriminant molecular tools within a single tumor has also to be evaluated. Finally, the molecular markers will have to be simple and cost-effective for transfer to clinical routine.

Individualizing patients’ prognosis and tumor biology appears as a necessary step for personalized medicine. In addition to tumor stage and tumor grade, the genomic classification may precise the risk stratification and thus help defining therapeutic strategy.

In adjuvant setting, it would be of particular interest to determine the individual risk of recurrence. Patients with molecular ‘good’ prognosis ACC could probably be proposed a simple follow-up after radical ‘R0’ resection and benefit from loco-regional treatments in case of recurrence. By contrast, patients with molecular aggressive ACC could benefit from closer follow-up and adjuvant systemic therapies like mitotane or even platinum chemotherapy.

Liquid biopsies represent another perspective for patients’ follow-up. Circulating miRNA (99, 104, 108, 109) and circulating tumor DNA (110, 111) are measurable in patients’ blood. Larger validation cohorts are needed to precise their ability for early detection of recurrence.

For advanced disease, one future direction will be the pharmacogenomics of ACC. Although overall response rate remains below 25% (12), a few patients show deep and durable responses to mitotane or to platinum-based chemotherapy. Additional studies are needed to precise whether some molecular markers can predict response to treatment and identify this minority of responders.

Genomics may also help identifying new therapeutic targets. Wnt/β-catenin pathway inhibitors (112) and cycle cell inhibitors, such as CDK4/CDK6 or Wee1 inhibitors (113), are currently in development and could offer new therapeutic strategies for ACC with alterations in these pathways. Demethylating agents could be proposed for hypermethylated tumors. Indeed, Suh et al. showed that decitabine inhibits proliferation in ACC cells (NCI-H295R) (114). Demethylating agents have mostly shown efficacy in hematological diseases, whereas they induced at best a stable disease in patients treated for solid tumors (115). However, their efficacy in CIMP ACC has not been evaluated so far. RNA interference therapy represents another promising approach to target molecular alterations. A recent preclinical study reported the first hopeful results of miRNA replacement in ACC, showing that systemic miR-7 administration reduces ACC xenograft growth (116).

Such strategies based on the molecular classification would need prospective validation in clinical trials.

Declaration of interest

The authors declare that there is no conflict of interest that could be perceived as prejudicing the impartiality of this review.

Funding

This work was supported by ITMO (Institut Thématique Multi-organismes) Cancer AVIESAN (Alliance pour les Sciences de la Vie et de la Santé) in part of Plan Cancer 2014–2019 (A J is receiving a PhD grant) and the COMETE TACTIC PHRC grant.

Acknowledgements

The authors thank the members of the Genomics and Signaling of Endocrine Tumors team of Cochin Institute for their thoughtful comments and helpful discussions.

References

  • 1

    Kerkhofs TMA, Verhoeven RHA, Van der Zwan JM, Dieleman J, Kerstens MN, Links TP, Van de Poll-Franse LV & Haak HR. Adrenocortical carcinoma: a population-based study on incidence and survival in the Netherlands since 1993. European Journal of Cancer 2013 49 25792586. (https://doi.org/10.1016/j.ejca.2013.02.034)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 2

    Fassnacht M, Johanssen S, Quinkler M, Bucsky P, Willenberg HS, Beuschlein F, Terzolo M, Mueller HH, Hahner S & Allolio B 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 243250. (https://doi.org/10.1002/cncr.24030)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 3

    Assié G, Antoni G, Tissier F, Caillou B, Abiven G, Gicquel C, Leboulleux S, Travagli JP, Dromain C & Bertagna X et al. Prognostic parameters of metastatic adrenocortical carcinoma. Journal of Clinical Endocrinology and Metabolism 2007 92 148154. (https://doi.org/10.1210/jc.2006-0706)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 4

    Kebebew E, Reiff E, Duh QY, Clark OH & McMillan A. Extent of disease at presentation and outcome for adrenocortical carcinoma: have we made progress? World Journal of Surgery 2006 30 872878. (https://doi.org/10.1007/s00268-005-0329-x)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 5

    Bilimoria KY, Shen WT, Elaraj D, Bentrem DJ, Winchester DJ, Kebebew E & Sturgeon C. Adrenocortical carcinoma in the United States: treatment utilization and prognostic factors. Cancer 2008 113 31303136. (https://doi.org/10.1002/cncr.23886)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 6

    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. Journal of Clinical Endocrinology and Metabolism 2014 99 455461. (https://doi.org/10.1210/jc.2013-2856)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 7

    Else T, Kim AC, Sabolch A, Raymond VM, Kandathil A, Caoili EM, Jolly S, Miller BS, Giordano TJ & Hammer GD. Adrenocortical carcinoma. Endocrine Reviews 2014 35 282326. (https://doi.org/10.1210/er.2013-1029)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 8

    Ayala-Ramirez M, Jasim S, Feng L, Ejaz S, Deniz F, Busaidy N, Waguespack SG, Naing A, Sircar K & Wood CG et al. Adrenocortical carcinoma: clinical outcomes and prognosis of 330 patients at a tertiary care center. European Journal of Endocrinology 2013 169 891899. (https://doi.org/10.1530/EJE-13-0519)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 9

    Icard P, Goudet P, Charpenay C, Andreassian B, Carnaille B, Chapuis Y, Cougard P, Henry JF & Proye C. Adrenocortical carcinomas: surgical trends and results of a 253-patient series from the French Association of Endocrine Surgeons study group. World Journal of Surgery 2001 25 891897. (https://doi.org/10.1007/s00268-001-0047-y)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 10

    Fassnacht M, Johanssen S, Fenske W, Weismann D, Agha A, Beuschlein F, Führer D, Jurowich C, Quinkler M & Petersenn S et al. Improved survival in patients with stage II adrenocortical carcinoma followed up prospectively by specialized centers. Journal of Clinical Endocrinology and Metabolism 2010 95 49254932. (https://doi.org/10.1210/jc.2010-0803)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 11

