1
|
Das L, Dutta P. Association of primary hyperparathyroidism with pituitary adenoma and management issues. Best Pract Res Clin Endocrinol Metab 2025; 39:101978. [PMID: 39915142 DOI: 10.1016/j.beem.2025.101978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/17/2025]
Abstract
The co-occurrence of primary hyperparathyroidism (PHPT) and pituitary adenomas (PAs) is often indicative of underlying genetic syndromes such as Multiple Endocrine Neoplasia type 1 (MEN1) and, less commonly, MEN4. Although both conditions can occur sporadically, their simultaneous presence warrants evaluation for genetic mutations, with MEN1 mutations being the most frequent cause. The management of concurrent PHPT and PAs, especially in MEN1 patients, presents unique challenges. Management complexities arise from the syndromic nature, involving both surgical and medical interventions tailored to each condition. PHPT often manifests earlier and more aggressively in MEN1, requiring surgical intervention. However, recurrence rates remain high due to multiglandular involvement. Pituitary adenomas in MEN1 are primarily prolactinomas, and treatment with dopamine agonists results in significant tumour control in most cases. Overall, PAs associated with MEN1 are generally responsive to medical therapy, but careful long-term monitoring is essential. The utility of genetic screening cannot be overstated, as it aids in early detection, risk stratification, and management of both the index case and affected family members by cascade screening. A multidisciplinary approach is crucial for optimizing outcomes, with ongoing surveillance to manage recurrence and associated complications. In summary, the co-occurrence of PHPT and PAs, particularly in the context of MEN1, necessitates an integrated management strategy. Genetic testing is key in confirming diagnosis and guiding treatment, while surgical and medical interventions should be tailored to the extent and nature of glandular involvement. Close monitoring for recurrence and proactive family screening are essential components of long-term care.
Collapse
Affiliation(s)
- Liza Das
- Department of Telemedicine, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India.
| | - Pinaki Dutta
- Department of Endocrinology, PGIMER, Chandigarh, India.
| |
Collapse
|
2
|
Lanzaro F, De Biasio D, Cesaro FG, Stampone E, Tartaglione I, Casale M, Bencivenga D, Marzuillo P, Roberti D. Childhood Multiple Endocrine Neoplasia (MEN) Syndromes: Genetics, Clinical Heterogeneity and Modifying Genes. J Clin Med 2024; 13:5510. [PMID: 39336996 PMCID: PMC11432259 DOI: 10.3390/jcm13185510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 08/15/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
Multiple endocrine neoplasia (MEN) syndromes are part of a spectrum of clinically well-defined tumor syndromes ultimately characterized by histologically similar tumors arising in patients and families with mutations in one of the following four genes: MEN1, RET, CDKN1B, and MAX. The high level of genetic and phenotypic heterogeneity has been linked to phenocopies and modifying genes, as well as unknown mechanisms that might be investigated in the future based on preclinical and translational considerations. MEN1, also known as Wermer's syndrome (OMIM *131100), is an autosomal dominant syndrome codifying for the most frequent MEN syndrome showing high penetrance due to mutations in the MEN1 gene; nevertheless, clinical manifestations vary among patients in terms of tumor localization, age of onset, and clinical aggressiveness/severity, even within the same families. This has been linked to the effect of modifying genes, as described in the review. MEN 2-2b-4 and 5 also show remarkable clinical heterogeneity. The traditional view of genetically predisposing monogenic or multifactorial disorders is no longer valid, and mandates a change in scientific focus. Phenotypes are indeed rarely consistent across genetic backgrounds and environments. In the future, understanding factors and genetic variants that control cellular functions and the expression of disease genes should provide insights into fundamental disease processes, providing implications for counseling and therapeutic and prophylactic possibilities.
Collapse
Affiliation(s)
- Francesca Lanzaro
- Department of Woman, Child and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, Via Luigi De Crecchio 2, 80138 Naples, Italy
| | - Delia De Biasio
- Department of Woman, Child and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, Via Luigi De Crecchio 2, 80138 Naples, Italy
| | - Francesco Giustino Cesaro
- Department of Woman, Child and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, Via Luigi De Crecchio 2, 80138 Naples, Italy
| | - Emanuela Stampone
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via Luigi De Crecchio, 7, 80138 Naples, Italy
| | - Immacolata Tartaglione
- Department of Woman, Child and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, Via Luigi De Crecchio 2, 80138 Naples, Italy
| | - Maddalena Casale
- Department of Woman, Child and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, Via Luigi De Crecchio 2, 80138 Naples, Italy
| | - Debora Bencivenga
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via Luigi De Crecchio, 7, 80138 Naples, Italy
| | - Pierluigi Marzuillo
- Department of Woman, Child and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, Via Luigi De Crecchio 2, 80138 Naples, Italy
| | - Domenico Roberti
- Department of Woman, Child and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, Via Luigi De Crecchio 2, 80138 Naples, Italy
| |
Collapse
|
3
|
Balen AH, Tamblyn J, Skorupskaite K, Munro MG. A comprehensive review of the new FIGO classification of ovulatory disorders. Hum Reprod Update 2024; 30:355-382. [PMID: 38412452 DOI: 10.1093/humupd/dmae003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 01/23/2024] [Indexed: 02/29/2024] Open
Abstract
BACKGROUND The World Health Organization (WHO) system for the classification of disorders of ovulation was produced 50 years ago and, by international consensus, has been updated by the International Federation of Gynecology and Obstetrics (FIGO). OBJECTIVE AND RATIONALE This review outlines in detail each component of the FIGO HyPO-P (hypothalamic, pituitary, ovarian, PCOS) classification with a concise description of each cause, and thereby provides a systematic method for diagnosis and management. SEARCH METHODS We searched the published articles in the PubMed database in the English-language literature until October 2022, containing the keywords ovulatory disorders; ovulatory dysfunction; anovulation, and each subheading in the FIGO HyPO-P classification. We did not include abstracts or conference proceedings because the data are usually difficult to assess. OUTCOMES We present the most comprehensive review of all disorders of ovulation, published systematically according to the logical FIGO classification. WIDER IMPLICATIONS Improving the diagnosis of an individual's ovulatory dysfunction will significantly impact clinical practice by enabling healthcare practitioners to make a precise diagnosis and plan appropriate management.
Collapse
Affiliation(s)
- Adam H Balen
- Leeds Centre for Reproductive Medicine, The University of Leeds, Leeds, UK
| | - Jennifer Tamblyn
- Leeds Centre for Reproductive Medicine, The University of Leeds, Leeds, UK
| | | | - Malcolm G Munro
- Department of Obstetrics and Gynecology, The University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
4
|
Bräutigam K, Nesti C, Riss P, Scheuba C, Niederle B, Grob T, Di Domenico A, Neuenschwander M, Mazal P, Köhn N, Trepp R, Perren A, Kaderli RM. Syndromic MEN1 parathyroid adenomas consist of both subclonal nodules and clonally independent tumors. Virchows Arch 2024; 484:789-798. [PMID: 38244045 PMCID: PMC11106174 DOI: 10.1007/s00428-023-03730-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 12/11/2023] [Accepted: 12/26/2023] [Indexed: 01/22/2024]
Abstract
Primary hyperparathyroidism with parathyroid tumors is a typical manifestation of Multiple Endocrine Neoplasia Type 1 (MEN1) and is historically termed "primary hyperplasia". Whether these tumors represent a multi-glandular clonal disease or hyperplasia has not been robustly proven so far. Loss of Menin protein expression is associated with inactivation of both alleles and a good surrogate for a MEN1 gene mutation. The cyclin-dependent kinase inhibitor 1B (CDKN1B) gene is mutated in MEN4 and encodes for protein p27 whose expression is poorly studied in the syndromic MEN1 setting.Here, we analyzed histomorphology and protein expression of Menin and p27 in parathyroid adenomas of 25 patients of two independent, well-characterized MEN1 cohorts. The pattern of loss of heterozygosity (LOH) was assessed by fluorescence in situ hybridization (FISH) in one MEN1-associated parathyroid adenoma. Further, next-generation sequencing (NGS) was performed on eleven nodules of four MEN1 patients.Morphologically, the majority of MEN1 adenomas consisted of multiple distinct nodules, in which Menin expression was mostly lost and p27 protein expression reduced. FISH analysis revealed that most nodules exhibited MEN1 loss, with or without the loss of centromere 11. NGS demonstrated both subclonal evolution and the existence of clonally unrelated tumors.Syndromic MEN1 parathyroid adenomas therefore consist of multiple clones with subclones, which supports the current concept of the novel WHO classification of parathyroid tumors (2022). p27 expression was lost in a large fraction of MEN1 parathyroids and must therefore be used with caution in suggesting MEN4.
