ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Clinical presentation and evaluation of adrenocortical tumors

Clinical presentation and evaluation of adrenocortical tumors
Literature review current through: Jan 2024.
This topic last updated: Apr 30, 2021.

INTRODUCTION — Unilateral tumors or masses of the adrenal gland are common. They are categorized as either functional (hormone-secreting) or silent and as either benign or malignant.

The majority of adrenocortical tumors are benign, nonfunctioning adenomas that are discovered incidentally on abdominal imaging studies (adrenal incidentalomas). (See "Evaluation and management of the adrenal incidentaloma".)

Others are benign, hormone-secreting adenomas that cause Cushing's syndrome, primary aldosteronism, or, much less commonly, virilization.

Adrenocortical carcinomas (ACCs) are rare, often aggressive tumors that may be functional and cause Cushing's syndrome and/or virilization, or nonfunctional and present as an abdominal mass or an incidental finding.

Pheochromocytomas are catecholamine-secreting tumors that arise from chromaffin cells of the adrenal medulla. They may be benign or malignant. These are discussed separately. (See "Treatment of pheochromocytoma in adults".)

The clinical presentation and evaluation of adrenocortical adenomas and staging of carcinomas will be reviewed here. Evaluation of adrenal incidentalomas and treatment of adrenocortical adenomas and carcinomas are discussed separately. The rarer bilateral micronodular and macronodular adrenal disorders that cause Cushing's syndrome are also reviewed separately. (See "Evaluation and management of the adrenal incidentaloma" and "Treatment of adrenocortical carcinoma" and "Cushing's syndrome due to primary bilateral macronodular adrenal hyperplasia" and "Cushing syndrome due to primary pigmented nodular adrenocortical disease".)

ADRENOCORTICAL ADENOMAS — Adrenocortical adenomas are benign neoplasms of adrenocortical cells that can secrete steroids independently from corticotropin (ACTH) or the renin-angiotensin system. They can produce syndromes of hypercortisolism and hyperaldosteronism and rarely produce virilization or feminization. Many are discovered incidentally on abdominal imaging studies (adrenal incidentalomas), while others present with symptoms of hormonal excess.

Incidentalomas — Adrenal tumors are most often discovered as an incidental finding, in over 4 percent of high-resolution abdominal imaging studies. Although most incidentalomas are nonfunctional, as many as 15 percent are functional, causing low levels of autonomous cortisol secretion (previously called subclinical Cushing's syndrome) [1,2]. In one series of 1004 adrenal incidentalomas, 85 percent were nonfunctional, 9.2 percent were found to secrete low levels of cortisol, 4.2 percent were pheochromocytomas, and 1.6 percent were aldosteronomas [3].

Adenomas secreting low levels of cortisol and pheochromocytoma are sufficiently common that all patients with an adrenal incidentaloma should be screened for these disorders. In addition, hypertensive patients should be screened for primary aldosteronism, even if the serum potassium concentration is normal. The approach to the adrenal incidentaloma is reviewed in detail separately. (See "Evaluation and management of the adrenal incidentaloma" and "Epidemiology and clinical manifestations of Cushing syndrome" and "Clinical presentation and diagnosis of pheochromocytoma" and "Pathophysiology and clinical features of primary aldosteronism".)

Pathogenesis — It is thought that the majority of adrenal tumors are of monoclonal origin. Most adrenocortical carcinomas (ACCs) are sporadic, but some occur as a component of hereditary cancer syndromes. (See 'Hereditary cancer syndromes' below.)

Aberrant receptors — Cortisol hypersecretion is the most frequent hormonal abnormality detected in patients with functioning unilateral adrenal adenomas. It had been assumed that the mechanism for this was non-ACTH-dependent autonomous cortisol secretion from the adenoma. However, it is now known that in a proportion of cases, particularly those with modest secretion [4], cortisol can be regulated by the aberrant expression and activation of G-protein coupled receptors (GPCRs) distinct from the melanocortin 2 (MC2)-ACTH receptor in the adrenocortical tissues. (See "Evaluation and management of the adrenal incidentaloma".)

The role of aberrant hormone receptors in adrenal adenomas, and more frequently in bilateral macronodular adrenal hyperplasia (BMAH), is reviewed in detail separately. (See "Causes and pathophysiology of Cushing syndrome", section on 'Primary adrenocortical hyperfunction' and "Cushing's syndrome due to primary bilateral macronodular adrenal hyperplasia".)

Reports of small numbers of cases indicate that one or more aberrant receptors may also be implicated in the renin-independent aldosterone-secreting adenomas and bilateral idiopathic hyperaldosteronism (IHA), including ACTH, gastric inhibitory polypeptide (GIP), 5-hydroxytryptamine receptor 4 (5-HT4), luteinizing hormone (LH), gonadotropin-releasing hormone (GnRH), vasopressin, or thyroid-stimulating hormone (TSH) [5-8].

Beta-catenin mutations (CTNNB1) — Constitutive activation of beta-catenin in the Wnt signaling pathway has been identified as a frequent alteration in benign and malignant adrenocortical tumors [9,10]. The increased occurrence of adrenal tumors in patients with mutations of adenomatous polyposis coli (APC) suggested that the Wnt/beta-catenin pathway could be involved in adrenal tumorigenesis [11]. This pathway is essential for embryonic development of the adrenal [12], and its ectopic constitutive activation is associated with cancer development in a number of tissues [13,14].

Activating mutations of exon 3 of the CTNNB1 gene (beta-catenin) are frequent in adrenocortical adenomas [15-21]. In one series of 39 adrenocortical tumors, an activating somatic mutation of the CTNNB1 gene was identified in 7 of 26 adrenocortical adenomas (27 percent) and in 4 of 13 ACCs (31 percent); most were point mutations altering the Ser45 of exon 3 [15]. In a second report, genetic alterations of exon 3 of beta-catenin were found in 5 of 33 adenomas (15 percent) but none in 4 ACCs or 13 BMAH [16].

In a larger study of 100 surgically removed adrenal adenomas (excluding aldosteronomas), 36 percent had CTNNB1 mutations, mostly in larger and nonsecreting adenomas, suggesting that the Wnt/beta-catenin pathway activation is associated with the development of less differentiated tumors [17]. Somatic CTNNB1 mutations may explain only approximately 50 percent of beta-catenin accumulation observed in adrenocortical tumors, indicating that other components of the Wnt pathway may be involved.

Mutations in adrenal Cushing's syndrome — Somatic mutations of protein kinase A (PKA) catalytic subunit (PRKACA) were identified in 22 of 59 adenomas (37 percent) from patients with overt Cushing's syndrome but not in adenomas secreting less cortisol [22]. This was confirmed in a second report; 35 percent of cases with overt Cushing's syndrome had the same mutation compared with only 11 percent of those with subclinical cortisol secretion [18]. In additional reports, the same mutation was found in over 50 percent of patients with Cushing's syndrome due to adrenal adenomas [10,19,21,23]. The most frequent hotspot p.Leu206Arg mutation is located in the active cleft of the catalytic subunit, inactivating the site where the regulatory subunit RII-beta usually binds, thus causing a constitutive PKA activation. Examination of a larger number of samples collected in several European centers identified additional somatic mutations of PRKACA with similar functional consequences; patients with PRKACA mutations had higher levels of cortisol after dexamethasone test and smaller adenomas than nonmutated ones [21,24]. The infrequent presence of PRKACA mutation in low cortisol-secreting adenomas may explain why they rarely progress to overt Cushing's syndrome with time.