    Luton JP, Cerdas S, Billaud L, Thomas G, Guilhaume B, Bertagna X, Laudat MH, Louvel A, Chapuis Y & Blondeau P. Clinical features of adrenocortical carcinoma, prognostic factors, and the effect of mitotane therapy. New England Journal of Medicine 1990 322 11951201. (https://doi.org/10.1056/NEJM199004263221705)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 12

    Fassnacht M, Terzolo M, Allolio B, Baudin E, Haak H, Berruti A, Welin S, Schade-Brittinger C, Lacroix A & Jarzab B et al. Combination chemotherapy in advanced adrenocortical carcinoma. New England Journal of Medicine 2012 366 21892197. (https://doi.org/10.1056/NEJMoa1200966)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 13

    Habra MA, Ejaz S, Feng L, Das P, Deniz F, Grubbs EG, Phan A, Waguespack SG, Ayala-Ramirez M & Jimenez C et al. A retrospective cohort analysis of the efficacy of adjuvant radiotherapy after primary surgical resection in patients with adrenocortical carcinoma. Journal of Clinical Endocrinology and Metabolism 2013 98 192197. (https://doi.org/10.1210/jc.2012-2367)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 14

    Fassnacht M, Hahner S, Polat B, Koschker AC, Kenn W, Flentje M & Allolio B. Efficacy of adjuvant radiotherapy of the tumor bed on local recurrence of adrenocortical carcinoma. Journal of Clinical Endocrinology and Metabolism 2006 91 45014504. (https://doi.org/10.1210/jc.2006-1007)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 15

    Tran TB, Postlewait LM, Maithel SK, Prescott JD, Wang TS, Glenn J, Phay JE, Keplinger K, Fields RC & Jin LX et al. Actual 10-year survivors following resection of adrenocortical carcinoma. Journal of Surgical Oncology 2016 114 971976. (https://doi.org/10.1002/jso.24439)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 16

    Ilias I, Alevizaki M, Philippou G, Anastasiou E & Souvatzoglou A. Sustained remission of metastatic adrenal carcinoma during long-term administration of low-dose mitotane. Journal of Endocrinological Investigation 2001 24 532535. (https://doi.org/10.1007/BF03343888)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 17

    Fujii Y, Kageyama Y, Kawakami S, Masuda H, Arisawa C, Akamatsu H, Akashi T & Kihara K. Successful long-term disease-free survival following multimodal treatments in a patient with a repeatedly recurrent refractory adrenal cortical carcinoma. International Journal of Urology 2003 10 445448. (https://doi.org/10.1046/j.1442-2042.2003.00651.x)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 18

    Nakano R, Satoh D, Nakajima H, Yoshimura Y, Miyoshi H, Yoshida K, Matsukawa H, Shiozaki S, Ichimura K & Okajima M et al. Repeated resections for liver metastasis from primary adrenocortical carcinoma: a case report. International Journal of Surgery Case Reports 2015 9 119122. (https://doi.org/10.1016/j.ijscr.2015.02.041)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 19

    De León DD, Lange BJ, Walterhouse D, Moshang T. Long-term (15 years) outcome in an infant with metastatic adrenocortical carcinoma. Journal of Clinical Endocrinology and Metabolism 2002 87 44524456. (https://doi.org/10.1210/jc.2001-011978)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 20

    El Ghorayeb N, Rondeau G, Latour M, Cohade C, Olney H, Lacroix A, Perrotte P, Sabourin A, Mazzuco TL, Bourdeau I. Rapid and complete remission of metastatic adrenocortical carcinoma persisting 10 years after treatment with mitotane monotherapy: case report and review of the literature. Medicine 2016 95 e3180. (https://doi.org/10.1097/MD.0000000000003180)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 21

    Assié G, Letouzé E, Fassnacht M, Jouinot A, Luscap W, Barreau O, Omeiri H, Rodriguez S, Perlemoine K & René-Corail F et al. Integrated genomic characterization of adrenocortical carcinoma. Nature Genetics 2014 46 607612. (https://doi.org/10.1038/ng.2953)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 22

    Zheng S, Cherniack AD, Dewal N, Moffitt RA, Danilova L, Murray BA, Lerario AM, Else T, Knijnenburg TA & Ciriello G et al. Comprehensive pan-genomic characterization of adrenocortical carcinoma. Cancer Cell 2016 29 723736. (https://doi.org/10.1016/j.ccell.2016.04.002)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 23

    Berruti A, Baudin E, Gelderblom H, Haak HR, Porpiglia F, Fassnacht M, Pentheroudakis G & ESMO Guidelines Working Group. Adrenal cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Annals of Oncology 2012 23 (Supplement 7) vii131vii138. (https://doi.org/10.1093/annonc/mds231)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 24

    Paton BL, Novitsky YW, Zerey M, Harrell AG, Norton HJ, Asbun H, Kercher KW & Heniford BT. Outcomes of adrenal cortical carcinoma in the United States. Surgery 2006 140 914920; discussion 919920. (https://doi.org/10.1016/j.surg.2006.07.035)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 25

    Stojadinovic A, Ghossein RA, Hoos A, Nissan A, Marshall D, Dudas M, Cordon-Cardo C, Jaques DP & Brennan MF. Adrenocortical carcinoma: clinical, morphologic, and molecular characterization. Journal of Clinical Oncology 2002 20 941950. (https://doi.org/10.1200/JCO.2002.20.4.941)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 26

    Abiven G, Coste J, Groussin L, Anract P, Tissier F, Legmann P, Dousset B, Bertagna X & Bertherat J. Clinical and biological features in the prognosis of adrenocortical cancer: poor outcome of cortisol-secreting tumors in a series of 202 consecutive patients. Journal of Clinical Endocrinology and Metabolism 2006 91 26502655. (https://doi.org/10.1210/jc.2005-2730)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 27

    Macfarlane DA. Cancer of the adrenal cortex; the natural history, prognosis and treatment in a study of fifty-five cases. Annals of the Royal College of Surgeons of England 1958 23 155186.