Collapse
Affiliation(s)
- Konstantin Bräutigam
- Institute of Tissue Medicine and Pathology, University of Bern, Murtenstr. 31, 3008, Bern, Switzerland.
| | - Cédric Nesti
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Philipp Riss
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Christian Scheuba
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Bruno Niederle
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Tobias Grob
- Institute of Tissue Medicine and Pathology, University of Bern, Murtenstr. 31, 3008, Bern, Switzerland
| | - Annunziata Di Domenico
- Institute of Tissue Medicine and Pathology, University of Bern, Murtenstr. 31, 3008, Bern, Switzerland
| | - Maja Neuenschwander
- Institute of Tissue Medicine and Pathology, University of Bern, Murtenstr. 31, 3008, Bern, Switzerland
| | - Peter Mazal
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Nastassja Köhn
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department of General Surgery, Cantonal Hospital of Aarau, Aarau, Switzerland
| | - Roman Trepp
- Department of Diabetes, Endocrinology, Nutritional Medicine and Metabolism, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Aurel Perren
- Institute of Tissue Medicine and Pathology, University of Bern, Murtenstr. 31, 3008, Bern, Switzerland
| | - Reto M Kaderli
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
5
|
Mazarico-Altisent I, Capel I, Baena N, Bella-Cueto MR, Barcons S, Guirao X, Albert L, Cano A, Pareja R, Caixàs A, Rigla M. Novel germline variants of CDKN1B and CDKN2C identified during screening for familial primary hyperparathyroidism. J Endocrinol Invest 2023; 46:829-840. [PMID: 36334246 PMCID: PMC10023768 DOI: 10.1007/s40618-022-01948-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 10/22/2022] [Indexed: 11/06/2022]
Abstract
PURPOSE CDKN1B mutations were established as a cause of multiple endocrine neoplasia 4 (MEN4) syndrome in patients with MEN1 phenotype without a mutation in the MEN1 gene. In addition, variants in other cyclin-dependent kinase inhibitors (CDKIs) were found in some MEN1-like cases without the MEN1 mutation. We aimed to describe novel germline mutations of these genes in patients with primary hyperparathyroidism (PHPT). METHODS During genetic screening for familial hyperparathyroidism, three novel CDKIs germline mutations in three unrelated cases between January 2019 and November 2021 were identified. In this report, we describe clinical features, DNA sequence analysis, and familial segregation studies based on these patients and their relatives. Genome-wide DNA study of loss of heterozygosity (LOH), copy number variation (CNV), and p27/kip immunohistochemistry was performed on tumour samples. RESULTS DNA screening was performed for atypical parathyroid adenomas in cases 1 and 2 and for cystic parathyroid adenoma and young age at diagnosis of PHPT in case 3. Genetic analysis identified likely pathogenic variants of CDKN1B in cases 1 and 2 and a variant of the uncertain significance of CDKN2C, with uniparental disomy in the tumour sample, in case 3. Neoplasm screening of probands showed other non-endocrine tumours in case 1 (colon adenoma with dysplasia and atypical lipomas) and case 2 (aberrant T-cell population) and a non-functional pituitary adenoma in case 3. CONCLUSION Germline mutations in CDKIs should be included in gene panels for genetic testing of primary hyperparathyroidism. New germline variants here described can be added to the current knowledge.
Collapse
Affiliation(s)
- I Mazarico-Altisent
- Endocrinology and Nutrition Department, Parc Taulí University Hospital, Institut d'Investigació i Innovació Parc Taulí (I3PT), Medicine Department, Universitat Autònoma de Barcelona, Parc Taulí 1, 08208, Sabadell, Barcelona, Spain.
| | - I Capel
- Endocrinology and Nutrition Department, Parc Taulí University Hospital, Institut d'Investigació i Innovació Parc Taulí (I3PT), Medicine Department, Universitat Autònoma de Barcelona, Parc Taulí 1, 08208, Sabadell, Barcelona, Spain
| | - N Baena
- Genetic Department, Parc Taulí University Hospital, Institut d'Investigació i Innovació Parc Taulí (I3PT), Medicine Department, Universitat Autònoma de Barcelona, Parc Taulí 1, 08208, Sabadell, Barcelona, Spain
| | - M R Bella-Cueto
- Pathology Department, Parc Taulí University Hospital, Institut d'Investigació i Innovació Parc Taulí (I3PT), Medicine Department, Universitat Autònoma de Barcelona, Parc Taulí 1, 08208, Sabadell, Barcelona, Spain
| | - S Barcons
- Surgery Department, Parc Taulí University Hospital, Institut d'Investigació i Innovació Parc Taulí (I3PT), Medicine Department, Universitat Autònoma de Barcelona, Parc Taulí 1, 08208, Sabadell, Barcelona, Spain
| | - X Guirao
- Surgery Department, Parc Taulí University Hospital, Institut d'Investigació i Innovació Parc Taulí (I3PT), Medicine Department, Universitat Autònoma de Barcelona, Parc Taulí 1, 08208, Sabadell, Barcelona, Spain
| | - L Albert
- Endocrinology and Nutrition Department, Parc Taulí University Hospital, Institut d'Investigació i Innovació Parc Taulí (I3PT), Medicine Department, Universitat Autònoma de Barcelona, Parc Taulí 1, 08208, Sabadell, Barcelona, Spain
| | - A Cano
- Endocrinology and Nutrition Department, Parc Taulí University Hospital, Institut d'Investigació i Innovació Parc Taulí (I3PT), Medicine Department, Universitat Autònoma de Barcelona, Parc Taulí 1, 08208, Sabadell, Barcelona, Spain
| | - R Pareja
- Endocrinology and Nutrition Department, Parc Taulí University Hospital, Institut d'Investigació i Innovació Parc Taulí (I3PT), Medicine Department, Universitat Autònoma de Barcelona, Parc Taulí 1, 08208, Sabadell, Barcelona, Spain
| | - A Caixàs
- Endocrinology and Nutrition Department, Parc Taulí University Hospital, Institut d'Investigació i Innovació Parc Taulí (I3PT), Medicine Department, Universitat Autònoma de Barcelona, Parc Taulí 1, 08208, Sabadell, Barcelona, Spain
| | - M Rigla
- Endocrinology and Nutrition Department, Parc Taulí University Hospital, Institut d'Investigació i Innovació Parc Taulí (I3PT), Medicine Department, Universitat Autònoma de Barcelona, Parc Taulí 1, 08208, Sabadell, Barcelona, Spain
| |
Collapse
|
6
|
Coopmans EC, Korbonits M. Molecular genetic testing in the management of pituitary disease. Clin Endocrinol (Oxf) 2022; 97:424-435. [PMID: 35349723 DOI: 10.1111/cen.14706] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 03/01/2022] [Accepted: 03/01/2022] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Most pituitary tumours occur sporadically without a genetically identifiable germline abnormality, a small but increasing proportion present with a genetic defect that predisposes to pituitary tumour development, either isolated (e.g., aryl hydrocarbon receptor-interacting protein, AIP) or as part of a tumour-predisposing syndrome (e.g., multiple endocrine neoplasia (MEN) type 1, Carney complex, McCune-Albright syndrome or pituitary tumour and paraganglioma association). Genetic alterations in sporadic pituitary adenomas may include somatic mutations (e.g., GNAS, USP8). In this review, we take a practical approach: which genetic syndromes should be considered in case of different presentation, such as tumour type, family history, age of onset and additional clinical features of the patient. DESIGN Review of the recent literature in the field of genetics of pituitary tumours. RESULTS Genetic testing in the management of pituitary disease is recommended in a significant minority of the cases. Understanding the genetic basis of the disease helps to identify patients and at-risk family members, facilitates early diagnosis and therefore better long-term outcome and opens up new pathways leading to tumorigenesis. CONCLUSION We provide a concise overview of the genetics of pituitary tumours and discuss the current challenges and implications of these genetic findings in clinical practice.
Collapse
Affiliation(s)
- Eva C Coopmans
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- Department of Medicine, Division of Endocrinology, Leiden University Medical Centre, Leiden, The Netherlands
- Department of Medicine, Endocrinology section, Pituitary Center Rotterdam, Erasmus University Medical Cente, Rotterdam, The Netherlands
| | - Márta Korbonits
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
7
|
Update in Pathogenesis, Diagnosis, and Therapy of Prolactinoma. Cancers (Basel) 2022; 14:cancers14153604. [PMID: 35892862 PMCID: PMC9331865 DOI: 10.3390/cancers14153604] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/20/2022] [Accepted: 07/22/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary This review updates recent advances in the pathogenesis, diagnosis, and therapy of prolactinoma. Prolactinomas, comprising 30–50% of all pituitary neuroendocrine tumors, frequently occur in females aged 20 to 50 and cause hypogonadism and infertility. In typical cases, female patients exhibit galactorrhea and amenorrhea due to serum prolactin (PRL) elevation, and during pregnancy, they should be carefully treated. During diagnosis, other causes of hyperprolactinemia must be excluded, and an MRI is useful for detecting pituitary neuroendocrine tumors. For the treatment of prolactinoma, dopamine agonists are effective in decreasing PRL levels and shrinking tumor size in most patients. Surgical treatment is recommended for patients who are resistant or intolerant to dopamine agonists. This review also discusses giant and malignant prolactinomas, prolactinoma-associated pregnancy, and new therapeutic approaches. Abstract Prolactinomas comprise 30–50% of all pituitary neuroendocrine tumors, frequently occur in females aged 20 to 50, and cause hypogonadism and infertility. In typical cases, female patients exhibit galactorrhea and amenorrhea due to serum prolactin (PRL) elevation, and patients during pregnancy should be carefully treated. During diagnosis, other causes of hyperprolactinemia must be excluded, and an MRI is useful for detecting pituitary neuroendocrine tumors. For treating prolactinoma, dopamine agonists (DAs) are effective for decreasing PRL levels and shrinking tumor size in most patients. Some DA-resistant cases and the molecular mechanisms of resistance to a DA are partially clarified. The side effects of a DA include cardiac valve alterations and impulse control disorders. Although surgical therapies are invasive, recent analysis shows that long-term remission rates are higher than from medical therapies. The treatments for giant or malignant prolactinomas are challenging, and the combination of medication, surgery, and radiation therapy should be considered. Regarding pathogenesis, somatic SF3B1 mutations were recently identified even though molecular mechanisms in most cases of prolactinoma have not been elucidated. To understand the pathogenesis of prolactinomas, the development of new therapeutic approaches for treatment-resistant patients is expected. This review updates the recent advances in understanding the pathogenesis, diagnosis, and therapy of prolactinoma.
