13
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      Overview of the 2022 WHO Classification of Adrenal Cortical Tumors

      review-article

      Read this article at

      ScienceOpenPublisherPMC
      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          The new WHO classification of adrenal cortical proliferations reflects translational advances in the fields of endocrine pathology, oncology and molecular biology. By adopting a question–answer framework, this review highlights advances in knowledge of histological features, ancillary studies, and associated genetic findings that increase the understanding of the adrenal cortex pathologies that are now reflected in the 2022 WHO classification. The pathological correlates of adrenal cortical proliferations include diffuse adrenal cortical hyperplasia, adrenal cortical nodular disease, adrenal cortical adenomas and adrenal cortical carcinomas. Understanding germline susceptibility and the clonal-neoplastic nature of individual adrenal cortical nodules in primary bilateral macronodular adrenal cortical disease, and recognition of the clonal-neoplastic nature of incidentally discovered non-functional subcentimeter benign adrenal cortical nodules has led to redefining the spectrum of adrenal cortical nodular disease. As a consequence, the most significant nomenclature change in the field of adrenal cortical pathology involves the refined classification of adrenal cortical nodular disease which now includes (a) sporadic nodular adrenocortical disease, (b) bilateral micronodular adrenal cortical disease, and (c) bilateral macronodular adrenal cortical disease (formerly known primary bilateral macronodular adrenal cortical hyperplasia). This group of clinicopathological entities are reflected in functional adrenal cortical pathologies. Aldosterone producing cortical lesions can be unifocal or multifocal, and may be bilateral with no imaging-detected nodule(s). Furthermore, not all grossly or radiologically identified adrenal cortical lesions may be the source of aldosterone excess. For this reason, the new WHO classification endorses the nomenclature of the HISTALDO classification which uses CYP11B2 immunohistochemistry to identify functional sites of aldosterone production to help predict the risk of bilateral disease in primary aldosteronism. Adrenal cortical carcinomas are subtyped based on their morphological features to include conventional, oncocytic, myxoid, and sarcomatoid subtypes. Although the classic histopathologic criteria for diagnosing adrenal cortical carcinomas have not changed, the 2022 WHO classification underscores the diagnostic and prognostic impact of angioinvasion (vascular invasion) in these tumors. Microscopic angioinvasion is defined as tumor cells invading through a vessel wall and forming a thrombus/fibrin-tumor complex or intravascular tumor cells admixed with platelet thrombus/fibrin. In addition to well-established Weiss and modified Weiss scoring systems, the new WHO classification also expands on the use of other multiparameter diagnostic algorithms (reticulin algorithm, Lin–Weiss–Bisceglia system, and Helsinki scoring system) to assist the workup of adrenal cortical neoplasms in adults. Accordingly, conventional carcinomas can be assessed using all multiparameter diagnostic schemes, whereas oncocytic neoplasms can be assessed using the Lin–Weiss–Bisceglia system, reticulin algorithm and Helsinki scoring system. Pediatric adrenal cortical neoplasms are assessed using the Wieneke system. Most adult adrenal cortical carcinomas show > 5 mitoses per 10 mm 2 and > 5% Ki67. The 2022 WHO classification places an emphasis on an accurate assessment of tumor proliferation rate using both the mitotic count (mitoses per 10 mm 2) and Ki67 labeling index which play an essential role in the dynamic risk stratification of affected patients. Low grade carcinomas have mitotic rate of ≤ 20 mitoses per 10 mm 2, whereas high-grade carcinomas show > 20 mitoses per 10 mm 2. Ki67-based tumor grading has not been endorsed in the new WHO classification, since the proliferation indices are continuous variables rather than being static thresholds in tumor biology. This new WHO classification emphasizes the role of diagnostic and predictive biomarkers in the workup of adrenal cortical neoplasms. Confirmation of the adrenal cortical origin of a tumor remains a critical requirement when dealing with non-functional lesions in the adrenal gland which may be mistaken for a primary adrenal cortical neoplasm. While SF1 is the most reliable biomarker in the confirmation of adrenal cortical origin, paranuclear IGF2 expression is a useful biomarker in the distinction of malignancy in adrenal cortical neoplasms. In addition to adrenal myelolipoma, the new classification of adrenal cortical tumors has introduced new sections including adrenal ectopia, based on the potential role of such ectopic tissue as a possible source of neoplastic proliferations as well as a potential mimicker of metastatic disease. Adrenal cysts are also discussed in the new classification as they may simulate primary cystic adrenal neoplasms or even adrenal cortical carcinomas in the setting of an adrenal pseudocyst.