    • Search Google Scholar
    • Export Citation
  • 28

    Sullivan M, Boileau M & Hodges CV. Adrenal cortical carcinoma. Journal of Urology 1978 120 660665. (https://doi.org/10.1016/S0022-5347(17)57317-6)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 29

    Lughezzani G, Sun M, Perrotte P, Jeldres C, Alasker A, Isbarn H, Budäus L, Shariat SF, Guazzoni G & Montorsi F et al. The European Network for the Study of Adrenal Tumors staging system is prognostically superior to the international union against cancer-staging system: a North American validation. European Journal of Cancer 2010 46 713719. (https://doi.org/10.1016/j.ejca.2009.12.007)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 30

    Libé R, Borget I, Ronchi CL, Zaggia B, Kroiss M, Kerkhofs T, Bertherat J, Volante M, Quinkler M & Chabre O et al. Prognostic factors in stage III-IV adrenocortical carcinomas (ACC): an European Network for the Study of Adrenal Tumor (ENSAT) study. Annals of Oncology 2015 26 21192125. (https://doi.org/10.1093/annonc/mdv329)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 31

    Canter DJ, Mallin K, Uzzo RG, Egleston BL, Simhan J, Walton J, Smaldone MC, Master VA, Bratslavsky G & Kutikov A. Association of tumor size with metastatic potential and survival in patients with adrenocortical carcinoma: an analysis of the National Cancer Database. Canadian Journal of Urology 2013 20 69156921.

    • Search Google Scholar
    • Export Citation
  • 32

    Berruti A, Fassnacht M, Haak H, Else T, Baudin E, Sperone P, Kroiss M, Kerkhofs T, Williams AR & Ardito A et al. Prognostic role of overt hypercortisolism in completely operated patients with adrenocortical cancer. European Urology 2014 65 832838. (https://doi.org/10.1016/j.eururo.2013.11.006)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 33

    Margonis GA, Kim Y, Tran TB, Postlewait LM, Maithel SK, Wang TS, Glenn JA, Hatzaras I, Shenoy R & Phay JE et al. Outcomes after resection of cortisol-secreting adrenocortical carcinoma. American Journal of Surgery 2016 211 11061113. (https://doi.org/10.1016/j.amjsurg.2015.09.020)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 34

    Beuschlein F, Weigel J, Saeger W, Kroiss M, Wild V, Daffara F, Libé R, Ardito A, Al Ghuzlan A & Quinkler M et al. Major prognostic role of Ki67 in localized adrenocortical carcinoma after complete resection. Journal of Clinical Endocrinology and Metabolism 2015 100 841849. (https://doi.org/10.1210/jc.2014-3182)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 35

    Cain DW & Cidlowski JA. Immune regulation by glucocorticoids. Nature Reviews Immunology 2017 17 233247. (https://doi.org/10.1038/nri.2017.1)

  • 36

    Sabbaga CC, Avilla SG, Schulz C, Garbers JC & Blucher D. Adrenocortical carcinoma in children: clinical aspects and prognosis. Journal of Pediatric Surgery 1993 28 841843. (https://doi.org/10.1016/0022-3468(93)90341-H)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 37

    Michalkiewicz E, Sandrini R, Figueiredo B, Miranda ECM, Caran E, Oliveira-Filho AG, Marques R, Pianovski MA, Lacerda L & Cristofani LM et al. Clinical and outcome characteristics of children with adrenocortical tumors: a report from the International Pediatric Adrenocortical Tumor Registry. Journal of Clinical Oncology 2004 22 838845. (https://doi.org/10.1200/JCO.2004.08.085)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 38

    Pinto EM, Chen X, Easton J, Finkelstein D, Liu Z, Pounds S, Rodriguez-Galindo C, Lund TC, Mardis ER & Wilson RK et al. Genomic landscape of paediatric adrenocortical tumours. Nature Communications 2015 6 6302. (https://doi.org/10.1038/ncomms7302)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 39

    Zini L, Capitanio U, Jeldres C, Lughezzani G, Sun M, Shariat SF, Isbarn H, Arjane P, Widmer H & Perrotte P et al. External validation of a nomogram predicting mortality in patients with adrenocortical carcinoma. BJU International 2009 104 16611667. (https://doi.org/10.1111/j.1464-410X.2009.08660.x)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 40

    Asare EA, Wang TS, Winchester DP, Mallin K, Kebebew E & Sturgeon C. A novel staging system for adrenocortical carcinoma better predicts survival in patients with stage I/II disease. Surgery 2014 156 13781385; discussion 13851386. (https://doi.org/10.1016/j.surg.2014.08.018)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 41

    Weiss LM. Comparative histologic study of 43 metastasizing and nonmetastasizing adrenocortical tumors. American Journal of Surgical Pathology 1984 8 163169. (https://doi.org/10.1097/00000478-198403000-00001)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 42

    Weiss LM, Medeiros LJ & Vickery AL. Pathologic features of prognostic significance in adrenocortical carcinoma. American Journal of Surgical Pathology 1989 13 202206. (https://doi.org/10.1097/00000478-198903000-00004)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 43

    Volante M, Bollito E, Sperone P, Tavaglione V, Daffara F, Porpiglia F, Terzolo M, Berruti A & Papotti M. Clinicopathological study of a series of 92 adrenocortical carcinomas: from a proposal of simplified diagnostic algorithm to prognostic stratification. Histopathology 2009 55 535543. (https://doi.org/10.1111/j.1365-2559.2009.03423.x)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 44

    Miller BS, Gauger PG, Hammer GD, Giordano TJ & Doherty GM. Proposal for modification of the ENSAT staging system for adrenocortical carcinoma using tumor grade. Langenbeck’s Archives of Surgery 2010 395 955961. (https://doi.org/10.1007/s00423-010-0698-y)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 45

    Pennanen M, Heiskanen I, Sane T, Remes S, Mustonen H, Haglund C & Arola J. Helsinki score-a novel model for prediction of metastases in adrenocortical carcinomas. Human Pathology 2015 46 404410. (https://doi.org/10.1016/j.humpath.2014.11.015)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 46