Collapse
|
8
|
Erickson LA, Mete O, Juhlin CC, Perren A, Gill AJ. Overview of the 2022 WHO Classification of Parathyroid Tumors. Endocr Pathol 2022; 33:64-89. [PMID: 35175514 DOI: 10.1007/s12022-022-09709-1] [Citation(s) in RCA: 116] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/29/2022] [Indexed: 12/18/2022]
Abstract
The 2022 WHO classification reflects increases in the knowledge of the underlying pathogenesis of parathyroid disease. In addition to the classic characteristic features of parathyroid neoplasms, subtleties in histologic features which may indicate an underlying genetic abnormality reflect increased understanding of the clinical manifestations, histologic, and genetic correlation in parathyroid disease. The importance of underlying genetic aberrancies is emphasized due to their significance to the care of the patient. Traditionally, the term "parathyroid hyperplasia" has been applied to multiglandular parathyroid disease; however, the concept of hyperplasia is generally no longer supported in the context of primary hyperparathyroidism since affected glands are usually composed of multiple "clonal" neoplastic proliferations. In light of these findings and management implications for patient care, the 2022 WHO classification endorses primary hyperparathyroidism-related multiglandular parathyroid disease (multiglandular multiple parathyroid adenomas) as a germline susceptibility-driven multiglandular parathyroid neoplasia. From such a perspective, pathologists can provide additional value to genetic triaging by recognizing morphological and immunohistochemical harbingers of MEN1, CDKN1B, MAX, and CDC73-related manifestations. In the current WHO classification, the term "parathyroid hyperplasia" is now used primarily in the setting of secondary hyperplasia which is most often caused by chronic renal failure. In addition to expansion in the histological features, including those that may be suggestive of an underlying genetic abnormality, there are additional nomenclature changes in the 2022 WHO classification reflecting increased understanding of the underlying pathogenesis of parathyroid disease. The new classification no longer endorses the use of "atypical parathyroid adenoma". This entity is now being replaced with the term of "atypical parathyroid tumor" to reflect a parathyroid neoplasm of uncertain malignant potential. The differential diagnoses of atypical parathyroid tumor are discussed along with the details of worrisome clinical and laboratory findings, and also features that define atypical histological and immunohistochemical findings to qualify for this diagnosis. The histological definition of parathyroid carcinoma still requires one of the following findings: (i) angioinvasion (vascular invasion) characterized by tumor invading through a vessel wall and associated thrombus, or intravascular tumor cells admixed with thrombus, (ii) lymphatic invasion, (iii) perineural (intraneural) invasion, (iv) local malignant invasion into adjacent anatomic structures, or (v) histologically/cytologically documented metastatic disease. In parathyroid carcinomas, the documentation of mitotic activity (e.g., mitoses per 10mm2) and Ki67 labeling index is recommended. Furthermore, the importance of complete submission of parathyroidectomy specimens for microscopic examination, and the crucial role of multiple levels along with ancillary biomarkers have expanded the diagnostic workup of atypical parathyroid tumors and parathyroid carcinoma to ensure accurate characterization of parathyroid neoplasms. The concept of parafibromin deficiency has been expanded upon and term "parafibromin deficient parathyroid neoplasm" is applied to a parathyroid neoplasm showing complete absence of nuclear parafibromin immunoreactivity. Nucleolar loss is considered as abnormal finding that requires further molecular testing to confirm its biological significance. The 2022 WHO classification emphasizes the role of molecular immunohistochemistry in parathyroid disease. 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 underlying pathogenesis of parathyroid disease that are now reflected in the updated classification and new entities in the 2022 WHO classification.
Collapse
Affiliation(s)
- Lori A Erickson
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 First St SW, Rochester, MN, 55901, USA.
| | - Ozgur Mete
- Department of Pathology, University Health Network, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - C Christofer Juhlin
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Department of Pathology and Cancer Diagnostics, Karolinska University Hospital, Stockholm, Sweden
| | - Aurel Perren
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Anthony J Gill
- Department of Anatomical Pathology, NSW Health Pathology, Royal North Shore Hospital, St Leonards, Sydney, NSW, Australia
- Cancer Diagnosis and Pathology Research Group, Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, Sydney, NSW, Australia
- University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
9
|
Nosé V, Gill A, Teijeiro JMC, Perren A, Erickson L. Overview of the 2022 WHO Classification of Familial Endocrine Tumor Syndromes. Endocr Pathol 2022; 33:197-227. [PMID: 35285003 DOI: 10.1007/s12022-022-09705-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/22/2022] [Indexed: 12/16/2022]
Abstract
This review of the familial tumor syndromes involving the endocrine organs is focused on discussing the main updates on the upcoming fifth edition of the WHO Classification of Endocrine and Neuroendocrine Tumors. This review emphasizes updates on histopathological and molecular genetics aspects of the most important syndromes involving the endocrine organs. We describe the newly defined Familial Cancer Syndromes as MAFA-related, MEN4, and MEN5 as well as the newly reported pathological findings in DICER1 syndrome. We also describe the updates done at the new WHO on the syndromic and non-syndromic familial thyroid diseases. We emphasize the problem of diagnostic criteria, mention the new genes that are possibly involved in this group, and at the same time, touching upon the role of some immunohistochemical studies that could support the diagnosis of some of these conditions. As pathologists play an important role in identifying tumors within a familial cancer syndrome, we highlight the most important clues for raising the suspicious of a syndrome. Finally, we highlight the challenges in defining these entities as well as determining their clinical outcome in comparison with sporadic tumors. Instead of the usual subject review, we present the highlights of the updates on familial cancer syndromes by answering select questions relevant to practicing pathologists.
Collapse
Affiliation(s)
- Vania Nosé
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA.
| | | | - José Manuel Cameselle Teijeiro
- Clinical University Hospital Santiago de Compostela and Medical Faculty, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Aurel Perren
- Institute of Pathology, University of Bern, Bern, Switzerland
| | | |
Collapse
|
10
|
Tang Y, Xie T, Wu S, Yang Q, Liu T, Li C, Liu S, Shao Z, Zhang X. Quantitative proteomics revealed the molecular characteristics of distinct types of granulated somatotroph adenomas. Endocrine 2021; 74:375-386. [PMID: 34043183 DOI: 10.1007/s12020-021-02767-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 05/15/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE Somatotroph adenomas are obviously heterogeneous in clinical characteristics, imaging performance, pathological diagnosis and therapeutic effect. The heterogeneity of the tumors, especially for SG and DG type adenomas, have attracted great interest in identifying the specific pathological markers and therapeutic targets of them. However, previous analyses of the molecular characteristics of the subtypes of somatotroph adenomas were performed at genomic and transcriptome level. The proteomic differences between the two subtypes of somatotroph adenomas are still unknown. METHODS Tumor samples were surgically removed from 10 sporadic pituitary somatotroph adenoma patients and grouped according to the pathological type. Tandem mass tag (TMT)-based quantitative proteomic analysis was employed to analyze the proteomic differences between SG and DG tumors. RESULTS In total, 228 differentially expressed proteins were identified between SG adenomas and DG adenomas. They were enriched mainly in extracellular matrix (ECM)-receptor interaction, leukocyte transendothelial migration, arrhythmogenic right ventricular cardiomyopathy and DNA replication pathways. Protein-protein interaction (PPI) network analysis indicated that Cadherin-1 and Catenin beta-1 were the most important key proteins in the differences between SG and DG adenomas. Immunohistochemistry (IHC) confirmed the expression levels of the key proteins. CONCLUSIONS This study provides large-scale proteome molecular characteristics of distinct granulation subtypes of somatotroph adenomas. Compared with DG adenomas, The differential protein of SG adenomas mostly enrich in invasive and proliferative functions and pathways at the proteomic level. Cadherin-1 and Catenin beta-1 play key roles in the different biological characteristics of the two tumor subtypes.
Collapse
Affiliation(s)
- Yifan Tang
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tao Xie
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Silin Wu
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qiaoqiao Yang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Tengfei Liu
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chen Li
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shuang Liu
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhiyong Shao
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Xiaobiao Zhang
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai, China.
- Digital Medical Research Center, Fudan University, Shanghai, China.