Somatic activating mutations of the Gs alpha subunit (GNAS) similar to those found in endocrine tumors of patients with McCune-Albright syndrome have been identified in 5 to 17 percent of cortisol-secreting adrenal adenomas [18,21,23]. Adenomas with PRKACA or GNAS mutations appear to be significantly smaller and secrete more cortisol than adenomas without these mutations, and individuals with these tumors presented at significantly younger ages [18,23].

Mutations in aldosterone-producing adenomas — The most frequent causes of primary aldosteronism include bilateral IHA (60 to 70 percent) and unilateral aldosterone-producing adenoma (APA, 30 to 40 percent). (See "Diagnosis of primary aldosteronism".)

This classical distinction between APA and IHA was challenged by the observation of adrenal cortex remodeling, with increased nodulation, reduced vascularization, and functional zona glomerulosa hyperplasia in the adrenal cortex adjacent to APAs [25,26].

Somatic mutations in KCNJ5 have been identified in patients with primary aldosteronism due to APAs. These mutations are more common in women than men; APAs with KCNJ5 mutations are larger than those without mutations. Somatic mutations in other important genes implicated in regulation of aldosterone synthesis (ATP1A1, ATP2B3, CACNA1D, CTNNB1, CLCN2 chloride channel ARMC5) have also been identified [27]. These mutations are reviewed in greater detail separately. (See "Pathophysiology and clinical features of primary aldosteronism", section on 'Mutations in aldosterone-producing adenomas'.)

Germline mutations in KCNJ5 or other ion channel genes have been identified in rare families with familial hyperaldosteronism type III but not in patients with APA or IHA. (See "Familial hyperaldosteronism", section on 'Mutations in KCNJ5 gene'.)

Clinical features

Cushing's syndrome — Approximately 10 percent of cases of overt Cushing's syndrome are due to adrenal adenomas. Both the degree of hypercortisolemia and many of the clinical manifestations of Cushing's syndrome tend to be less severe in patients >50 years of age [28]; this may be related to the fact that younger patients with overt Cushing's syndrome are more likely to harbor PRKACA mutations [18,19,22-24]. As noted above, low levels of ACTH-independent cortisol secretion (mild hypercortisolism without clinical manifestations of Cushing's syndrome) is common in patients with adrenal incidentalomas. However, glucose intolerance and hypertension are common in these patients [29], and patients may be at increased risk of cardiovascular morbidity [30,31]. The causes, clinical features, and diagnosis of Cushing's syndrome are reviewed in detail separately. (See "Causes and pathophysiology of Cushing syndrome" and "Epidemiology and clinical manifestations of Cushing syndrome" and "Establishing the diagnosis of Cushing syndrome".)

Primary aldosteronism — Aldosteronomas are rare causes of adrenal incidentaloma because most have been diagnosed by the time they reach a size detectable by computed tomography (CT) or magnetic resonance imaging (MRI). However, using more carefully selected control groups to establish normal cutoffs and 48-hour low-dose dexamethasone tests and saline loading test under dexamethasone in 151 patients with adrenal incidentalomas, the prevalence of aldosterone excess was 24 percent, with combined excess of cortisol and aldosterone in 12 percent [32,33]. Renin-independent, incompletely suppressible (primary) hypersecretion of aldosterone is an increasingly recognized but still underdiagnosed cause of hypertension [34]; it is estimated to be responsible for 5 to 13 percent of hypertension in humans and can reach 20 percent in those with resistant hypertension and up to 35 percent of patients with sleep apnea. The classic presenting signs of primary aldosteronism were previously hypertension and hypokalemia, but potassium levels are frequently normal in modern-day series of aldosteronomas; screening for primary aldosteronism should be conducted in all patients with adrenal incidentaloma and hypertension. (See "Pathophysiology and clinical features of primary aldosteronism" and "Diagnosis of primary aldosteronism", section on 'Variable presentation'.)

Androgen and estrogen-secreting tumors — Androgen-secreting adrenal tumors are usually malignant, but benign tumors have also been described in women. In a report of 21 women with androgen-secreting tumors, malignant tumors (n = 10) were larger at presentation than benign tumors (n = 11; 14 versus 9 cm, respectively). In addition, serum testosterone levels were 2.6-fold higher in the women with malignant tumors [35]. Benign cortisol-secreting adenomas can also produce small amounts of androgens, but the serum androgen levels are usually not elevated [36].

Estrogen-secreting tumors are rare and are usually malignant [37,38]. In males, it can present as feminization with gynecomastia, decreased libido, testicular atrophy; in women, it can present with breast tenderness and dysfunctional uterine bleeding.

Imaging — The maximum diameter of the adrenal mass is predictive of malignancy. Most adrenal adenomas are less than 4 cm in diameter. In contrast, most ACCs are greater than 4 cm in diameter when discovered. (See "Evaluation and management of the adrenal incidentaloma", section on 'Size'.)

The "imaging phenotype" of an adrenal tumor refers to the characteristics of the mass on CT or MRI. The lipid-rich nature of cortical adenomas is helpful in distinguishing this benign tumor from ACC. Evaluation of lipid content, through assessment of mass density, may be even more informative than the size of the mass in capturing either primary or secondary adrenal malignancies. (See "Evaluation and management of the adrenal incidentaloma", section on 'Imaging phenotype'.)

As an example, CT attenuation of a benign adenoma, as expressed in Hounsfield units (HU), is usually <10 on an unenhanced scan (ie, has the density of fat), which suggests that the likelihood that it is a benign adenoma is nearly 100 percent.

In contrast, ACCs are usually of larger size, have higher attenuation values, and may also display features such as inhomogeneity, irregular borders, calcifications, invasion of surrounding structures or lymph node enlargement. Precontrast HU >20 and <50 percent contrast washout at 10 minutes is more suspicious for malignancy [39]. (See "Evaluation and management of the adrenal incidentaloma", section on 'Imaging phenotype'.)

Although CT remains the primary adrenal imaging procedure, MRI has advantages in certain clinical situations to characterize local invasion from adrenal carcinoma. (See "Evaluation and management of the adrenal incidentaloma", section on 'MRI'.)

The use of fluorodeoxyglucose (FDG)-positron emission tomography (PET) scanning is emerging as a useful tool to distinguish ACC from benign adenomas with elevated HU or delayed contrast washout values [40]. (See 'Radiographic studies' below.)

Metomidate (MTO) is an inhibitor of 11-beta-hydroxylase (CYP11B1) and aldosterone synthetase (CYP11B2), with high affinity for these enzymes. C-MTO PET can distinguish tumors of adrenocortical origin from noncortical lesions but cannot distinguish benign from malignant adrenocortical lesions [41,42]. Wider availability and combination of FDG and MTO PET studies may well be useful to distinguish primary and isolated metastatic adrenal lesions from benign adrenocortical lesions.