    Duregon E, Cappellesso R, Maffeis V, Zaggia B, Ventura L, Berruti A, Terzolo M, Fassina A, Volante M & Papotti M. Validation of the prognostic role of the ‘Helsinki Score’ in 225 cases of adrenocortical carcinoma. Human Pathology 2017 62 17. (https://doi.org/10.1016/j.humpath.2016.09.035)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 47

    Tissier F, Aubert S, Leteurtre E, Al Ghuzlan A, Patey M, Decaussin M, Doucet L, Gobet F, Hoang C & Mazerolles C et al. Adrenocortical tumors: improving the practice of the Weiss system through virtual microscopy: a National Program of the French Network INCa-COMETE. American Journal of Surgical Pathology 2012 36 11941201. (https://doi.org/10.1097/PAS.0b013e31825a6308)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 48

    Terzolo M, Boccuzzi A, Bovio S, Cappia S, De Giuli P, Alì A, Paccotti P, Porpiglia F, Fontana D & Angeli A. Immunohistochemical assessment of Ki-67 in the differential diagnosis of adrenocortical tumors. Urology 2001 57 176182. (https://doi.org/10.1016/S0090-4295(00)00852-9)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 49

    Morimoto R, Satoh F, Murakami O, Suzuki T, Abe T, Tanemoto M, Abe M, Uruno A, Ishidoya S & Arai Y et al. Immunohistochemistry of a proliferation marker Ki67/MIB1 in adrenocortical carcinomas: Ki67/MIB1 labeling index is a predictor for recurrence of adrenocortical carcinomas. Endocrine Journal 2008 55 4955. (https://doi.org/10.1507/endocrj.K07-079)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 50

    McNicol AM, Struthers AL, Nolan CE, Hermans J & Haak HR. Proliferation in adrenocortical tumors: correlation with clinical outcome and p53 status. Endocrine Pathology 1997 8 2936. (https://doi.org/10.1007/BF02739705)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 51

    Duregon E, Molinaro L, Volante M, Ventura L, Righi L, Bolla S, Terzolo M, Sapino A & Papotti MG. Comparative diagnostic and prognostic performances of the hematoxylin-eosin and phospho-histone H3 mitotic count and Ki-67 index in adrenocortical carcinoma. Modern Pathology 2014 27 12461254. (https://doi.org/10.1038/modpathol.2013.230)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 52

    Choi YM, Kwon H, Jeon MJ, Sung TY, Hong SJ, Kim TY, Kim WB, Shong YK, Lee JL & Song DE et al. Clinicopathological features associated with the prognosis of patients with adrenal cortical carcinoma: usefulness of the Ki-67 index. Medicine 2016 95 e3736. (https://doi.org/10.1097/MD.0000000000003736)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 53

    Dowsett M, Nielsen TO, A’Hern R, Bartlett J, Coombes RC, Cuzick J, Ellis M, Henry NL, Hugh JC & Lively T et al. Assessment of Ki67 in breast cancer: recommendations from the International Ki67 in Breast Cancer working group. Journal of the National Cancer Institute 2011 103 16561664. (https://doi.org/10.1093/jnci/djr393)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 54

    Papathomas TG, Pucci E, Giordano TJ, Lu H, Duregon E, Volante M, Papotti M, Lloyd RV, Tischler AS & van Nederveen FH et al. An international Ki67 reproducibility study in adrenal cortical carcinoma. American Journal of Surgical Pathology 2016 40 569576. (https://doi.org/10.1097/PAS.0000000000000574)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 55

    Margonis GA, Kim Y, Prescott JD, Tran TB, Postlewait LM, Maithel SK, Wang TS, Evans DB, Hatzaras I & Shenoy R et al. Adrenocortical carcinoma: impact of surgical margin status on long-term outcomes. Annals of Surgical Oncology 2016 23 134141. (https://doi.org/10.1245/s10434-015-4803-x)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 56

    Tissier F, Cavard C, Groussin L, Perlemoine K, Fumey G, Hagneré AM, René-Corail F, Jullian E, Gicquel C & Bertagna X 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 Research 2005 65 76227627. (https://doi.org/10.1158/0008-5472.CAN-05-0593)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 57

    Ragazzon B, Libé R, Gaujoux S, Assié G, Fratticci A, Launay P, Clauser E, Bertagna X, Tissier F & de Reyniès A et al. Transcriptome analysis reveals that p53 and {beta}-catenin alterations occur in a group of aggressive adrenocortical cancers. Cancer Research 2010 70 82768281. (https://doi.org/10.1158/0008-5472.CAN-10-2014)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 58

    Gaujoux S, Grabar S, Fassnacht M, Ragazzon B, Launay P, Libé R, Chokri I, Audebourg A, Royer B & Sbiera S et al. β-Catenin activation is associated with specific clinical and pathologic characteristics and a poor outcome in adrenocortical carcinoma. Clinical Cancer Research 2011 17 328336. (https://doi.org/10.1158/1078-0432.CCR-10-2006)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 59

    Heaton JH, Wood MA, Kim AC, Lima LO, Barlaskar FM, Almeida MQ, Fragoso MCBV, Kuick R, Lerario AM & Simon DP et al. Progression to adrenocortical tumorigenesis in mice and humans through insulin-like growth factor 2 and β-catenin. American Journal of Pathology 2012 181 10171033. (https://doi.org/10.1016/j.ajpath.2012.05.026)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 60

    Kovach AE, Nucera C, Lam QT, Nguyen A, Dias-Santagata D & Sadow PM. Genomic and immunohistochemical analysis in human adrenal cortical neoplasia reveal beta-catenin mutations as potential prognostic biomarker. Discoveries 2015 3 e40. (https://doi.org/10.15190/d.2015.32)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 61

    Libè R, Groussin L, Tissier F, Elie C, René-Corail F, Fratticci A, Jullian E, Beck-Peccoz P, Bertagna X & Gicquel C et al. Somatic TP53 mutations are relatively rare among adrenocortical cancers with the frequent 17p13 loss of heterozygosity. Clinical Cancer Research 2007 13 844850. (https://doi.org/10.1158/1078-0432.CCR-06-2085)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 62