- Shanghai Key Laboratory of Medical Image Computing and Computer-Assisted Intervention, Shanghai, China.
| |
Collapse
|
11
|
Böhm M, Gellner R. [Neuroendorine paraneoplastic syndromes]. Hautarzt 2021; 72:299-306. [PMID: 33661337 DOI: 10.1007/s00105-021-04778-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/03/2021] [Indexed: 10/22/2022]
Abstract
Skin is commonly affected by neuroendorine paraneoplastic syndromes (PNS). This is due to the expression of receptors in the skin by which abnormally secreted neuroendocrine hormones and mediators elicit directly, and indirectly, cutaneous key signs and thus facilitate early diagnosis of these diseases. In acromegaly, induction of the growth hormone-insulin-like growth factor‑1 axis results in trophic changes of the acral portions of the skin and mucosal membranes including cutis verticis gyrata. The skin signs of non-iatrogenic Cushing syndrome are identical with those of exogenous prolonged intake of glucocorticoids: centripetal accumulation of adipose tissue, plethora and striae distensae. Episodic flushing of the face and trunk (together with explosive diarrhea) is a key feature of carcinoid tumors. Fibrotic remodeling of the heart and retroperitoneal space, and less commonly of the skin, are important complications mediated by abnormally secreted 5‑hydroxytryptamine (serotonin, 5‑HT), the latter eliciting profibrotic responses on HT2B-receptor-expressing fibroblasts. Androgen-secreting tumors lead to well-established receptor-mediated cutaneous signs of peripheral hyperandrogenisms: seborrhea, acne, hirsutism, and androgenetic alopecia. In contrast, the pathogenesis of necrolytic migratory erythema as a key feature of glucagonoma remains incompletely understood and is thought to be related to hypoaminoacidemia. This review summarizes the clinical features of neuroendocrine PNS with skin involvement, elucidates its underlying pathophysiology, lists differential diagnoses, and explains key diagnostic steps and principal therapeutic options. An interdisciplinary approach is essential to provide the best care of all patients with neuroendocrine PNS.
Collapse
Affiliation(s)
- M Böhm
- Klinik für Hautkrankheiten, Universitätsklinikum Münster, Von-Esmarch-Str. 58, 48149, Münster, Deutschland.
| | - R Gellner
- Medizinische Klinik B, Universitätsklinikum Münster, Münster, Deutschland
| |
Collapse
|
12
|
Hinojosa-Amaya JM, Lam-Chung CE, Cuevas-Ramos D. Recent Understanding and Future Directions of Recurrent Corticotroph Tumors. Front Endocrinol (Lausanne) 2021; 12:657382. [PMID: 33986726 PMCID: PMC8111286 DOI: 10.3389/fendo.2021.657382] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 03/29/2021] [Indexed: 11/13/2022] Open
Abstract
Corticotroph tumors (CTs) are pituitary neoplasms arising from the Tpit lineage, which may or not express adrenocorticotrophic hormone (ACTH). Functioning CTs cause Cushing's disease (CD), which has high morbidity and mortality due to hypercortisolemia. "Non-functioning" or silent CTs (SCT) and the Crooke's cell subtypes do not cause CD and may be asymptomatic until manifested by compressive symptoms and are more frequently found as macroadenoma. Both tend toward more aggressive behavior, recurrence, and a higher rate of malignant transformation to pituitary carcinoma. Tumorigenesis involves genetic, epigenetic, and post-transcriptional disruption of cell-cycle regulators, which increase cell proliferation, POMC overexpression, ACTH transcription, and/or hypersecretion. Furthermore, functioning CTs develop resistance to glucocorticoid-mediated negative feedback on ACTH secretion, through increased expression of testicular orphan nuclear receptor 4 (TR4), heat-shock protein 90 (HSP90), and loss-of-function mutation of CDK5 and ABL enzyme substrate 1 (CABLES1) gene. Overt autonomous hypercortisolemia is difficult to control, and multiple diagnostic studies and therapeutic modalities are commonly required. Cell-cycle regulation depends mainly on p27, cyclin E, cyclin-dependent kinases (CDKs), and the retinoblastoma protein (Rb)/E2F1 transcription factor complex. Gain-of-function mutations of ubiquitin-specific protease (USP) 8, USP48, and BRAF genes may subsequently cause overexpression of epithelial growth factor receptor (EGFR), and enhance POMC transcription, cell proliferation, and tumor growth. Epigenetic changes through micro RNAs and decreased DNA deacetylation by histone deacetylase type 2 (HDAC2), may also affect tumor growth. All the former mechanisms may become interesting therapeutic targets for CTs, aside from temozolomide, currently used for aggressive tumors. Potential therapeutic agents are EGFR inhibitors such as gefitinib and lapatinib, the purine analog R-roscovitine by dissociation of CDK2/Cyclin E complex, the HSP90 inhibitor silibinin (novobiocin), to reduce resistance to glucocorticoid-mediated negative feedback, and BRAF inhibitors vemurafenib and dabrafenib in BRAF V600E positive tumors. This review summarizes the molecular mechanisms related to CTs tumorigenesis, their diagnostic approach, and provides an update of the potential novel therapies, from the lab bench to the clinical translation.
Collapse
Affiliation(s)
- José Miguel Hinojosa-Amaya
- Pituitary Clinic, Endocrinology Division, Department of Medicine, Hospital Universitario “Dr. José E. González” UANL, Monterrey, Mexico
| | - César Ernesto Lam-Chung
- Neuroendocrinology Clinic, Department of Endocrinology and Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Daniel Cuevas-Ramos
- Neuroendocrinology Clinic, Department of Endocrinology and Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- *Correspondence: Daniel Cuevas-Ramos,
| |
Collapse
|
13
|
Abstract
Pituitary adenomas are common intracranial neoplasms, with diverse phenotypes. Most of these tumors occur sporadically and are not part of genetic disorders. Over the last decades numerous genetic studies have led to identification of somatic and germline mutations associated with pituitary tumors, which has advanced the understanding of pituitary tumorigenesis. Exploring the genetic background of pituitary neuroendocrine tumors can lead to early diagnosis associated with better outcomes, and their molecular mechanisms should lead to novel targeted therapies even for sporadic tumors. This article summarizes the genes and the syndromes associated with pituitary tumors.
Collapse
Affiliation(s)
- Sayka Barry
- Centre for Endocrinology, William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Márta Korbonits
- Centre for Endocrinology, William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK.
| |
Collapse
|
14
|
Abstract
Parathyroid gland excision specimens are common and sometimes underestimated cases that many surgical pathologists encounter regularly. In the vast majority of cases, these will be spot diagnoses of sporadic primary parathyroid adenomas or, perhaps, hyperplasias commonly in the setting of renal failure. However, a small but significant number of parathyroid gland excisions may be due to heritable disease. In most cases, hereditary disease is suspected by the referring clinicians. Nevertheless, a subset of these are undetected which is significant, particularly in the setting of the multiple endocrine neoplasia (MEN), and the hyperparathyroidism jaw tumour (HPT-JT) syndromes. There have been recent advances in recognition of the morphological and immunohistochemical characteristics of these tumours and hyperplasias. While hereditary kindreds are over-represented at specialist referral centres, with awareness of the characteristic clinical and morphological features, the general surgical pathologist is frequently able to suggest the possibility of hereditary parathyroid disease. We therefore provide a succinct guide for pathologists to increase the recognition of hereditary parathyroid disease.
Collapse
MESH Headings
- Diagnosis, Differential
- Diagnostic Techniques, Endocrine/standards
- Genetic Diseases, Inborn/diagnosis
- Genetic Diseases, Inborn/metabolism
- Genetic Diseases, Inborn/pathology
- Humans
- Hyperparathyroidism, Primary/diagnosis
- Hyperparathyroidism, Primary/genetics
- Hyperparathyroidism, Primary/metabolism
- Hyperparathyroidism, Primary/pathology
- Immunohistochemistry
- Parathyroid Diseases/diagnosis
- Parathyroid Diseases/genetics
- Parathyroid Diseases/metabolism
- Parathyroid Diseases/pathology
- Parathyroid Glands/metabolism
- Parathyroid Glands/pathology
- Pathologists/standards
- Pathologists/statistics & numerical data
- Practice Patterns, Physicians'/standards
Collapse
Affiliation(s)
- John Turchini
- Anatomical Pathology, Douglass Hanly Moir Pathology, Macquarie Park, NSW, 2113, Australia.
- Discipline of Pathology, MQ Health, Macquarie University, Macquarie Park, NSW, 2113, Australia.
- Sydney Medical School, The University of Sydney, Sydney, 2006, Australia.
- Cancer Diagnosis and Pathology Group, Kolling Institute of Medical Research, St Leonards, NSW, 2065, Australia.
| | - Anthony J Gill
- Sydney Medical School, The University of Sydney, Sydney, 2006, Australia
- Cancer Diagnosis and Pathology Group, Kolling Institute of Medical Research, St Leonards, NSW, 2065, Australia
- Department of Anatomical Pathology, NSW Health Pathology, Royal North Shore Hospital, St Leonards, NSW, 2065, Australia
| |
Collapse
|
15
|
Abstract
Lactotroph adenomas, also called prolactinomas and prolactin-secreting adenomas, constitute nearly 80% of functioning pituitary tumors and about 30-50% of all adenomas in the clinical practice. Lactotroph adenomas occur in the general population at a prevalence of 45/100,000, are more common in women, but also involve men and children of both sexes. Most lactotroph adenomas are microadenomas occurring in reproductive-age women who present with oligo/amenorrhea, galactorrhea, and infertility. In men and elderly women, lactotroph adenomas are usually macroadenomas and are most commonly associated with symptoms of a tumoral mass, including headaches, neurologic defects, and visual loss. Although clinical and laboratory features may differ depending on patient's gender and age, the histopathology of the tumors is similar. Lactotroph adenomas are histologically classified into three subtypes: the common sparsely granulated lactotroph adenoma, and the rare densely granulated lactotroph adenoma and acidophilic stem cell adenoma. We will review the main pathological features of the lactotroph adenomas and some of their characteristics that may predict biological behavior and responsiveness to treatment.