ADRENOCORTICAL CARCINOMA

Epidemiology — Adrenocortical carcinomas (ACCs) are rare; the incidence is approximately one to two per million population per year [43-45]. However, the incidence is approximately 10-fold higher in children in southern Brazil, where a number of environmental and genetic risk factors have been identified [46,47].

Although ACC can develop at any age, there is a bimodal age distribution, with disease peaks before the age of five and in the fourth to fifth decade of life [43]. In general, the level of aggressiveness and pace of disease progression are more rapid in adults than in children.

Women develop ACCs more often than men (female-to-male ratio 1.5 to 2.5:1) [48-50]. In the southern Brazil population, girls are also more commonly affected than boys (1.6:1) [51]. Studies using human adrenal cancer cell line suggest proliferative effects of estrogen, but it is unclear whether this may explain the higher occurrence of ACC in women [52].

Pathogenesis

Hereditary cancer syndromes — Although most cases of ACC appear to be sporadic, some have been described as a component of several hereditary cancer syndromes [53,54]:

Li-Fraumeni syndrome (also known as the Sarcoma, Breast, Leukemia, and Adrenal gland [SBLA] cancer syndrome) is inherited as an autosomal dominant disorder and is associated with inactivating mutations of the TP53 tumor suppressor gene on chromosome 17p. (See "Li-Fraumeni syndrome".)

Beckwith-Wiedemann syndrome (Wilms' tumor, neuroblastoma, hepatoblastoma, and ACC), associated with abnormalities in 11p15. (See "Congenital cytogenetic abnormalities" and "Rhabdomyosarcoma in childhood and adolescence: Epidemiology, pathology, and molecular pathogenesis".)

Multiple endocrine neoplasia type 1 (MEN1; parathyroid, pituitary, and pancreatic neuroendocrine tumors and adrenal adenomas, as well as carcinomas), associated with inactivating mutations of the MEN1 gene on chromosome 11q. Unilateral or bilateral adrenal tumors can be found in 20 to 40 percent of patients with MEN1; the majority are benign tumors, usually nonfunctional, but they can present with excess production of aldosterone or cortisol. (See "Multiple endocrine neoplasia type 1: Genetics".)

Sporadic ACC — Although the molecular mechanisms underlying tumorigenesis in many of the hereditary syndromes described above are well characterized, the molecular pathogenesis of sporadic adrenocortical carcinomas (ACCs) is less well understood [55]. A multistep tumor progression model, similar to that described for colorectal cancer, has been proposed [53,56,57]. (See "Molecular genetics of colorectal cancer".)

TP53 gene, located on chromosome 17p13, is the most frequently mutated gene in human cancers. A role for the TP53 tumor suppressor gene in sporadic ACCs is suggested by the frequent finding of loss of heterozygosity (LOH) at the 17p13 locus in sporadic ACCs [58,59]. Although loss of heterozygosity at 17p13 is common, only approximately one-third of these tumors have a mutation of TP53 [60-67]. This suggests that another as yet unidentified suppressor gene is present in this locus [67].

In contrast, a distinct germline TP53 mutation (R337H) has been identified in a high percentage of children with adrenocortical tumors from southern Brazil [60,61] and North America [68,69], which may at least partly explain the higher frequency of ACC in this population. Amplification of the steroidogenic factor 1 (SF-1) gene in tumors also has been reported in this population [70].

In a study that used next-generation sequencing to determine the contribution of germline predisposing gene mutations in 1051 children and adolescents with cancer, mutations that were deemed to be pathogenic or probably pathogenic were identified in 90 patients (8.5 percent). The 39 patients with adrenocortical cancers carried the highest prevalence of mutations (69 percent), which were mostly in the TP53 gene [71]. Importantly, family history did not predict the presence of an underlying predisposition syndrome in most patients.

Another chromosomal locus that is strongly implicated in the pathogenesis of ACC is 11p, the area of abnormality in Beckwith-Wiedemann syndrome [72] and the site of the insulin-like growth factor-2 (IGF-2) gene. LOH at the 11p15 locus and overexpression of IGF-2 have been associated with the malignant phenotype in sporadic ACCs [58,73,74]. However, other growth-related tumor suppressor genes at this locus may also be involved [59].

Activating somatic mutation of the CTNNB1 gene has also been identified in ACCs (see 'Beta-catenin mutations (CTNNB1)' above). Wnt/beta-catenin pathway activation was an independent predictor of less favorable disease-free and overall survival in patients with resected primary adrenal carcinoma [75].

Using exome sequencing, nucleotide polymorphisms arrays, and confirmation in 123 ACCs, mutations in ZNRF3, encoding a cell-surface E3 ubiquitin ligase, were identified in 21 percent [76]. ZNRF3 is thought to be a potential tumor suppressor gene related to the beta-catenin pathway. Other known mutations associated with ACC were also observed (CTNNB1, TP53, CDKN2A [cyclin-dependent kinase inhibitor 2A], RB1 [retinoblastoma 1], and MEN1 [multiple endocrine neoplasias]). Two distinct groups of ACCs, those with poor or good outcome, could be identified by their genetic features. A comprehensive Cancer Genome Atlas (TCGA) group characterization of ACC identified additional driver genes, including PRKAR1A, RPL22, TERF2, CCNE1, and NF1. Whole-genome doubling was associated with an aggressive clinical course increased telomerase reverse transcriptase (TERT) [77].

There is an emerging interest in detecting such mutations in circulating cell-free tumor DNA for the follow-up of patients with ACC [78].

Clinical presentation — Approximately 60 percent of ACCs are sufficiently secretory to present clinical syndrome of hormone excess [43,45,49,79-81]. Adults with hormone-secreting ACCs usually present with Cushing's syndrome alone (45 percent), or a mixed Cushing's and virilization syndrome, with overproduction of both glucocorticoids and androgens (25 percent) [43,82]. Fewer than 10 percent present with virilization alone, but the presence of virilization in a patient with an adrenal neoplasm suggests an ACC rather than an adenoma.

The clinical symptoms associated with glucocorticoid excess, such as weight gain, weakness, and insomnia, usually develop very rapidly (over three to six months). Patients who have coexisting hypersecretion of adrenal androgens may not experience the typical catabolic effects of glucocorticoid excess (muscle and skin atrophy).

Feminization and hyperaldosteronism occur in fewer than 10 percent of cases [43].

Most patients with nonfunctioning tumors (or more precisely with modest production of steroids) present with clinical manifestations related to tumor growth (ie, abdominal or flank pain) or with an incidentally found adrenal mass detected on radiographic imaging performed for a different reason; constitutional symptoms (weight loss, anorexia) are frequent in this clinical context (see "Evaluation and management of the adrenal incidentaloma"). Uncommonly, a varicocele or fever and leucocytosis from tumor necrosis or production of chemokines may occur [83].

The impact of clinical characteristics on outcome of ACC is controversial. In initial cohort studies, nonfunctioning ACCs were more common in older adults and tended to progress more rapidly than functioning tumors [84,85]. However, in a second study, patients with functioning ACC and cortisol hypersecretion (versus androgen secretion) had shorter survival (hazard ratio [HR] 3.9) [86]. Overt Cushing's syndrome represents an important cause of morbidity when patients are treated surgically or with chemotherapy, due to the increased risk of infections and metabolic or vascular complications.