    Waldmann J, Patsalis N, Fendrich V, Langer P, Saeger W, Chaloupka B, Ramaswamy A, Fassnacht M, Bartsch DK & Slater EP. Clinical impact of TP53 alterations in adrenocortical carcinomas. Langenbeck’s Archives of Surgery 2012 397 209216. (https://doi.org/10.1007/s00423-011-0868-6)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 63

    Sbiera S, Schmull S, Assie G, Voelker HU, Kraus L, Beyer M, Ragazzon B, Beuschlein F, Willenberg HS & Hahner S et al. High diagnostic and prognostic value of steroidogenic factor-1 expression in adrenal tumors. Journal of Clinical Endocrinology and Metabolism 2010 95 E161E171. (https://doi.org/10.1210/jc.2010-0653)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 64

    Fenske W, Völker HU, Adam P, Hahner S, Johanssen S, Wortmann S, Schmidt M, Morcos M, Müller-Hermelink HK & Allolio B et al. Glucose transporter GLUT1 expression is an stage-independent predictor of clinical outcome in adrenocortical carcinoma. Endocrine-Related Cancer 2009 16 919928. (https://doi.org/10.1677/ERC-08-0211)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 65

    Waldmann J, Feldmann G, Slater EP, Langer P, Buchholz M, Ramaswamy A, Saeger W, Rothmund M & Fendrich V. Expression of the zinc-finger transcription factor Snail in adrenocortical carcinoma is associated with decreased survival. British Journal of Cancer 2008 99 19001907. (https://doi.org/10.1038/sj.bjc.6604755)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 66

    Pinheiro C, Granja S, Longatto-Filho A, Faria AM, Fragoso MCBV, Lovisolo SM, Lerário AM, Almeida MQ, Baltazar F & Zerbini MCN. Metabolic reprogramming: a new relevant pathway in adult adrenocortical tumors. Oncotarget 2015 6 4440344421. (https://doi.org/10.18632/oncotarget.5623)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 67

    Volante M, Sperone P, Bollito E, Frangipane E, Rosas R, Daffara F, Terzolo M, Berruti A & Papotti M. Matrix metalloproteinase type 2 expression in malignant adrenocortical tumors: diagnostic and prognostic significance in a series of 50 adrenocortical carcinomas. Modern Pathology 2006 19 15631569. (https://doi.org/10.1038/modpathol.3800683)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 68

    Sarkaria IS, Stojadinovic A, Talbot SG, Hoos A, Dudas ME, Brennan MF, Ghossein RA & Singh B. Squamous cell carcinoma-related oncogene is highly expressed in developing, normal, and adenomatous adrenal tissue but not in aggressive adrenocortical carcinomas. Surgery 2004 136 11221128. (https://doi.org/10.1016/j.surg.2004.06.041)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 69

    Faria AM, Sbiera S, Ribeiro TC, Soares IC, Mariani BMP, Freire DS, de Sousa GRV, Lerario AM, Ronchi CL & Deutschbein T et al. Expression of LIN28 and its regulatory microRNAs in adult adrenocortical cancer. Clinical Endocrinology 2015 82 481488. (https://doi.org/10.1111/cen.12607)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 70

    Ip JCY, Pang TCY, Glover AR, Soon P, Zhao JT, Clarke S, Robinson BG, Gill AJ & Sidhu SB. Immunohistochemical validation of overexpressed genes identified by global expression microarrays in adrenocortical carcinoma reveals potential predictive and prognostic biomarkers. Oncologist 2015 20 247256. (https://doi.org/10.1634/theoncologist.2014-0392)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 71

    Gicquel C, Bertagna X, Gaston V, Coste J, Louvel A, Baudin E, Bertherat J, Chapuis Y, Duclos JM & Schlumberger M et al. Molecular markers and long-term recurrences in a large cohort of patients with sporadic adrenocortical tumors. Cancer Research 2001 61 67626767.

    • Search Google Scholar
    • Export Citation
  • 72

    de Fraipont F, El Atifi M, Cherradi N, Le Moigne G, Defaye G, Houlgatte R, Bertherat J, Bertagna X, Plouin PF & Baudin E et al. Gene expression profiling of human adrenocortical tumors using complementary deoxyribonucleic acid microarrays identifies several candidate genes as markers of malignancy. Journal of Clinical Endocrinology and Metabolism 2005 90 18191829. (https://doi.org/10.1210/jc.2004-1075)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 73

    de Reyniès A, Assié G, Rickman DS, Tissier F, Groussin L, René-Corail F, Dousset B, Bertagna X, Clauser E, Bertherat J. Gene expression profiling reveals a new classification of adrenocortical tumors and identifies molecular predictors of malignancy and survival. Journal of Clinical Oncology 2009 27 11081115. (https://doi.org/10.1200/JCO.2008.18.5678)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 74

    Giordano TJ, Kuick R, Else T, Gauger PG, Vinco M, Bauersfeld J, Sanders D, Thomas DG, Doherty G & Hammer G. Molecular classification and prognostication of adrenocortical tumors by transcriptome profiling. Clinical Cancer Research 2009 15 668676. (https://doi.org/10.1158/1078-0432.CCR-08-1067)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 75

    Laurell C, Velázquez-Fernández D, Lindsten K, Juhlin C, Enberg U, Geli J, Höög A, Kjellman M, Lundeberg J & Hamberger B et al. Transcriptional profiling enables molecular classification of adrenocortical tumours. European Journal of Endocrinology 2009 161 141152. (https://doi.org/10.1530/EJE-09-0068)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 76

    Fragoso MCBV, Almeida MQ, Mazzuco TL, Mariani BMP, Brito LP, Gonçalves TC, Alencar GA, Lima LO, Faria AM & Bourdeau I et al. Combined expression of BUB1B, DLGAP5, and PINK1 as predictors of poor outcome in adrenocortical tumors: validation in a Brazilian cohort of adult and pediatric patients. European Journal of Endocrinology 2012 166 6167. (https://doi.org/10.1530/EJE-11-0806)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 77

    Demeure MJ, Coan KE, Grant CS, Komorowski RA, Stephan E, Sinari S, Mount D & Bussey KJ. PTTG1 overexpression in adrenocortical cancer is associated with poor survival and represents a potential therapeutic target. Surgery 2013 154 14051416; discussion 1416. (https://doi.org/10.1016/j.surg.2013.06.058)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 78