Collapse
Affiliation(s)
- M Beatriz S Lopes
- Division of Neuropathology, Department of Pathology, University of Virginia School of Medicine, 1215 Lee Street, HEP-Room 3060, Charlottesville, VA, 22908-0214, USA.
| |
Collapse
|
16
|
Abstract
Hyperprolactinaemia is one of the most common problems in clinical endocrinology. It relates with various aetiologies (physiological, pharmacological, pathological), the clarification of which requires careful history taking and clinical assessment. Analytical issues (presence of macroprolactin or of the hook effect) need to be taken into account when interpreting the prolactin values. Medications and sellar/parasellar masses (prolactin secreting or acting through “stalk effect”) are the most common causes of pathological hyperprolactinaemia. Hypogonadism and galactorrhoea are well-recognized manifestations of prolactin excess, although its implications on bone health, metabolism and immune system are also expanding. Treatment mainly aims at restoration and maintenance of normal gonadal function/fertility, and prevention of osteoporosis; further specific management strategies depend on the underlying cause. In this review, we provide an update on the diagnostic and management approaches for the patient with hyperprolactinaemia and on the current data looking at the impact of high prolactin on metabolism, cardiovascular and immune systems.
Collapse
|
17
|
The Clinicopathological Spectrum of Acromegaly. J Clin Med 2019; 8:jcm8111962. [PMID: 31766255 PMCID: PMC6912315 DOI: 10.3390/jcm8111962] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 11/08/2019] [Accepted: 11/11/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Acromegaly results from a persistent excess in growth hormone with clinical features that may be subtle or severe. The most common cause of acromegaly is a pituitary tumor that causes excessive production of growth hormone (GH), and rare cases are due to an excess of the GH-releasing hormone (GHRH) or the ectopic production of GH. OBJECTIVE Discuss the different diseases that present with manifestations of GH excess and clinical acromegaly, emphasizing the distinct clinical and radiological characteristics of the different pathological entities. METHODS We performed a narrative review of the published clinicopathological information about acromegaly. An English-language search for relevant studies was conducted on PubMed from inception to 1 August 2019. The reference lists of relevant studies were also reviewed. RESULTS Pituitary tumors that cause GH excess have several variants, including pure somatotroph tumors that can be densely or sparsely granulated, or plurihormonal tumors that include mammosomatotroph, mixed somatotroph-lactotroph tumors and mature plurihomonal Pit1-lineage tumors, acidophil stem cell tumors and poorly-differentiated Pit1-lineage tumors. Each tumor type has a distinct pathophysiology, resulting in variations in clinical manifestations, imaging and responses to therapies. CONCLUSION Detailed clinicopathological information will be useful in the era of precision medicine, in which physicians tailor the correct treatment modality to each patient.
Collapse
|
18
|
Genetics of Pituitary Tumours. EXPERIENTIA. SUPPLEMENTUM 2019. [PMID: 31588533 DOI: 10.1007/978-3-030-25905-1_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2023]
Abstract
Pituitary tumours are relatively common in the general population. Most often they occur sporadically, with somatic mutations accounting for a significant minority of somatotroph and corticotroph adenomas. Pituitary tumours can also develop secondary to germline mutations as part of a complex syndrome or as familial isolated pituitary adenomas. Tumours occurring in a familial setting may present at a younger age and can behave more aggressively with resistance to treatment. This chapter will focus on the genetics and molecular pathogenesis of pituitary tumours.
Collapse
|
19
|
Fuentes-Fayos AC, García-Martínez A, Herrera-Martínez AD, Jiménez-Vacas JM, Vázquez-Borrego MC, Castaño JP, Picó A, Gahete MD, Luque RM. Molecular determinants of the response to medical treatment of growth hormone secreting pituitary neuroendocrine tumors. MINERVA ENDOCRINOL 2019; 44:109-128. [PMID: 30650942 DOI: 10.23736/s0391-1977.19.02970-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Acromegaly is a chronic systemic disease mainly caused by a growth hormone (GH)-secreting pituitary neuroendocrine tumor (PitNETs), which is associated with many health complications and increased mortality when not adequately treated. Transsphenoidal surgery is considered the treatment of choice in GH-secreting PitNETs, but patients in whom surgery cannot be considered or with persistent disease after surgery require medical therapy. Treatment with available synthetic somatostatin analogues (SSAs) is considered the mainstay in the medical management of acromegaly which exert their beneficial effects through the binding to a family of G-protein coupled receptors encoded by 5 genes (SSTR1-5). However, although it has been demonstrated that the SST1-5 receptors are physically present in tumor cells, SSAs are in many cases ineffective (i.e. approximately 10-30% of patients with GH-secreting PitNET are unresponsive to SSAs), suggesting that other cellular/molecular determinants could be essential for the response to the pharmacological treatment in patients with GH-secreting PitNETs. Therefore, the scrutiny of these determinants might be used for the identification of subgroups of patients in whom an appropriate pharmacological treatment can be successfully employed (responders vs. non-responders). In this review, we will describe some of the existing, classical and novel, genetic and molecular determinants involved in the response of patients with GH-secreting PitNETs to the available therapeutic treatments, as well as new molecular/therapeutic approaches that could be potentially useful for the treatment of GH-secreting PitNETs.
Collapse
Affiliation(s)
- Antonio C Fuentes-Fayos
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Cordoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain.,Reina Sofia University Hospital (HURS), Cordoba, Spain.,CIBER Physiopathology of Obesity and Nutrition (CIBERobn), Cordoba, Spain
| | - Araceli García-Martínez
- Research Laboratory, Hospital General Universitario de Alicante-Institute for Health and Biomedical Research (ISABIAL), Alicante, Spain
| | - Aura D Herrera-Martínez
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Cordoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain.,Reina Sofia University Hospital (HURS), Cordoba, Spain.,CIBER Physiopathology of Obesity and Nutrition (CIBERobn), Cordoba, Spain
| | - Juan M Jiménez-Vacas
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Cordoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain.,Reina Sofia University Hospital (HURS), Cordoba, Spain.,CIBER Physiopathology of Obesity and Nutrition (CIBERobn), Cordoba, Spain
| | - Mari C Vázquez-Borrego
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Cordoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain.,Reina Sofia University Hospital (HURS), Cordoba, Spain.,CIBER Physiopathology of Obesity and Nutrition (CIBERobn), Cordoba, Spain
| | - Justo P Castaño
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Cordoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain.,Reina Sofia University Hospital (HURS), Cordoba, Spain.,CIBER Physiopathology of Obesity and Nutrition (CIBERobn), Cordoba, Spain
| | - Antonio Picó
- Department of Endocrinology and Nutrition, Hospital General Universitario de Alicante-ISABIAL, Miguel Hernández University, CIBERER, Alicante, Spain
| | - Manuel D Gahete
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Cordoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain.,Reina Sofia University Hospital (HURS), Cordoba, Spain.,CIBER Physiopathology of Obesity and Nutrition (CIBERobn), Cordoba, Spain
| | - Raúl M Luque
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Cordoba, Spain - .,Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain.,Reina Sofia University Hospital (HURS), Cordoba, Spain.,CIBER Physiopathology of Obesity and Nutrition (CIBERobn), Cordoba, Spain
| |
Collapse
|
20
|
Sidaraite A, Liutkeviciene R, Glebauskiene B, Vilkeviciute A, Kriauciuniene L. Associations of cholesteryl ester transfer protein (CETP) gene variants with pituitary adenoma. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2019; 164:189-195. [PMID: 31012439 DOI: 10.5507/bp.2019.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Accepted: 04/04/2019] [Indexed: 01/06/2023] Open
Abstract
AIM The aim was to evaluate the association of CETP (rs5882 and rs708272) single nucleotide polymorphisms with the presence, invasiveness, hormonal activity and recurrence of pituitary adenoma (PA). METHODS The study group included 142 patients with PA and the control group, 753 healthy subjects. The genotyping of CETP (rs5882 and rs708272) was performed using a real-time PCR method. RESULTS After statistical analysis we found that CETP rs708272 genotype G/A under the over-dominant model was associated with the decreased odds of PA (OR=0.637; 95%CI: 0.443-0.917; P=0.015), active PA (OR=0.538; 95%CI: 0.335-0.865; P =0.01) and non-recurrent PA (OR=0.602; 95% CI: 0.402 - 0.902; P =0.014). When compared to controls, the rs708272 genotype G/A was less frequent in the active PA subgroup (37.5% vs 52.7%, P =0.009) and the non-recurrent PA subgroup (40.2% vs 52.7%, P=0.013), while the rs5882 genotype A/A was less frequent in the non-recurrent PA subgroup (37.5% vs 46.2%, P=0.015). CONCLUSION Our study showed that CETP rs708272 genotype G/A may be associated with a decreased risk of PA.