Children usually present with virilization (84 percent), while isolated glucocorticoid excess (Cushing's syndrome) is much less common (6 percent) [51,87].

Diagnostic evaluation — A careful history and physical examination should be performed to exclude signs and symptoms of pheochromocytoma, hyperaldosteronism, hyperandrogenism, and Cushing's syndrome.

Hormonal evaluation — The evaluation in apparently asymptomatic patients has been debated. Even in asymptomatic patients, the European Network for the Study of Adrenal Tumors (ENSAT) recommends performing the following tests to determine the secretory activity of the tumor: fasting blood glucose, serum potassium, cortisol, corticotropin (ACTH), 24-hour urinary free cortisol, fasting serum cortisol at 8 AM following a 1 mg dose of dexamethasone at bedtime, adrenal androgens (dehydroepiandrosterone sulfate [DHEAS], androstenedione, testosterone, 17-hydroxyprogesterone), and serum estradiol in men and postmenopausal women [88].

Adrenal carcinomas are typically inefficient steroid producers, but they secrete excessive amounts of adrenal steroid precursors due to decreased expression of several steroidogenic enzymes (which also results in diminished cortisol production). Even in patients with adrenal carcinomas who presumably did not produce excess steroids, more sensitive methods such as gas chromatography/mass spectrometry identify increased urinary metabolites of several steroids and precursors of androgens (pregnanediol, pregnanetriol, androsterone, etiocholanolone) or glucocorticoids (17-hydroxyprogesterone, tetrahydro-11-deoxycortisol, cortisol, 6-hydroxycortisol, tetrahydrocortisol, and alpha-cortol); this is different from cortisol-secreting adenomas, which produce cortisol, but little androgens [89]. Low serum aldosterone concentrations, but normal or high serum or urinary concentrations of aldosterone precursors (ie, deoxycorticosterone, 18-hydroxydeoxycorticosterone, corticosterone, and 18-hydroxycorticosterone, tetrahydro-11-deoxycorticosterone [THDOC], and 5 alpha-THDOC) are found in most adrenal carcinomas but not in adrenal adenomas [89,90].

The ENSAT also recommends that plasma metanephrines or urinary metanephrines and catecholamines be obtained in all patients to exclude pheochromocytoma and that plasma aldosterone and renin be obtained in patients with hypertension and/or hypokalemia (see "Establishing the cause of Cushing syndrome" and "Adrenal hyperandrogenism" and "Pathophysiology and clinical features of primary aldosteronism" and "Evaluation and management of the adrenal incidentaloma"). Hormonal evaluation may help in establishing the adrenal origin of the tumor and provide tumor markers that can be useful during follow-up to estimate the presence of residual tumor or tumor recurrence after surgery.

Radiographic studies — As noted above, computed tomography (CT) scanning can usually distinguish adenomas from ACCs. Magnetic resonance imaging (MRI) is complementary to CT, in that local invasion and involvement of the vena cava are more readily identifiable. These and other radiographic imaging studies that are useful in the evaluation of adrenal masses are discussed elsewhere. (See "Evaluation and management of the adrenal incidentaloma", section on 'Evaluation for malignancy'.)

Positron emission tomography (PET) scanning with fluorodeoxyglucose (FDG) is of value for identifying unilateral adrenal tumors with a higher index of suspicion for malignancy [40,90-94]. In a report of 26 patients with nonsecreting adrenal masses (13 benign, 13 malignant [6 ACC, 1 sarcoma, 6 metastases]), FDG uptake was observed in all of the malignant and none of the benign lesions [91]. However, others have shown that a minority of benign cortical adenomas accumulate FDG, although to a lesser extent [40,95].

Integrated or "fused" PET-CT imaging improves the performance of PET because adrenal adenomas can be better differentiated from nonadenomas using a combination of CT attenuation measurements plus the intensity of FDG uptake, as described by the standardized uptake value (SUV) for the adrenal lesion [96-98]. In the largest series, 150 patients with a total of 175 adrenal masses underwent PET-CT; the final diagnosis was based on histology in six cases, prolonged imaging follow-up in 118, and morphologic imaging criteria in 51 [96]. Using PET alone (with an SUV cutoff of 3.1), the sensitivity, specificity, positive predictive value, and negative predictive values for malignant lesions versus adenomas were 99, 92, 89, and 99 percent, respectively. The corresponding values for PET-CT were 100, 98, 97, and 100 percent, respectively. PET-CT correctly classified all masses less than 1.5 cm in diameter.

Alternative PET scan tracers are under study (eg, 11C-metomidate [MTO]) that might further improve specificity [41,99,100].

The most common sites of distant spread for ACC are the liver, lungs, lymph nodes, and bone [43,86]. For this reason, CT imaging of the chest and liver, as well as bone scan, are typically included in the staging workup if an ACC is suspected based upon the imaging evaluation or clinical presentation. It is not yet clear that PET and/or PET-CT are sufficiently accurate to replace all of these other radiographic studies as a single staging method in patients with suspected ACC [92]. While PET is more sensitive than CT or radiographic bone scans for distant metastases in a variety of clinical settings, small lesions may be missed [90].

On the other hand, FDG-PET-CT is complementary to regular CT in the follow-up of patients with adrenal carcinoma, and it is increasingly utilized in the regular follow-up evaluations; however, its precise role (first-line test, second-line after positive/dubious conventional imaging) and timing remain to be better defined [88].

Fine-needle aspiration biopsy — Cytology from a specimen obtained by fine-needle aspiration (FNA) cannot distinguish a benign adrenal mass from adrenal carcinoma. It can, however, distinguish between an adrenal tumor and a metastatic tumor.

Thus, FNA is sometimes performed when there is a suspicion of cancer outside the adrenal gland or in the patient undergoing a staging evaluation for a known cancer [101,102]. Pheochromocytoma should always be excluded by biochemical testing before attempting FNA biopsy of an adrenal mass. (See "Clinical presentation and diagnosis of pheochromocytoma", section on 'Approach to initial evaluation'.)

Even when diagnostic material is available, the distinction between benign and malignant adrenocortical tumors may be difficult and should only be made by a pathologist experienced in using the microscopic Weiss criteria [103,104]. The only definitive diagnostic criterion for a malignant adrenocortical tumor is distant metastasis or the presence of local invasion. In the absence of these findings, the Weiss histopathologic system is the most commonly used method for assessing the likelihood of malignant behavior because of its simplicity and reliability [103,105]. The five criteria used in the updated Weiss system include: >6 mitoses/50 high-power fields, ≤25 percent clear tumor cells in cytoplasm, abnormal mitoses, necrosis, and capsular invasion [105]. Each criterion is scored 0 when absent, or 2 for the first two criteria and 1 for the last three when present; the threshold for malignancy is a total score ≥3.

Other immunohistochemical criteria utilized include Ki-67 proliferation index, but cutoff values between benign and malignant lesions vary from 1.5 to 10 percent [106]. Overexpression of TP53, IGF-2, and cyclin E are found in ACC but are not sufficiently discriminatory [106]. Several markers (such as alpha-inhibin, Melan A, SF-1) can confirm the primary adrenal origin [88,107].

Staging — A variety of staging systems have been used for adrenocortical cancer [72,80,81,108-111]. The variability in disease classification used in individual studies complicates the comparison of reported results.