          Related collections

          Most cited references236

          • Record: found
          • Abstract: found
          • Article: not found

          A prospective study of the prevalence of primary aldosteronism in 1,125 hypertensive patients.

          We prospectively investigated the prevalence of curable forms of primary aldosteronism (PA) in newly diagnosed hypertensive patients. The prevalence of curable forms of PA is currently unknown, although retrospective data suggest that it is not as low as commonly perceived. Consecutive hypertensive patients referred to 14 hypertension centers underwent a diagnostic protocol composed of measurement of Na+ and K+ in serum and 24-h urine, sitting plasma renin activity, and aldosterone at baseline and after 50 mg captopril. The patients with an aldosterone/renin ratio >40 at baseline, and/or >30 after captopril, and/or a probability of PA (by a logistic discriminant function) > or =50% underwent imaging tests and adrenal vein sampling (AVS) or adrenocortical scintigraphy to identify the underlying adrenal pathology. An aldosterone-producing adenoma (APA) was diagnosed in patients who in addition to excess autonomous aldosterone secretion showed: 1) lateralized aldosterone secretion at AVS or adrenocortical scintigraphy, 2) adenoma at surgery and pathology, and 3) a blood pressure decrease after adrenalectomy. Evidence of excess autonomous aldosterone secretion without such criteria led to a diagnosis of idiopathic hyperaldosteronism (IHA). A total of 1,180 patients (age 46 +/- 12 years) were enrolled; a conclusive diagnosis was attained in 1,125 (95.3%). Of these, 54 (4.8%) had an APA and 72 (6.4%) had an IHA. There were more APA (62.5%) and fewer IHA cases (37.5%) at centers where AVS was available (p = 0.002); the opposite occurred where AVS was unavailable. In newly diagnosed hypertensive patients referred to hypertension centers, the prevalence of APA is high (4.8%). The availability of AVS is essential for an accurate identification of the adrenocortical pathologies underlying PA.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Integrated genomic characterization of adrenocortical carcinoma.

            Adrenocortical carcinomas (ACCs) are aggressive cancers originating in the cortex of the adrenal gland. Despite overall poor prognosis, ACC outcome is heterogeneous. We performed exome sequencing and SNP array analysis of 45 ACCs and identified recurrent alterations in known driver genes (CTNNB1, TP53, CDKN2A, RB1 and MEN1) and in genes not previously reported in ACC (ZNRF3, DAXX, TERT and MED12), which we validated in an independent cohort of 77 ACCs. ZNRF3, encoding a cell surface E3 ubiquitin ligase, was the most frequently altered gene (21%) and is a potential new tumor suppressor gene related to the β-catenin pathway. Our integrated genomic analyses further identified two distinct molecular subgroups with opposite outcome. The C1A group of ACCs with poor outcome displayed numerous mutations and DNA methylation alterations, whereas the C1B group of ACCs with good prognosis displayed specific deregulation of two microRNA clusters. Thus, aggressive and indolent ACCs correspond to two distinct molecular entities driven by different oncogenic alterations.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              K+ channel mutations in adrenal aldosterone-producing adenomas and hereditary hypertension.