Collapse
Affiliation(s)
- Agne Sidaraite
- Lithuanian University of Health Sciences, Medical Academy, Eiveniu 2, Kaunas, Lithuania
| | - Rasa Liutkeviciene
- Neuroscience Institute, Lithuanian University of Health Sciences, Medical Academy, Eiveniu 2, Kaunas, Lithuania.,Department of Ophthalmology, Lithuanian University of Health Sciences, Medical Academy, Eiveniu 2, Kaunas, Lithuania
| | - Brigita Glebauskiene
- Department of Ophthalmology, Lithuanian University of Health Sciences, Medical Academy, Eiveniu 2, Kaunas, Lithuania
| | - Alvita Vilkeviciute
- Neuroscience Institute, Lithuanian University of Health Sciences, Medical Academy, Eiveniu 2, Kaunas, Lithuania
| | - Loresa Kriauciuniene
- Neuroscience Institute, Lithuanian University of Health Sciences, Medical Academy, Eiveniu 2, Kaunas, Lithuania.,Department of Ophthalmology, Lithuanian University of Health Sciences, Medical Academy, Eiveniu 2, Kaunas, Lithuania
| |
Collapse
|
21
|
Renner U, Ciato D, Stalla GK. Recent advances in understanding corticotroph pituitary tumor initiation and progression. F1000Res 2018; 7. [PMID: 30228864 PMCID: PMC6117851 DOI: 10.12688/f1000research.14789.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/23/2018] [Indexed: 11/20/2022] Open
Abstract
Cushing’s disease is the most frequent form of hypercortisolism and is caused by hypophyseal corticotroph adenomas secreting excessive amounts of adrenocorticotropic hormone. Most of the tumors develop sporadically and only a limited number of corticotroph adenomas have been found to be associated with different neuroendocrine syndromes or with familial isolated pituitary adenomas. The pathogenic mechanisms of corticotroph adenomas are largely unknown, but the discovered aberrant chaperoning activity of heat shock protein 90 on the one hand and the presence of ubiquitin-specific protease 8 mutations on the other hand partially explained the causes of their development. Corticotroph tumors arise initially as benign microadenomas but with time form invasively growing aggressive macroadenomas which can switch to corticotroph carcinomas in extremely rare cases. The mechanisms through which corticotroph tumors escape from glucocorticoid negative feedback are still poorly understood, as are the processes that trigger the progression of benign corticotroph adenomas toward aggressive and malignant phenotypes. This review summarizes recent findings regarding initiation and progression of corticotroph pituitary tumors.
Collapse
Affiliation(s)
- Ulrich Renner
- Max Planck Institute of Psychiatry, Clinical Neuroendocrinology Group, Munich, Germany
| | - Denis Ciato
- Max Planck Institute of Psychiatry, Clinical Neuroendocrinology Group, Munich, Germany
| | - Günter K Stalla
- Max Planck Institute of Psychiatry, Clinical Neuroendocrinology Group, Munich, Germany
| |
Collapse
|
22
|
It's raining MEN: Putting order to multiple endocrine neoplasms. ACTA ACUST UNITED AC 2018; 65:245-246. [PMID: 29706177 DOI: 10.1016/j.endinu.2018.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Accepted: 03/22/2018] [Indexed: 11/21/2022]
|
23
|
Hannah-Shmouni F, Stratakis CA. An update on the genetics of benign pituitary adenomas in children and adolescents. ACTA ACUST UNITED AC 2018; 1:19-24. [PMID: 30555957 DOI: 10.1016/j.coemr.2018.04.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Pituitary adenomas in children and adolescents are rare tumors that often result from a tumor predisposition syndrome. Several inherited causes for pituitary adenomas have been identified in the last few years, including multiple endocrine neoplasia type 1 and 4, Carney's complex, Tuberous sclerosis, DICER1 syndrome, neurofibromatosis type 1, McCune Albright syndrome, familial isolated pituitary adenoma, and pituitary adenoma association due to defects in succinate dehydrogenase genes. Recently, our group discovered X-linked acrogigantism (X-LAG), a new pediatric disorder that is caused by an Xq26.3 genomic duplication (involving the GPR101 gene). Genes that predispose to pediatric Cushing disease, including CABLES1 and USP8, were also recently identified. Genetic screening and counseling of affected or at risk individuals is a key component of their comprehensive care. In this review, we provide an up-to-date discussion on the latest pediatric genetic discoveries associated with pituitary adenomas with a focus on familial syndromes.
Collapse
Affiliation(s)
- Fady Hannah-Shmouni
- Section on Endocrinology & Genetics (SEGEN), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Constantine A Stratakis
- Section on Endocrinology & Genetics (SEGEN), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| |
Collapse
|
24
|
Nagata Y, Inoshita N, Fukuhara N, Yamaguchi-Okada M, Nishioka H, Iwata T, Yoshimoto K, Yamada S. Growth hormone-producing pituitary adenomas in childhood and young adulthood: clinical features and outcomes. Pituitary 2018; 21:1-9. [PMID: 28849339 DOI: 10.1007/s11102-017-0836-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE Growth hormone (GH)-producing pituitary adenomas (PAs) in childhood or young adulthood are rare, and the details surrounding these tumors remain enigmatic. We present the clinical, pathological and genetic features of this disease. METHODS We identified 25 patients aged 20 years or younger with GH-producing PAs who underwent surgery between 2003 and 2016 at Toranomon Hospital in Tokyo. We retrospectively reviewed the clinical data, treatment outcomes and pathological features of these patients to shed light on childhood acromegaly. RESULTS The cohort comprised 14 male and 11 female patients whose average age at the time of surgery was 17.3 years. Germline AIP mutations were present in 5 of 13 patients examined, and Carney complex was identified in 2 of 25 patients. The mean maximum tumor diameter was 26.7 mm, and total resection assessed during surgery was achieved in 17 patients. Based on their respective pathological findings, patients were divided into the following 4 groups: sparsely granulated adenomas (5), densely granulated (DG) adenomas (6), plurihormonal adenomas (9), and silent subtype 3 (SS3) adenomas (5). During the mean follow-up period of 50.3 months, complete endocrinological remission was achieved in 14 of 25 patients (56%) by surgery alone and in 19 patients (76%) after postoperative adjuvant therapy. CONCLUSIONS GH-producing PAs in young patients are intriguing and difficult to treat due to their distinct tumor characteristics, including a lower incidence of the DG subtype and a higher incidence of SS3 adenomas and genetic abnormalities. Therefore, multi-modal therapies are essential to achieve optimal clinical outcomes.
Collapse
Affiliation(s)
- Yuichi Nagata
- Department of Hypothalamic and Pituitary Surgery, Toranomon Hospital, 2-2-2 Toranomon, Minato-ku, Tokyo, 105-8470, Japan.
| | - Naoko Inoshita
- Department of Pathology, Toranomon Hospital, Tokyo, Japan
| | - Noriaki Fukuhara
- Department of Hypothalamic and Pituitary Surgery, Toranomon Hospital, 2-2-2 Toranomon, Minato-ku, Tokyo, 105-8470, Japan
| | - Mitsuo Yamaguchi-Okada
- Department of Hypothalamic and Pituitary Surgery, Toranomon Hospital, 2-2-2 Toranomon, Minato-ku, Tokyo, 105-8470, Japan
| | - Hiroshi Nishioka
- Department of Hypothalamic and Pituitary Surgery, Toranomon Hospital, 2-2-2 Toranomon, Minato-ku, Tokyo, 105-8470, Japan
| | - Takeo Iwata
- Department of Medical Pharmacology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Katsuhiko Yoshimoto
- Department of Medical Pharmacology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Shozo Yamada
- Department of Hypothalamic and Pituitary Surgery, Toranomon Hospital, 2-2-2 Toranomon, Minato-ku, Tokyo, 105-8470, Japan
| |
Collapse
|
25
|
Newey PJ, Thakker RV. Multiple Endocrine Neoplasia Syndromes. GENETICS OF BONE BIOLOGY AND SKELETAL DISEASE 2018:699-732. [DOI: 10.1016/b978-0-12-804182-6.00038-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
26
|
Guilmette J, Nosé V. Hereditary and familial thyroid tumours. Histopathology 2017; 72:70-81. [DOI: 10.1111/his.13373] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 08/22/2017] [Accepted: 08/24/2017] [Indexed: 01/16/2023]
Affiliation(s)
| | - Vania Nosé
- Massachusetts General Hospital; Boston MA USA
| |
Collapse
|
27
|
Alrezk R, Hannah-Shmouni F, Stratakis CA. MEN4 and CDKN1B mutations: the latest of the MEN syndromes. Endocr Relat Cancer 2017; 24:T195-T208. [PMID: 28824003 PMCID: PMC5623937 DOI: 10.1530/erc-17-0243] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 08/18/2017] [Indexed: 12/14/2022]
Abstract
Multiple endocrine neoplasia (MEN) refers to a group of autosomal dominant disorders with generally high penetrance that lead to the development of a wide spectrum of endocrine and non-endocrine manifestations. The most frequent among these conditions is MEN type 1 (MEN1), which is caused by germline heterozygous loss-of-function mutations in the tumor suppressor gene MEN1 MEN1 is characterized by primary hyperparathyroidism (PHPT) and functional or nonfunctional pancreatic neuroendocrine tumors and pituitary adenomas. Approximately 10% of patients with familial or sporadic MEN1-like phenotype do not have MEN1 mutations or deletions. A novel MEN syndrome was discovered, initially in rats (MENX), and later in humans (MEN4), which is caused by germline mutations in the putative tumor suppressor CDKN1B The most common phenotype of the 19 established cases of MEN4 that have been described to date is PHPT followed by pituitary adenomas. Recently, somatic or germline mutations in CDKN1B were also identified in patients with sporadic PHPT, small intestinal neuroendocrine tumors, lymphoma and breast cancer, demonstrating a novel role for CDKN1B as a tumor susceptibility gene for other neoplasms. In this review, we report on the genetic characterization and clinical features of MEN4.