Adults — The eighth tumor, node, metastases (TNM) staging system from the combined American Joint Committee on Cancer (AJCC)/Union for International Cancer Control (UICC) was revised in 2017 for implementation in January 2018 (table 1) [112]. The TNM classification definitions were the same as those initially proposed by the UICC/World Health Organization (WHO) in 2004 [109], which were in turn based upon the Sullivan modification of the original McFarlane staging system [72].

In the eighth edition of the TNM staging system, stage IV disease now, in contrast to the previous 2004 AJCC/UICC classification (table 2), is limited to patients with distant metastases, and those with local invasion or invasion of adjacent organs (T3 or T4) but without distant metastases are classified as having stage III disease. These changes are consistent with the modifications initially proposed by the ENSAT group [113] and confirmed by others (table 2) [114].

The significantly better survival of patients with stage III ACC without distant disease as compared with those with distant metastases (stage IV) was shown in the series from the German Adrenocortical Cancer Registry described above (HR for death 0.44, 95% CI 0.25-0.78) [113].

Based on the eighth edition of the TNM staging system and by the ENSAT classification, the five-year, disease-specific survival rates in 416 adult cases reported to the German ACC registry were [113]:

Stage I – Confined to the adrenal gland without local invasion or distant metastases; greatest tumor dimension ≤5 cm (T1N0M0): 82 percent

Stage II – Same as stage I but with tumor size >5 cm without risk factors (T2N0M0): 61 percent

Stage III – Tumor of any size with at least one of the following factors: tumor infiltration in surrounding tissues (T3), tumor invasion into tumor thrombus in the vena cava or renal vein (T4), positive lymph nodes (N1) but no distant metastases: 50 percent

Stage IV – Distant metastases: 13 percent

Most contemporary treatment studies use the modification for stage groupings proposed by ENSAT over the 2004 UICC/AJCC classification (table 2).

A modification has been proposed for the ENSAT staging system that incorporates histologic grade of differentiation (table 3). (See "Treatment of adrenocortical carcinoma", section on 'Stage and margin status'.)

Children — The staging systems described above were developed primarily for adults [81,115,116]. A slightly different system has been used for children, which identifies only three prognostically distinct groups [46,51]:

Completely resected small tumors (200 g or smaller) – Excellent prognosis (five-year, event-free survival 91 percent in one series of 228 children [51])

Completely resected large tumors (over 200 g) – Intermediate prognosis (five-year, event-free survival 52 percent in the same series [51])

Residual or distant metastatic disease – Poor prognosis

Although ACC has historically been considered to have a poor prognosis, newer data suggest that survival may be improving. (See "Treatment of adrenocortical carcinoma", section on 'Prognosis'.)

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Adrenal cancer" and "Society guideline links: Adrenal incidentaloma".)

INFORMATION FOR PATIENTS — UpToDate offers two types of patient education materials, "The Basics" and "Beyond the Basics." The Basics patient education pieces are written in plain language, at the 5th to 6th grade reading level, and they answer the four or five key questions a patient might have about a given condition. These articles are best for patients who want a general overview and who prefer short, easy-to-read materials. Beyond the Basics patient education pieces are longer, more sophisticated, and more detailed. These articles are written at the 10th to 12th grade reading level and are best for patients who want in-depth information and are comfortable with some medical jargon.

Here are the patient education articles that are relevant to this topic. We encourage you to print or e-mail these topics to your patients. (You can also locate patient education articles on a variety of subjects by searching on "patient info" and the keyword(s) of interest.)

Basics topic (see "Patient education: Adrenal cancer (The Basics)")

SUMMARY — Unilateral tumors or masses of the adrenal gland are common. They are categorized as either functional (hormone-secreting) or silent, and as either benign or malignant.

The majority of adrenocortical tumors are benign, nonfunctioning adenomas that are discovered incidentally on abdominal imaging studies (adrenal incidentalomas). (See "Evaluation and management of the adrenal incidentaloma".)

Others are benign, hormone-secreting adenomas that cause Cushing's syndrome, primary aldosteronism or, much less commonly, virilization or feminization. Pheochromocytomas are adrenomedullary, not adrenocortical tumors. (See 'Adrenocortical adenomas' above.)

Adrenocortical carcinomas (ACCs) are rare, frequently aggressive tumors, which may be functional and cause Cushing's syndrome and/or virilization, or nonfunctional and present as an abdominal mass or an incidental finding. (See 'Adrenocortical carcinoma' above.)

Most adrenal adenomas are less than 4 cm in diameter. In contrast, most ACCs are greater than 4 cm in diameter when discovered. The lipid-rich nature of cortical adenomas is helpful in distinguishing this benign tumor from carcinoma on computed tomography (CT) scanning. (See 'Imaging' above.)

At initial presentation, approximately 50 percent of adult patients with ACC have relatively advanced disease stage. (See 'Staging' above.)

DISCLOSURE — The views expressed in this topic are those of the author(s) and do not reflect the official views or policy of the United States Government or its components.