              Endocrine tumors such as aldosterone-producing adrenal adenomas (APAs), a cause of severe hypertension, feature constitutive hormone production and unrestrained cell proliferation; the mechanisms linking these events are unknown. We identify two recurrent somatic mutations in and near the selectivity filter of the potassium (K(+)) channel KCNJ5 that are present in 8 of 22 human APAs studied. Both produce increased sodium (Na(+)) conductance and cell depolarization, which in adrenal glomerulosa cells produces calcium (Ca(2+)) entry, the signal for aldosterone production and cell proliferation. Similarly, we identify an inherited KCNJ5 mutation that produces increased Na(+) conductance in a Mendelian form of severe aldosteronism and massive bilateral adrenal hyperplasia. These findings explain pathogenesis in a subset of patients with severe hypertension and implicate loss of K(+) channel selectivity in constitutive cell proliferation and hormone production.
                Bookmark

                Author and article information

                Contributors
                ozgur.mete2@uhn.ca
                Journal
                Endocr Pathol
                Endocr Pathol
                Endocrine Pathology
                Springer US (New York )
                1046-3976
                1559-0097
                14 March 2022
                : 1-42
                Affiliations
                [1 ]GRID grid.231844.8, ISNI 0000 0004 0474 0428, Department of Pathology, , University Health Network, ; Toronto, ON Canada
                [2 ]Endocrine Oncology Site, Princess Margaret Cancer Centre, Toronto, ON Canada
                [3 ]GRID grid.17063.33, ISNI 0000 0001 2157 2938, Department of Laboratory Medicine and Pathobiology, , University of Toronto, ; Toronto, ON Canada
                [4 ]GRID grid.66875.3a, ISNI 0000 0004 0459 167X, Department of Laboratory Medicine and Pathology, , Mayo Clinic, ; Rochester, MN USA
                [5 ]GRID grid.4714.6, ISNI 0000 0004 1937 0626, Department of Oncology-Pathology, , Karolinska Institutet, ; Stockholm, Sweden
                [6 ]GRID grid.24381.3c, ISNI 0000 0000 9241 5705, Department of Pathology and Cancer Diagnostics, , Karolinska University Hospital, ; Stockholm, Sweden
                [7 ]GRID grid.7692.a, ISNI 0000000090126352, Princess Maxima Center for Pediatric Oncology, and Department of Pathology, , University Medical Center Utrecht, ; Utrecht, The Netherlands
                [8 ]GRID grid.69566.3a, ISNI 0000 0001 2248 6943, Department of Pathology, , Tohoku University School of Medicine, ; Sendai, Japan
                [9 ]GRID grid.7605.4, ISNI 0000 0001 2336 6580, Department of Pathology, , University of Turin, ; Turin, Italy
                [10 ]GRID grid.7605.4, ISNI 0000 0001 2336 6580, Department of Oncology, , University of Turin, ; Turin, Italy
                Author information
                http://orcid.org/0000-0003-0469-2801
                http://orcid.org/0000-0002-3666-2546
                http://orcid.org/0000-0002-5945-9081
                http://orcid.org/0000-0001-6871-1296
                http://orcid.org/0000-0002-6600-8641
                http://orcid.org/0000-0003-0719-105X
                http://orcid.org/0000-0002-2713-1700
                Article
                9710
                10.1007/s12022-022-09710-8
                8920443
                35288842
                24b7fdd3-0b25-46fc-b047-6c80b7c0df4d
                © The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature 2022

                This article is made available via the PMC Open Access Subset for unrestricted research re-use and secondary analysis in any form or by any means with acknowledgement of the original source. These permissions are granted for the duration of the World Health Organization (WHO) declaration of COVID-19 as a global pandemic.

                History
                : 12 February 2022
                Categories
                Article

                Endocrinology & Diabetes
                adrenal cortical adenoma,adrenal cortical carcinoma,adrenal cortical hyperplasia,adrenal cortical nodular disease,primary bilateral macronodular adrenal cortical hyperplasia,igf2,primary aldosteronism,cushing syndrome,virilization and feminization,adrenal incidentaloma,reticulin algorithm,reticulin histochemistry,lynch syndrome,who classification,biomarkers

                Comments

                Comment on this article