Collapse
Affiliation(s)
- Rami Alrezk
- The National Institute of Diabetes and Digestive and Kidney DiseasesNational Institutes of Health, Bethesda, Maryland, USA
| | - Fady Hannah-Shmouni
- Section on Endocrinology & Geneticsthe Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland, USA
| | - Constantine A Stratakis
- Section on Endocrinology & Geneticsthe Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland, USA
| |
Collapse
|
28
|
Chen L, Zhou L, Zhang M, Shang L, Zhang P, Wang W, Fang C, Li J, Xu T, Tan H, Zhang P, Qiu M, Yu X, Jin K, Chen Y, Chen H, Lin R, Zhang Q, Shen L, Chen M, Li J, Li L, Chen J. Clinicopathological features and prognostic validity of WHO grading classification of SI-NENs. BMC Cancer 2017; 17:521. [PMID: 28778195 PMCID: PMC5543548 DOI: 10.1186/s12885-017-3490-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 07/18/2017] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND The clinicopathological characteristics of small intestinal neuroendocrine neoplasms (SI-NENs) and the prognostic validity of WHO grading classification for SI-NENs are still unknown in Asian patients. METHODS 277 patients and 8315 patients with SI-NENs were retrieved respectively from eleven Chinese hospitals and Surveillance, Epidemiology, and End Results (SEER) cancer registry. Overall survival was used as the major study outcome. Survival analysis using Kaplan-Meier analysis with log-rank test and cox regression analysis were applied. RESULTS Clinicopathological characteristics of SI-NENs were quite different among different races. Duodenum was the predominant tumor site in Chinese patients and Asian/Pacific Islander patients but not in white patients from SEER database. Patients with duodenal NENs tended to have more localized disease than patients with jejunal/ileal NENs which were confirmed by patients from SEER database. Grade 3 or poorly differentiated/undifferentiated tumor were more common and tumor size was significantly larger in ampullary NENs compared with that in non-ampullary duodenal NENs. As for the prognostic validity of WHO grading classification, survival between patients with grade 1 and grade 2 disease was not significantly different. Ki-67 index of 5% might be a better threshold between grade 1 and grade 2 than Ki-67 index of 2% in SI-NENs. CONCLUSIONS Our study revealed that the clinicopathological characteristics of SI-NENs among different races were quite different. This might because duodenal NENs was much more common in Chinese patients and Asian/Pacific Islander patients. Duodenal NENs and jejunal/ileal NENs, ampullary and non-ampullary duodenal NENs shared different characteristics. Ki-67 index of 5% might be a better threshold between grade 1 and grade 2 in SI-NENs.
Collapse
Affiliation(s)
- Luohai Chen
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan II Road, Yuexiu District, Guangzhou, 510080 China
| | - Lin Zhou
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Meng Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Liang Shang
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong University, No.324 Jingwu Road, Huaiyin District, Jinan, 250021 China
| | - Panpan Zhang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis & Translational Research under Ministry of Education, Peking University Cancer Hospital & Beijing Cancer Hospital, No.52 Fucheng Road, Haidian District, Beijing, 100142 China
| | - Wei Wang
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Cheng Fang
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Jingnan Li
- Department of Gastroenterology, Peking Union Medical College Hospital, Beijing, China
| | - Tianming Xu
- Department of Gastroenterology, Peking Union Medical College Hospital, Beijing, China
| | - Huangying Tan
- Department of Integrative Oncology, China-Japan Friendship Hospital, Beijing, China
| | - Pan Zhang
- Department of Integrative Oncology, China-Japan Friendship Hospital, Beijing, China
| | - Meng Qiu
- Department of Medical Oncology, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Kaizhou Jin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Ye Chen
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Huishan Chen
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Rong Lin
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Qin Zhang
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis & Translational Research under Ministry of Education, Peking University Cancer Hospital & Beijing Cancer Hospital, No.52 Fucheng Road, Haidian District, Beijing, 100142 China
| | - Minhu Chen
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan II Road, Yuexiu District, Guangzhou, 510080 China
| | - Jie Li
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis & Translational Research under Ministry of Education, Peking University Cancer Hospital & Beijing Cancer Hospital, No.52 Fucheng Road, Haidian District, Beijing, 100142 China
| | - Leping Li
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong University, No.324 Jingwu Road, Huaiyin District, Jinan, 250021 China
| | - Jie Chen
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan II Road, Yuexiu District, Guangzhou, 510080 China
| |
Collapse
|
29
|
Abstract
Although most of pituitary adenomas are benign, they may cause significant burden to patients. Sporadic adenomas represent the vast majority of the cases, where recognized somatic mutations (eg, GNAS or USP8), as well as altered gene-expression profile often affecting cell cycle proteins have been identified. More rarely, germline mutations predisposing to pituitary adenomas -as part of a syndrome (eg, MEN1 or Carney complex), or isolated to the pituitary (AIP or GPR101) can be identified. These alterations influence the biological behavior, clinical presentations and therapeutic responses, and their full understanding helps to provide appropriate care for these patients.
Collapse
Affiliation(s)
- Pedro Marques
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Márta Korbonits
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK.
| |
Collapse
|
30
|
Syro LV, Rotondo F, Serna CA, Ortiz LD, Kovacs K. Pathology of GH-producing pituitary adenomas and GH cell hyperplasia of the pituitary. Pituitary 2017; 20:84-92. [PMID: 27586499 DOI: 10.1007/s11102-016-0748-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Histologic, immunohistochemical and electron microscopic studies have provided conclusive evidence that a marked diversity exists between tumors which secrete growth hormone (GH) in excess. GH cell hyperplasia can also be associated with acromegaly in patients with extrapituitary GH-releasing hormone secreting tumors or in familial pituitary tumor syndromes. MATERIALS AND METHODS A literature search was performed for information regarding pathology, GH-producing tumors and acromegaly. RESULTS This review summarizes the current knowledge on the morphology of GH-producing and silent GH adenomas, as well as GH hyperplasia of the pituitary. CONCLUSION The importance of morphologic classification and identification of different subgroups of patients with GH-producing adenomas and their impact on clinical management is discussed.
Collapse
Affiliation(s)
- Luis V Syro
- Department of Neurosurgery, Hospital Pablo Tobon Uribe and Clinica Medellin, Medellin, Colombia.
| | - Fabio Rotondo
- Department of Laboratory Medicine, Division of Pathology, St. Michael's Hospital, University of Toronto, Toronto, ON, Canada
| | - Carlos A Serna
- Laboratorio de Patologia y Citologia Rodrigo Restrepo. Department of Pathology, Clinica Las Americas, Universidad CES, Medellin, Colombia
| | - Leon D Ortiz
- Division of Neuro-oncology, Instituto de Cancerologia, Clinica Las Americas, Medellin, Colombia
| | - Kalman Kovacs
- Department of Laboratory Medicine, Division of Pathology, St. Michael's Hospital, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
31
|
Maxwell JE, Howe JR. Pancreatic neuroendocrine tumors. BLUMGART'S SURGERY OF THE LIVER, BILIARY TRACT AND PANCREAS, 2-VOLUME SET 2017:997-1006.e3. [DOI: 10.1016/b978-0-323-34062-5.00065-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
32
|
Ferraù F, Romeo PD, Puglisi S, Ragonese M, Torre ML, Scaroni C, Occhi G, De Menis E, Arnaldi G, Trimarchi F, Cannavò S. Analysis of GPR101 and AIP genes mutations in acromegaly: a multicentric study. Endocrine 2016; 54:762-767. [PMID: 26815903 DOI: 10.1007/s12020-016-0862-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 01/08/2016] [Indexed: 12/17/2022]
Abstract
This multicentric study aimed to investigate the prevalence of the G protein-coupled receptor 101 (GPR101) p.E308D variant and aryl hydrocarbon receptor interacting protein (AIP) gene mutations in a representative cohort of Italian patients with acromegaly. 215 patients with GH-secreting pituitary adenomas, referred to 4 Italian referral centres for pituitary diseases, have been included. Three cases of gigantism were present. Five cases were classified as FIPA. All the patients have been screened for germline AIP gene mutations and GPR101 gene p.E308D variant. Heterozygous AIP gene variants have been found in 7 patients (3.2 %). Five patients carried an AIP mutation (2.3 %; 4 females): 3 patients harboured the p.R3O4Q mutation, one had the p.R304* mutation and the last one the IVS3+1G>A mutation. The prevalence of AIP mutations was 3.3 % and 2.8 % when considering only the patients diagnosed when they were <30 or <40-year old, respectively. Furthermore, 2.0 % of the patients with a pituitary macroadenoma and 4.2 % of patients resistant to somatostatin analogues treatment were found to harbour an AIP gene mutation. None of the patients was found to carry the GPR101 p.E308D variant. The prevalence of AIP gene mutations among our sporadic and familial acromegaly cases was similar to that one reported in previous studies, but lower when considering only the cases diagnosed before 40 years of age. The GPR101 p.E308D change is unlikely to have a role in somatotroph adenomas tumorigenesis, since none of our sporadic or familial patients tested positive for this variant.