  1. Fassnacht M, Arlt W, Bancos I, et al. Management of adrenal incidentalomas: European Society of Endocrinology Clinical Practice Guideline in collaboration with the European Network for the Study of Adrenal Tumors. Eur J Endocrinol 2016; 175:G1.
  2. Kebebew E. Adrenal Incidentaloma. N Engl J Med 2021; 384:1542.
  3. Mantero F, Terzolo M, Arnaldi G, et al. A survey on adrenal incidentaloma in Italy. Study Group on Adrenal Tumors of the Italian Society of Endocrinology. J Clin Endocrinol Metab 2000; 85:637.
  4. Reznik Y, Lefebvre H, Rohmer V, et al. Aberrant adrenal sensitivity to multiple ligands in unilateral incidentaloma with subclinical autonomous cortisol hypersecretion: a prospective clinical study. Clin Endocrinol (Oxf) 2004; 61:311.
  5. Lampron A, Bourdeau I, Oble S, et al. Regulation of aldosterone secretion by several aberrant receptors including for glucose-dependent insulinotropic peptide in a patient with an aldosteronoma. J Clin Endocrinol Metab 2009; 94:750.
  6. Zwermann O, Suttmann Y, Bidlingmaier M, et al. Screening for membrane hormone receptor expression in primary aldosteronism. Eur J Endocrinol 2009; 160:443.
  7. Ye P, Mariniello B, Mantero F, et al. G-protein-coupled receptors in aldosterone-producing adenomas: a potential cause of hyperaldosteronism. J Endocrinol 2007; 195:39.
  8. Albiger NM, Sartorato P, Mariniello B, et al. A case of primary aldosteronism in pregnancy: do LH and GNRH receptors have a potential role in regulating aldosterone secretion? Eur J Endocrinol 2011; 164:405.
  9. Mazzuco TL, Durand J, Chapman A, et al. Genetic aspects of adrenocortical tumours and hyperplasias. Clin Endocrinol (Oxf) 2012; 77:1.
  10. Bonnet-Serrano F, Bertherat J. Genetics of tumors of the adrenal cortex. Endocr Relat Cancer 2018; 25:R131.
  11. Smith TG, Clark SK, Katz DE, et al. Adrenal masses are associated with familial adenomatous polyposis. Dis Colon Rectum 2000; 43:1739.
  12. Kim AC, Reuter AL, Zubair M, et al. Targeted disruption of beta-catenin in Sf1-expressing cells impairs development and maintenance of the adrenal cortex. Development 2008; 135:2593.
  13. Polakis P. Wnt signaling and cancer. Genes Dev 2000; 14:1837.
  14. Kikuchi A. Tumor formation by genetic mutations in the components of the Wnt signaling pathway. Cancer Sci 2003; 94:225.
  15. Tissier F, Cavard C, Groussin L, et al. Mutations of beta-catenin in adrenocortical tumors: activation of the Wnt signaling pathway is a frequent event in both benign and malignant adrenocortical tumors. Cancer Res 2005; 65:7622.
  16. Tadjine M, Lampron A, Ouadi L, Bourdeau I. Frequent mutations of beta-catenin gene in sporadic secreting adrenocortical adenomas. Clin Endocrinol (Oxf) 2008; 68:264.
  17. Bonnet S, Gaujoux S, Launay P, et al. Wnt/β-catenin pathway activation in adrenocortical adenomas is frequently due to somatic CTNNB1-activating mutations, which are associated with larger and nonsecreting tumors: a study in cortisol-secreting and -nonsecreting tumors. J Clin Endocrinol Metab 2011; 96:E419.
  18. Goh G, Scholl UI, Healy JM, et al. Recurrent activating mutation in PRKACA in cortisol-producing adrenal tumors. Nat Genet 2014; 46:613.
  19. Cao Y, He M, Gao Z, et al. Activating hotspot L205R mutation in PRKACA and adrenal Cushing's syndrome. Science 2014; 344:913.
  20. Thiel A, Reis AC, Haase M, et al. PRKACA mutations in cortisol-producing adenomas and adrenal hyperplasia: a single-center study of 60 cases. Eur J Endocrinol 2015; 172:677.
  21. Ronchi CL, Di Dalmazi G, Faillot S, et al. Genetic Landscape of Sporadic Unilateral Adrenocortical Adenomas Without PRKACA p.Leu206Arg Mutation. J Clin Endocrinol Metab 2016; 101:3526.
  22. Beuschlein F, Fassnacht M, Assié G, et al. Constitutive activation of PKA catalytic subunit in adrenal Cushing's syndrome. N Engl J Med 2014; 370:1019.
  23. Sato Y, Maekawa S, Ishii R, et al. Recurrent somatic mutations underlie corticotropin-independent Cushing's syndrome. Science 2014; 344:917.
  24. Di Dalmazi G, Kisker C, Calebiro D, et al. Novel somatic mutations in the catalytic subunit of the protein kinase A as a cause of adrenal Cushing's syndrome: a European multicentric study. J Clin Endocrinol Metab 2014; 99:E2093.
  25. Nishimoto K, Nakagawa K, Li D, et al. Adrenocortical zonation in humans under normal and pathological conditions. J Clin Endocrinol Metab 2010; 95:2296.
  26. Boulkroun S, Samson-Couterie B, Dzib JF, et al. Adrenal cortex remodeling and functional zona glomerulosa hyperplasia in primary aldosteronism. Hypertension 2010; 56:885.
  27. Zennaro MC, Boulkroun S, Fernandes-Rosa F. Genetic Causes of Functional Adrenocortical Adenomas. Endocr Rev 2017; 38:516.
  28. Katayama M, Nomura K, Ujihara M, et al. Age-dependent decline in cortisol levels and clinical manifestations in patients with ACTH-independent Cushing's syndrome. Clin Endocrinol (Oxf) 1998; 49:311.
  29. Terzolo M, Pia A, Alì A, et al. Adrenal incidentaloma: a new cause of the metabolic syndrome? J Clin Endocrinol Metab 2002; 87:998.
  30. Di Dalmazi G, Vicennati V, Garelli S, et al. Cardiovascular events and mortality in patients with adrenal incidentalomas that are either non-secreting or associated with intermediate phenotype or subclinical Cushing's syndrome: a 15-year retrospective study. Lancet Diabetes Endocrinol 2014; 2:396.
  31. Debono M, Bradburn M, Bull M, et al. Cortisol as a marker for increased mortality in patients with incidental adrenocortical adenomas. J Clin Endocrinol Metab 2014; 99:4462.
  32. Piaditis GP, Kaltsas GA, Androulakis II, et al. High prevalence of autonomous cortisol and aldosterone secretion from adrenal adenomas. Clin Endocrinol (Oxf) 2009; 71:772.
  33. Vaidya A, Mulatero P, Baudrand R, Adler GK. The Expanding Spectrum of Primary Aldosteronism: Implications for Diagnosis, Pathogenesis, and Treatment. Endocr Rev 2018; 39:1057.
  34. Funder JW, Carey RM, Fardella C, et al. Case detection, diagnosis, and treatment of patients with primary aldosteronism: an endocrine society clinical practice guideline. J Clin Endocrinol Metab 2008; 93:3266.
  35. Moreno S, Montoya G, Armstrong J, et al. Profile and outcome of pure androgen-secreting adrenal tumors in women: experience of 21 cases. Surgery 2004; 136:1192.
  36. Kamenicky P, Houdoin L, Ferlicot S, et al. Benign cortisol-secreting adrenocortical adenomas produce small amounts of androgens. Clin Endocrinol (Oxf) 2007; 66:778.
  37. Fassnacht M, Libé R, Kroiss M, Allolio B. Adrenocortical carcinoma: a clinician's update. Nat Rev Endocrinol 2011; 7:323.
  38. Moreno S, Guillermo M, Decoulx M, et al. Feminizing adreno-cortical carcinomas in male adults. A dire prognosis. Three cases in a series of 801 adrenalectomies and review of the literature. Ann Endocrinol (Paris) 2006; 67:32.