    Drelon C, Berthon A, Mathieu M, Ragazzon B, Kuick R, Tabbal H, Septier A, Rodriguez S, Batisse-Lignier M & Sahut-Barnola I et al. EZH2 is overexpressed in adrenocortical carcinoma and is associated with disease progression. Human Molecular Genetics 2016 25 27892800. (https://doi.org/10.1093/hmg/ddw136)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 79

    Ruggiero C, Doghman-Bouguerra M, Sbiera S, Sbiera I, Parsons M, Ragazzon B, Morin A, Robidel E, Favier J & Bertherat J et al. Dosage-dependent regulation of VAV2 expression by steroidogenic factor-1 drives adrenocortical carcinoma cell invasion. Science Signaling 2017 10 eaal2464. (https://doi.org/10.1126/scisignal.aal2464)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 80

    Sbiera S, Sbiera I, Ruggiero C, Doghman-Bouguerra M, Korpershoek E, de Krijger RR, Ettaieb H, Haak H, Volante M & Papotti M et al. Assessment of VAV2 expression refines prognostic prediction in adrenocortical carcinoma. Journal of Clinical Endocrinology and Metabolism 2017 102 34913498. (https://doi.org/10.1210/jc.2017-00984)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 81

    Juhlin CC, Goh G, Healy JM, Fonseca AL, Scholl UI, Stenman A, Kunstman JW, Brown TC, Overton JD & Mane SM et al. Whole-exome sequencing characterizes the landscape of somatic mutations and copy number alterations in adrenocortical carcinoma. Journal of Clinical Endocrinology and Metabolism 2015 100 E493E502. (https://doi.org/10.1210/jc.2014-3282)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 82

    Sidhu S, Marsh DJ, Theodosopoulos G, Philips J, Bambach CP, Campbell P, Magarey CJ, Russell CFJ, Schulte KM & Röher HD et al. Comparative genomic hybridization analysis of adrenocortical tumors. Journal of Clinical Endocrinology and Metabolism 2002 87 34673474. (https://doi.org/10.1210/jcem.87.7.8697)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 83

    Stephan EA, Chung TH, Grant CS, Kim S, Von Hoff DD, Trent JM & Demeure MJ. Adrenocortical carcinoma survival rates correlated to genomic copy number variants. Molecular Cancer Therapeutics 2008 7 425431. (https://doi.org/10.1158/1535-7163.MCT-07-0267)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 84

    Barreau O, de Reynies A, Wilmot-Roussel H, Guillaud-Bataille M, Auzan C, René-Corail F, Tissier F, Dousset B, Bertagna X & Bertherat J et al. Clinical and pathophysiological implications of chromosomal alterations in adrenocortical tumors: an integrated genomic approach. Journal of Clinical Endocrinology and Metabolism 2012 97 E301E311. (https://doi.org/10.1210/jc.2011-1588)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 85

    De Martino MC, Al Ghuzlan A, Aubert S, Assié G, Scoazec JY, Leboulleux S, Do Cao C, Libè R, Nozières C & Lombès M et al. Molecular screening for a personalized treatment approach in advanced adrenocortical cancer. Journal of Clinical Endocrinology and Metabolism 2013 98 40804088. (https://doi.org/10.1210/jc.2013-2165)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 86

    Ronchi CL, Sbiera S, Leich E, Henzel K, Rosenwald A, Allolio B & Fassnacht M. Single nucleotide polymorphism array profiling of adrenocortical tumors – evidence for an adenoma carcinoma sequence? PLoS ONE 2013 8 e73959. (https://doi.org/10.1371/journal.pone.0073959)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 87

    Garinet S, Néou M, de La Villéon B, Faillot S, Sakat J, Da Fonseca JP, Jouinot A, Le Tourneau C, Kamal M & Luscap-Rondof W et al. Calling chromosome alterations, DNA methylation statuses, and mutations in tumors by simple targeted next-generation sequencing: a solution for transferring integrated pangenomic studies into routine practice? Journal of Molecular Diagnostics 2017 19 776787. (https://doi.org/10.1016/j.jmoldx.2017.06.005)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 88

    Barreau O, Assié G, Wilmot-Roussel H, Ragazzon B, Baudry C, Perlemoine K, René-Corail F, Bertagna X, Dousset B & Hamzaoui N et al. Identification of a CpG island methylator phenotype in adrenocortical carcinomas. Journal of Clinical Endocrinology and Metabolism 2013 98 E174E184. (https://doi.org/10.1210/jc.2012-2993)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 89

    Rechache NS, Wang Y, Stevenson HS, Killian JK, Edelman DC, Merino M, Zhang L, Nilubol N, Stratakis CA & Meltzer PS et al. DNA methylation profiling identifies global methylation differences and markers of adrenocortical tumors. Journal of Clinical Endocrinology and Metabolism 2012 97 E1004E1013. (https://doi.org/10.1210/jc.2011-3298)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 90

    Fonseca AL, Kugelberg J, Starker LF, Scholl U, Choi M, Hellman P, Åkerström G, Westin G, Lifton RP & Björklund P et al. Comprehensive DNA methylation analysis of benign and malignant adrenocortical tumors. Genes, Chromosomes and Cancer 2012 51 949960. (https://doi.org/10.1002/gcc.21978)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 91

    Jouinot A, Assie G, Libe R, Fassnacht M, Papathomas T, Barreau O, De la Villeon B, Faillot S, Hamzaoui N & Neou M et al. DNA methylation is an independent prognostic marker of survival in adrenocortical cancer. Journal of Clinical Endocrinology and Metabolism 2016 102 923932. (https://doi.org/10.1210/jc.2016-3205)

    • Search Google Scholar
    • Export Citation
  • 92

    Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 2004 116 281297. (https://doi.org/10.1016/S0092-8674(04)00045-5)