  39. Szolar DH, Korobkin M, Reittner P, et al. Adrenocortical carcinomas and adrenal pheochromocytomas: mass and enhancement loss evaluation at delayed contrast-enhanced CT. Radiology 2005; 234:479.
  40. Groussin L, Bonardel G, Silvéra S, et al. 18F-Fluorodeoxyglucose positron emission tomography for the diagnosis of adrenocortical tumors: a prospective study in 77 operated patients. J Clin Endocrinol Metab 2009; 94:1713.
  41. Hahner S, Stuermer A, Kreissl M, et al. [123 I]Iodometomidate for molecular imaging of adrenocortical cytochrome P450 family 11B enzymes. J Clin Endocrinol Metab 2008; 93:2358.
  42. Khan TS, Sundin A, Juhlin C, et al. 11C-metomidate PET imaging of adrenocortical cancer. Eur J Nucl Med Mol Imaging 2003; 30:403.
  43. Ng L, Libertino JM. Adrenocortical carcinoma: diagnosis, evaluation and treatment. J Urol 2003; 169:5.
  44. Hsing AW, Nam JM, Co Chien HT, et al. Risk factors for adrenal cancer: an exploratory study. Int J Cancer 1996; 65:432.
  45. Allolio B, Fassnacht M. Clinical review: Adrenocortical carcinoma: clinical update. J Clin Endocrinol Metab 2006; 91:2027.
  46. Sandrini R, Ribeiro RC, DeLacerda L. Childhood adrenocortical tumors. J Clin Endocrinol Metab 1997; 82:2027.
  47. Figueiredo BC, Stratakis CA, Sandrini R, et al. Comparative genomic hybridization analysis of adrenocortical tumors of childhood. J Clin Endocrinol Metab 1999; 84:1116.
  48. Xiao XR, Ye LY, Shi LX, et al. Diagnosis and treatment of adrenal tumours: a review of 35 years' experience. Br J Urol 1998; 82:199.
  49. Luton JP, Cerdas S, Billaud L, et al. Clinical features of adrenocortical carcinoma, prognostic factors, and the effect of mitotane therapy. N Engl J Med 1990; 322:1195.
  50. Wooten MD, King DK. Adrenal cortical carcinoma. Epidemiology and treatment with mitotane and a review of the literature. Cancer 1993; 72:3145.
  51. Michalkiewicz E, Sandrini R, Figueiredo B, et al. Clinical and outcome characteristics of children with adrenocortical tumors: a report from the International Pediatric Adrenocortical Tumor Registry. J Clin Oncol 2004; 22:838.
  52. Sirianni R, Zolea F, Chimento A, et al. Targeting estrogen receptor-α reduces adrenocortical cancer (ACC) cell growth in vitro and in vivo: potential therapeutic role of selective estrogen receptor modulators (SERMs) for ACC treatment. J Clin Endocrinol Metab 2012; 97:E2238.
  53. Koch CA, Pacak K, Chrousos GP. The molecular pathogenesis of hereditary and sporadic adrenocortical and adrenomedullary tumors. J Clin Endocrinol Metab 2002; 87:5367.
  54. Sidhu S, Sywak M, Robinson B, Delbridge L. Adrenocortical cancer: recent clinical and molecular advances. Curr Opin Oncol 2004; 16:13.
  55. Sidhu S, Marsh DJ, Theodosopoulos G, et al. Comparative genomic hybridization analysis of adrenocortical tumors. J Clin Endocrinol Metab 2002; 87:3467.
  56. Bernard MH, Sidhu S, Berger N, et al. A case report in favor of a multistep adrenocortical tumorigenesis. J Clin Endocrinol Metab 2003; 88:998.
  57. Libé R, Bertherat J. Molecular genetics of adrenocortical tumours, from familial to sporadic diseases. Eur J Endocrinol 2005; 153:477.
  58. Gicquel C, Bertagna X, Gaston V, et al. Molecular markers and long-term recurrences in a large cohort of patients with sporadic adrenocortical tumors. Cancer Res 2001; 61:6762.
  59. Bourcigaux N, Gaston V, Logié A, et al. High expression of cyclin E and G1 CDK and loss of function of p57KIP2 are involved in proliferation of malignant sporadic adrenocortical tumors. J Clin Endocrinol Metab 2000; 85:322.
  60. Stojadinovic A, Ghossein RA, Hoos A, et al. Adrenocortical carcinoma: clinical, morphologic, and molecular characterization. J Clin Oncol 2002; 20:941.
  61. Latronico AC, Pinto EM, Domenice S, et al. An inherited mutation outside the highly conserved DNA-binding domain of the p53 tumor suppressor protein in children and adults with sporadic adrenocortical tumors. J Clin Endocrinol Metab 2001; 86:4970.
  62. Wagner J, Portwine C, Rabin K, et al. High frequency of germline p53 mutations in childhood adrenocortical cancer. J Natl Cancer Inst 1994; 86:1707.
  63. Ribeiro RC, Sandrini F, Figueiredo B, et al. An inherited p53 mutation that contributes in a tissue-specific manner to pediatric adrenal cortical carcinoma. Proc Natl Acad Sci U S A 2001; 98:9330.
  64. Reincke M, Karl M, Travis WH, et al. p53 mutations in human adrenocortical neoplasms: immunohistochemical and molecular studies. J Clin Endocrinol Metab 1994; 78:790.
  65. Ohgaki H, Kleihues P, Heitz PU. p53 mutations in sporadic adrenocortical tumors. Int J Cancer 1993; 54:408.
  66. Barzon L, Chilosi M, Fallo F, et al. Molecular analysis of CDKN1C and TP53 in sporadic adrenal tumors. Eur J Endocrinol 2001; 145:207.
  67. Libè R, Groussin L, Tissier F, et al. Somatic TP53 mutations are relatively rare among adrenocortical cancers with the frequent 17p13 loss of heterozygosity. Clin Cancer Res 2007; 13:844.
  68. Pinto EM, Chen X, Easton J, et al. Genomic landscape of paediatric adrenocortical tumours. Nat Commun 2015; 6:6302.
  69. Wasserman JD, Novokmet A, Eichler-Jonsson C, et al. Prevalence and functional consequence of TP53 mutations in pediatric adrenocortical carcinoma: a children's oncology group study. J Clin Oncol 2015; 33:602.
  70. Figueiredo BC, Cavalli LR, Pianovski MA, et al. Amplification of the steroidogenic factor 1 gene in childhood adrenocortical tumors. J Clin Endocrinol Metab 2005; 90:615.
  71. Zhang J, Walsh MF, Wu G, et al. Germline Mutations in Predisposition Genes in Pediatric Cancer. N Engl J Med 2015; 373:2336.
  72. Sullivan M, Boileau M, Hodges CV. Adrenal cortical carcinoma. J Urol 1978; 120:660.
  73. Gicquel C, Raffin-Sanson ML, Gaston V, et al. Structural and functional abnormalities at 11p15 are associated with the malignant phenotype in sporadic adrenocortical tumors: study on a series of 82 tumors. J Clin Endocrinol Metab 1997; 82:2559.
  74. Gicquel C, Bertagna X, Schneid H, et al. Rearrangements at the 11p15 locus and overexpression of insulin-like growth factor-II gene in sporadic adrenocortical tumors. J Clin Endocrinol Metab 1994; 78:1444.
  75. Gaujoux S, Grabar S, Fassnacht M, et al. β-catenin activation is associated with specific clinical and pathologic characteristics and a poor outcome in adrenocortical carcinoma. Clin Cancer Res 2011; 17:328.
  76. Assié G, Letouzé E, Fassnacht M, et al. Integrated genomic characterization of adrenocortical carcinoma. Nat Genet 2014; 46:607.
  77. Zheng S, Cherniack AD, Dewal N, et al. Comprehensive Pan-Genomic Characterization of Adrenocortical Carcinoma. Cancer Cell 2016; 29:723.