  • 93

    Lu J, Getz G, Miska EA, Alvarez-Saavedra E, Lamb J, Peck D, Sweet-Cordero A, Ebert BL, Mak RH & Ferrando AA et al. MicroRNA expression profiles classify human cancers. Nature 2005 435 834838. (https://doi.org/10.1038/nature03702)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 94

    Soon PSH, Tacon LJ, Gill AJ, Bambach CP, Sywak MS, Campbell PR, Yeh MW, Wong SG, Clifton-Bligh RJ & Robinson BG et al. miR-195 and miR-483-5p identified as predictors of poor prognosis in adrenocortical cancer. Clinical Cancer Research 2009 15 76847692. (https://doi.org/10.1158/1078-0432.CCR-09-1587)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 95

    Tömböl Z, Szabó PM, Molnár V, Wiener Z, Tölgyesi G, Horányi J, Riesz P, Reismann P, Patócs A & Likó I et al. Integrative molecular bioinformatics study of human adrenocortical tumors: microRNA, tissue-specific target prediction, and pathway analysis. Endocrine-Related Cancer 2009 16 895906. (https://doi.org/10.1677/ERC-09-0096)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 96

    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 16301639. (https://doi.org/10.1002/cncr.25724)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 97

    Özata DM, Caramuta S, Velázquez-Fernández D, Akçakaya P, Xie H, Höög A, Zedenius J, Bäckdahl M, Larsson C, Lui WO. The role of microRNA deregulation in the pathogenesis of adrenocortical carcinoma. Endocrine-Related Cancer 2011 18 643655. (https://doi.org/10.1530/ERC-11-0082)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 98

    Schmitz KJ, Helwig J, Bertram S, Sheu SY, Suttorp AC, Seggewiss J, Willscher E, Walz MK, Worm K & Schmid KW. Differential expression of microRNA-675, microRNA-139-3p and microRNA-335 in benign and malignant adrenocortical tumours. Journal of Clinical Pathology 2011 64 529535. (https://doi.org/10.1136/jcp.2010.085621)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 99

    Chabre O, Libé R, Assie G, Barreau O, Bertherat J, Bertagna X, Feige JJ & Cherradi N. Serum miR-483-5p and miR-195 are predictive of recurrence risk in adrenocortical cancer patients. Endocrine-Related Cancer 2013 20 579594. (https://doi.org/10.1530/ERC-13-0051)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 100

    Duregon E, Rapa I, Votta A, Giorcelli J, Daffara F, Terzolo M, Scagliotti GV, Volante M & Papotti M. MicroRNA expression patterns in adrenocortical carcinoma variants and clinical pathologic correlations. Human Pathology 2014 45 15551562. (https://doi.org/10.1016/j.humpath.2014.04.005)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 101

    Feinmesser M, Benbassat C, Meiri E, Benjamin H, Lebanony D, Lebenthal Y, de Vries L, Drozd T, Spector Y. Specific microRNAs differentiate adrenocortical adenomas from carcinomas and correlate with weiss histopathologic system. Applied Immunohistochemistry and Molecular Morphology 2015 23 522531. (https://doi.org/10.1097/PAI.0000000000000117)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 102

    Caramuta S, Lee L, Ozata DM, Akçakaya P, Xie H, Höög A, Zedenius J, Bäckdahl M, Larsson C & Lui WO. Clinical and functional impact of TARBP2 over-expression in adrenocortical carcinoma. Endocrine-Related Cancer 2013 20 551564. (https://doi.org/10.1530/ERC-13-0098)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 103

    de Sousa GRV, Ribeiro TC, Faria AM, Mariani BMP, Lerario AM, Zerbini MCN, Soares IC, Wakamatsu A, Alves VAF & Mendonca BB et al. Low DICER1 expression is associated with poor clinical outcome in adrenocortical carcinoma. Oncotarget 2015 6 2272422733. (https://doi.org/10.18632/oncotarget.4261)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 104

    Patel D, Boufraqech M, Jain M, Zhang L, He M, Gesuwan K, Gulati N, Nilubol N, Fojo T & Kebebew E. MiR-34a and miR-483-5p are candidate serum biomarkers for adrenocortical tumors. Surgery 2013 154 12241228; discussion 1229. (https://doi.org/10.1016/j.surg.2013.06.022)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 105

    Morris KV & Mattick JS. The rise of regulatory RNA. Nature Reviews Genetics 2014 15 423437. (https://doi.org/10.1038/nrg3722)

  • 106

    Prensner JR & Chinnaiyan AM. The emergence of lncRNAs in cancer biology. Cancer Discovery 2011 1 391407. (https://doi.org/10.1158/2159-8290.CD-11-0209)

  • 107

    Glover AR, Zhao JT, Ip JC, Lee JC, Robinson BG, Gill AJ, Soon PSH & Sidhu SB. Long noncoding RNA profiles of adrenocortical cancer can be used to predict recurrence. Endocrine-Related Cancer 2015 22 99109. (https://doi.org/10.1530/ERC-14-0457)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 108

    Szabó DR, Luconi M, Szabó PM, Tóth M, Szücs N, Horányi J, Nagy Z, Mannelli M, Patócs A & Rácz K et al. Analysis of circulating microRNAs in adrenocortical tumors. Laboratory Investigation 2014 94 331339. (https://doi.org/10.1038/labinvest.2013.148)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 109

    Salvianti F, Canu L, Poli G, Armignacco R, Scatena C, Cantini G, Di Franco A, Gelmini S, Ercolino T & Pazzagli M et al. New insights in the clinical and translational relevance of miR483-5p in adrenocortical cancer. Oncotarget 2017 8 6552565533. (https://doi.org/10.18632/oncotarget.19118)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 110

    Creemers SG, Korpershoek E, Atmodimedjo PN, Dinjens WNM, van Koetsveld PM, Feelders RA, Hofland LJ. Identification of mutations in cell-free circulating tumor DNA in adrenocortical carcinoma: a case series. Journal of Clinical Endocrinology and Metabolism 2017 102 36113615. (https://doi.org/10.1210/jc.2017-00174)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 111

    Garinet S, Nectoux J, Neou M, Pasmant E, Jouinot A, Sibony M, Orhant L, Pipoli da Fonseca J, Perlemoine K & Bricaire L et al. Detection and monitoring of circulating tumor DNA in adrenocortical carcinoma. Endocrine-Related Cancer 2018 25 L13L17. (https://doi.org/10.1530/ERC-17-0467)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 112

    Bahrami A, Amerizadeh F, ShahidSales S, Khazaei M, Ghayour-Mobarhan M, Sadeghnia HR, Maftouh M, Hassanian SM & Avan A. Therapeutic potential of targeting Wnt/β-catenin pathway in treatment of colorectal cancer: rational and progress. Journal of Cellular Biochemistry 2017 118 19791983. (https://doi.org/10.1002/jcb.25903)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 113

    Otto T & Sicinski P. Cell cycle proteins as promising targets in cancer therapy. Nature Reviews Cancer 2017 17 93115. (https://doi.org/10.1038/nrc.2016.138)

  • 114

    Suh I, Weng J, Fernandez-Ranvier G, Shen WT, Duh QY, Clark OH & Kebebew E. Antineoplastic effects of decitabine, an inhibitor of DNA promoter methylation, in adrenocortical carcinoma cells. Archives of Surgery 2010 145 226232. (https://doi.org/10.1001/archsurg.2009.292)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 115

    Overman MJ, Morris V, Moinova H, Manyam G, Ensor J, Lee MS, Eng C, Kee B, Fogelman D & Shroff RT et al. Phase I/II study of azacitidine and capecitabine/oxaliplatin (CAPOX) in refractory CIMP-high metastatic colorectal cancer: evaluation of circulating methylated vimentin. Oncotarget 2016 7 6749567506. (https://doi.org/10.18632/oncotarget.11317)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 116

    Glover AR, Zhao JT, Gill AJ, Weiss J, Mugridge N, Kim E, Feeney AL, Ip JC, Reid G & Clarke S et al. MicroRNA-7 as a tumor suppressor and novel therapeutic for adrenocortical carcinoma. Oncotarget 2015 6 3667536688. (https://doi.org/10.18632/oncotarget.5383)

    • PubMed
    • Search Google Scholar
    • Export Citation

 

  • Collapse
  • Expand

     European Society of Endocrinology

Sept 2018 onwards Past Year Past 30 Days
Abstract Views 0 0 0
Full Text Views 1114 1114 64
PDF Downloads 761 761 70
  • View in gallery

    Disease-specific survival stratified on the 2008 ENSAT staging classification. From Fassnacht et al. (2). HR, hazard ratio with 95% confidence intervals; P, log-rank P-value.

  • View in gallery

    Recurrence-free (A) and overall (B) survival stratified on the Ki67 index in stages I–III ACC after R0 surgery. From Beuschlein et al. (34).

  • View in gallery

    Integrated genomics of ACC. From Assié et al. (A and B) and Zheng et al. (C and D). (A) Integrated genomics of the ENSAT cohort. Tumors were divided into 2 groups from mRNA expression. DNA methylation, miRNA expression clusters, mutation rate and alterations in key genes and pathways converge with transcriptome clusters. (B) Overall survival stratified on the ENSAT genomic classification. (C) Integrated genomics of the TCGA cohort. Tumors were divided into 3 groups by clustering the clusters from mRNA expression (red), chromosome alterations profiles (black), DNA methylation profiles (blue); miRNA expression (purple). (D) Event-free survival stratified on the TCGA genomic classification.

  • View in gallery

    Global molecular classification of ACC. Gene expression, chromosome alterations and DNA methylation profiles divided ACC into 3 prognostic groups. These groups are also associated with some other molecular events – including miRNA expression and somatic mutations – and clinical characteristics. Gene expression profiling defines poor/intermediate prognostic ‘C1A’ and better prognostic ‘C1B’ clusters. The ‘C1A’ group of poor prognosis is also characterized by ‘noisy’ chromosome profile (numerous chromosome losses and gains) and hypermethylation in CpG islands (CIMP). The ‘C1A’ group of intermediate prognosis displays ‘noisy’ profile or ‘CIMP’ but not both. The ‘C1B’ group shows a small number of molecular events – the most common alteration being a downregulation of the DLK1-MEG3 miR cluster (14q32.2) – and a better prognosis.

  • 1

    Kerkhofs TMA, Verhoeven RHA, Van der Zwan JM, Dieleman J, Kerstens MN, Links TP, Van de Poll-Franse LV & Haak HR. Adrenocortical carcinoma: a population-based study on incidence and survival in the Netherlands since 1993. European Journal of Cancer 2013 49 25792586. (https://doi.org/10.1016/j.ejca.2013.02.034)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 2

    Fassnacht M, Johanssen S, Quinkler M, Bucsky P, Willenberg HS, Beuschlein F, Terzolo M, Mueller HH, Hahner S & Allolio B 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 243250. (https://doi.org/10.1002/cncr.24030)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 3

    Assié G, Antoni G, Tissier F, Caillou B, Abiven G, Gicquel C, Leboulleux S, Travagli JP, Dromain C & Bertagna X et al. Prognostic parameters of metastatic adrenocortical carcinoma. Journal of Clinical Endocrinology and Metabolism 2007 92 148154. (https://doi.org/10.1210/jc.2006-0706)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 4

    Kebebew E, Reiff E, Duh QY, Clark OH & McMillan A. Extent of disease at presentation and outcome for adrenocortical carcinoma: have we made progress? World Journal of Surgery 2006 30 872878. (https://doi.org/10.1007/s00268-005-0329-x)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 5

    Bilimoria KY, Shen WT, Elaraj D, Bentrem DJ, Winchester DJ, Kebebew E & Sturgeon C. Adrenocortical carcinoma in the United States: treatment utilization and prognostic factors. Cancer 2008 113 31303136. (https://doi.org/10.1002/cncr.23886)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 6

    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. Journal of Clinical Endocrinology and Metabolism 2014 99 455461. (https://doi.org/10.1210/jc.2013-2856)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 7

    Else T, Kim AC, Sabolch A, Raymond VM, Kandathil A, Caoili EM, Jolly S, Miller BS, Giordano <