  78. Creemers SG, Korpershoek E, Atmodimedjo PN, et al. Identification of Mutations in Cell-Free Circulating Tumor DNA in Adrenocortical Carcinoma: A Case Series. J Clin Endocrinol Metab 2017; 102:3611.
  79. Crucitti F, Bellantone R, Ferrante A, et al. The Italian Registry for Adrenal Cortical Carcinoma: analysis of a multiinstitutional series of 129 patients. The ACC Italian Registry Study Group. Surgery 1996; 119:161.
  80. Vassilopoulou-Sellin R, Schultz PN. Adrenocortical carcinoma. Clinical outcome at the end of the 20th century. Cancer 2001; 92:1113.
  81. Icard P, Goudet P, Charpenay C, et al. Adrenocortical carcinomas: surgical trends and results of a 253-patient series from the French Association of Endocrine Surgeons study group. World J Surg 2001; 25:891.
  82. Wajchenberg BL, Albergaria Pereira MA, Medonca BB, et al. Adrenocortical carcinoma: clinical and laboratory observations. Cancer 2000; 88:711.
  83. Schteingart DE, Giordano TJ, Benitez RS, et al. Overexpression of CXC chemokines by an adrenocortical carcinoma: a novel clinical syndrome. J Clin Endocrinol Metab 2001; 86:3968.
  84. Hogan TF, Gilchrist KW, Westring DW, Citrin DL. A clinical and pathological study of adrenocortical carcinoma: therapeutic implications. Cancer 1980; 45:2880.
  85. Hough AJ, Hollifield JW, Page DL, Hartmann WH. Prognostic factors in adrenal cortical tumors. A mathematical analysis of clinical and morphologic data. Am J Clin Pathol 1979; 72:390.
  86. Abiven G, Coste J, Groussin L, et al. Clinical and biological features in the prognosis of adrenocortical cancer: poor outcome of cortisol-secreting tumors in a series of 202 consecutive patients. J Clin Endocrinol Metab 2006; 91:2650.
  87. Stewart JN, Flageole H, Kavan P. A surgical approach to adrenocortical tumors in children: the mainstay of treatment. J Pediatr Surg 2004; 39:759.
  88. Fassnacht M, Allolio B. Clinical management of adrenocortical carcinoma. Best Pract Res Clin Endocrinol Metab 2009; 23:273.
  89. Arlt W, Biehl M, Taylor AE, et al. Urine steroid metabolomics as a biomarker tool for detecting malignancy in adrenal tumors. J Clin Endocrinol Metab 2011; 96:3775.
  90. Mackie GC, Shulkin BL, Ribeiro RC, et al. Use of [18F]fluorodeoxyglucose positron emission tomography in evaluating locally recurrent and metastatic adrenocortical carcinoma. J Clin Endocrinol Metab 2006; 91:2665.
  91. Maurea S, Klain M, Mainolfi C, et al. The diagnostic role of radionuclide imaging in evaluation of patients with nonhypersecreting adrenal masses. J Nucl Med 2001; 42:884.
  92. Becherer A, Vierhapper H, Pötzi C, et al. FDG-PET in adrenocortical carcinoma. Cancer Biother Radiopharm 2001; 16:289.
  93. Leboulleux S, Dromain C, Bonniaud G, et al. Diagnostic and prognostic value of 18-fluorodeoxyglucose positron emission tomography in adrenocortical carcinoma: a prospective comparison with computed tomography. J Clin Endocrinol Metab 2006; 91:920.
  94. Tenenbaum F, Groussin L, Foehrenbach H, et al. 18F-fluorodeoxyglucose positron emission tomography as a diagnostic tool for malignancy of adrenocortical tumours? Preliminary results in 13 consecutive patients. Eur J Endocrinol 2004; 150:789.
  95. Minn H, Salonen A, Friberg J, et al. Imaging of adrenal incidentalomas with PET using (11)C-metomidate and (18)F-FDG. J Nucl Med 2004; 45:972.
  96. Metser U, Miller E, Lerman H, et al. 18F-FDG PET/CT in the evaluation of adrenal masses. J Nucl Med 2006; 47:32.
  97. Caoili EM, Korobkin M, Brown RK, et al. Differentiating adrenal adenomas from nonadenomas using (18)F-FDG PET/CT: quantitative and qualitative evaluation. Acad Radiol 2007; 14:468.
  98. Blake MA, Slattery JM, Kalra MK, et al. Adrenal lesions: characterization with fused PET/CT image in patients with proved or suspected malignancy--initial experience. Radiology 2006; 238:970.
  99. Zettinig G, Mitterhauser M, Wadsak W, et al. Positron emission tomography imaging of adrenal masses: (18)F-fluorodeoxyglucose and the 11beta-hydroxylase tracer (11)C-metomidate. Eur J Nucl Med Mol Imaging 2004; 31:1224.
  100. Hennings J, Lindhe O, Bergström M, et al. [11C]metomidate positron emission tomography of adrenocortical tumors in correlation with histopathological findings. J Clin Endocrinol Metab 2006; 91:1410.
  101. Jhala NC, Jhala D, Eloubeidi MA, et al. Endoscopic ultrasound-guided fine-needle aspiration biopsy of the adrenal glands: analysis of 24 patients. Cancer 2004; 102:308.
  102. Kocijancic K, Kocijancic I, Guna F. Role of sonographically guided fine-needle aspiration biopsy of adrenal masses in patients with lung cancer. J Clin Ultrasound 2004; 32:12.
  103. Weiss LM, Medeiros LJ, Vickery AL Jr. Pathologic features of prognostic significance in adrenocortical carcinoma. Am J Surg Pathol 1989; 13:202.
  104. Medeiros LJ, Weiss LM. New developments in the pathologic diagnosis of adrenal cortical neoplasms. A review. Am J Clin Pathol 1992; 97:73.
  105. Aubert S, Wacrenier A, Leroy X, et al. Weiss system revisited: a clinicopathologic and immunohistochemical study of 49 adrenocortical tumors. Am J Surg Pathol 2002; 26:1612.
  106. Lau SK, Weiss LM. The Weiss system for evaluating adrenocortical neoplasms: 25 years later. Hum Pathol 2009; 40:757.
  107. Tissier, F. Pathological pattern of adrenal cortical carcinoma. In: Adrenal Cancer, Bertagna, X. (Eds), John Libbey Eurotext Ltd., 2006. p.25.
  108. MACFARLANE DA. Cancer of the adrenal cortex; the natural history, prognosis and treatment in a study of fifty-five cases. Ann R Coll Surg Engl 1958; 23:155.
  109. World Health Organization. Pathology and Genetics of Tumours of Endocrine Organs, DeLellis RA, Lloyd RV, Heitz PU, Eng C (Eds), IARC, Lyon, France 2004.
  110. Kebebew E, Reiff E, Duh QY, et al. Extent of disease at presentation and outcome for adrenocortical carcinoma: have we made progress? World J Surg 2006; 30:872.
  111. Lee JE, Berger DH, el-Naggar AK, et al. Surgical management, DNA content, and patient survival in adrenal cortical carcinoma. Surgery 1995; 118:1090.
  112. Phan AT, Grogan RH, Rohren E, Perrier ND. Adrenal Cortical Carcinoma. In: AJCC Cancer Staging Manual, 8th, Amin MB (Ed), Springer, New York 2017. p.911.
  113. Fassnacht M, Johanssen S, Quinkler M, et al. Limited prognostic value of the 2004 International Union Against Cancer staging classification for adrenocortical carcinoma: proposal for a Revised TNM Classification. Cancer 2009; 115:243.
  114. Lughezzani G, Sun M, Perrotte P, 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. Eur J Cancer 2010; 46:713.
  115. Cohn K, Gottesman L, Brennan M. Adrenocortical carcinoma. Surgery 1986; 100:1170.
  116. Henley DJ, van Heerden JA, Grant CS, et al. Adrenal cortical carcinoma--a continuing challenge. Surgery 1983; 94:926.
Topic 137 Version 29.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