Collapse
Affiliation(s)
- Francesco Ferraù
- Endocrine Unit, Department of Clinical and Experimental Medicine, University of Messina, UOC di Endocrinologia, Pad. H, 4° piano, AOU Policlinico Gaetano Martino, Via Consolare Valeria, 1, 98125, Messina, Italy.
| | - P D Romeo
- Endocrine Unit, Department of Clinical and Experimental Medicine, University of Messina, UOC di Endocrinologia, Pad. H, 4° piano, AOU Policlinico Gaetano Martino, Via Consolare Valeria, 1, 98125, Messina, Italy
| | - S Puglisi
- Endocrine Unit, Department of Clinical and Experimental Medicine, University of Messina, UOC di Endocrinologia, Pad. H, 4° piano, AOU Policlinico Gaetano Martino, Via Consolare Valeria, 1, 98125, Messina, Italy
| | - M Ragonese
- Endocrine Unit, Department of Clinical and Experimental Medicine, University of Messina, UOC di Endocrinologia, Pad. H, 4° piano, AOU Policlinico Gaetano Martino, Via Consolare Valeria, 1, 98125, Messina, Italy
| | - M L Torre
- Endocrine Unit, Department of Clinical and Experimental Medicine, University of Messina, UOC di Endocrinologia, Pad. H, 4° piano, AOU Policlinico Gaetano Martino, Via Consolare Valeria, 1, 98125, Messina, Italy
| | - C Scaroni
- Endocrinology Unit, Department of Medicine, Padova University Hospital, Padua, Italy
| | - G Occhi
- Department of Biology, University of Padova, Padua, Italy
| | - E De Menis
- Department of Internal Medicine, General Hospital of Montebelluna, Treviso, Italy
| | - G Arnaldi
- Department of Endocrinology and Metabolic Diseases, Azienda Ospedaliera Universitaria Ospedali Riuniti di Ancona, Ancona, Italy
| | - F Trimarchi
- Endocrine Unit, Department of Clinical and Experimental Medicine, University of Messina, UOC di Endocrinologia, Pad. H, 4° piano, AOU Policlinico Gaetano Martino, Via Consolare Valeria, 1, 98125, Messina, Italy
| | - S Cannavò
- Endocrine Unit, Department of Clinical and Experimental Medicine, University of Messina, UOC di Endocrinologia, Pad. H, 4° piano, AOU Policlinico Gaetano Martino, Via Consolare Valeria, 1, 98125, Messina, Italy
| |
Collapse
|
33
|
Yamada T, Kanoh M, Nabe S, Yasuoka T, Suzuki J, Matsumoto A, Kuwahara M, Maruyama S, Fujimoto T, Sakisuka R, Yasukawa M, Yamashita M. Menin Plays a Critical Role in the Regulation of the Antigen-Specific CD8+ T Cell Response upon Listeria Infection. THE JOURNAL OF IMMUNOLOGY 2016; 197:4079-4089. [PMID: 27798149 DOI: 10.4049/jimmunol.1502295] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 09/12/2016] [Indexed: 02/06/2023]
Abstract
Menin, a tumor suppressor protein, is encoded by the MEN1 gene in humans. Certain germinal mutations of MEN1 induce an autosomal-dominant syndrome that is characterized by concurrent parathyroid adenomas and several other tumor types. Although menin is also expressed in hematopoietic lineages, its role in CD8+ T cells remains unclear. We generated Meninflox/flox CD4-Cre (Menin-KO) mice by crossing Meninflox/flox mice with CD4-Cre transgenic (Tg) mice to determine the role of menin in CD8+ T cells. Wild-type (WT) and Menin-KO mice were infected with Listeria monocytogenes expressing OVA to analyze the immune response of Ag-specific CD8+ T cells. Menin deficiency resulted in an impaired primary immune response by CD8+ T cells. On day 7, there were fewer Menin-KO OVA-specific CD8+ T cells compared with WT cells. Next, we adoptively transferred WT and Menin-KO OT-1 Tg CD8+ T cells into congenic recipient mice and infected them with L. monocytogenes expressing OVA to determine the CD8+ T cell-intrinsic effect. Menin-KO OT-1 Tg CD8+ T cells were outcompeted by the WT cells upon infection. Increased expression of Blimp-1 and T-bet, cell cycle inhibitors, and proapoptotic genes was observed in the Menin-KO OT-1 Tg CD8+ T cells upon infection. These data suggest that menin inhibits differentiation into terminal effectors and positively controls proliferation and survival of Ag-specific CD8+ T cells that are activated upon infection. Collectively, our study uncovered an important role for menin in the immune response of CD8+ T cells to infection.
Collapse
Affiliation(s)
- Takeshi Yamada
- Department of Infection and Host Defenses, Graduate School of Medicine, Ehime University, Shitsukawa, Toon, Ehime 791-0295, Japan;
| | - Makoto Kanoh
- Department of Infection and Host Defenses, Graduate School of Medicine, Ehime University, Shitsukawa, Toon, Ehime 791-0295, Japan
| | - Shogo Nabe
- Department of Hematology, Clinical Immunology, and Infectious diseases, Graduate School of Medicine, Ehime University, Shitsukawa, Toon, Ehime 791-0295, Japan
| | - Toshiaki Yasuoka
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Ehime University, Shitsukawa, Toon, Ehime 791-0295, Japan
| | - Junpei Suzuki
- Department of Hematology, Clinical Immunology, and Infectious diseases, Graduate School of Medicine, Ehime University, Shitsukawa, Toon, Ehime 791-0295, Japan.,Department of Immunology, Graduate School of Medicine, Ehime University, Shitsukawa, Toon, Ehime 791-0295, Japan; and.,Translational Research Center, Ehime University Hospital, Shitsukawa, Toon, Ehime 791-0295, Japan
| | - Akira Matsumoto
- Department of Infection and Host Defenses, Graduate School of Medicine, Ehime University, Shitsukawa, Toon, Ehime 791-0295, Japan
| | - Makoto Kuwahara
- Department of Immunology, Graduate School of Medicine, Ehime University, Shitsukawa, Toon, Ehime 791-0295, Japan; and.,Translational Research Center, Ehime University Hospital, Shitsukawa, Toon, Ehime 791-0295, Japan
| | - Saho Maruyama
- Department of Immunology, Graduate School of Medicine, Ehime University, Shitsukawa, Toon, Ehime 791-0295, Japan; and
| | - Takuya Fujimoto
- Department of Infection and Host Defenses, Graduate School of Medicine, Ehime University, Shitsukawa, Toon, Ehime 791-0295, Japan
| | - Ryo Sakisuka
- Department of Infection and Host Defenses, Graduate School of Medicine, Ehime University, Shitsukawa, Toon, Ehime 791-0295, Japan
| | - Masaki Yasukawa
- Department of Hematology, Clinical Immunology, and Infectious diseases, Graduate School of Medicine, Ehime University, Shitsukawa, Toon, Ehime 791-0295, Japan
| | - Masakatsu Yamashita
- Department of Immunology, Graduate School of Medicine, Ehime University, Shitsukawa, Toon, Ehime 791-0295, Japan; and.,Translational Research Center, Ehime University Hospital, Shitsukawa, Toon, Ehime 791-0295, Japan
| |
Collapse
|
34
|
Hannah-Shmouni F, Trivellin G, Stratakis CA. Genetics of gigantism and acromegaly. Growth Horm IGF Res 2016; 30-31:37-41. [PMID: 27657986 PMCID: PMC5154831 DOI: 10.1016/j.ghir.2016.08.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 07/28/2016] [Accepted: 08/09/2016] [Indexed: 12/11/2022]
Abstract
Gigantism and acromegaly are rare disorders that are caused by excessive GH secretion and/or high levels of its mediator, IGF-1. Gigantism occurs when excess GH or IGF-1 lead to increased linear growth, before the end of puberty and epiphyseal closure. The majority of cases arise from a benign GH-secreting pituitary adenoma, with an incidence of pituitary gigantism and acromegaly of approximately 8 and 11 per million person-years, respectively. Over the past two decades, our increasing understanding of the molecular and genetic etiologies of pituitary gigantism and acromegaly yielded several genetic causes, including multiple endocrine neoplasia type 1 and 4, McCune-Albright syndrome, Carney complex, familial isolated pituitary adenoma, pituitary adenoma association due to defects in familial succinate dehydrogenase genes, and the recently identified X-linked acrogigantism. The early diagnosis of these conditions helps guide early intervention, screening, and genetic counseling of patients and their family members. In this review, we provide a concise and up-to-date discussion on the genetics of gigantism and acromegaly.
Collapse
Affiliation(s)
- Fady Hannah-Shmouni
- Section on Endocrinology & Genetics (SEGEN), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Giampaolo Trivellin
- Section on Endocrinology & Genetics (SEGEN), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Constantine A Stratakis
- Section on Endocrinology & Genetics (SEGEN), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892, USA.
| |
Collapse
|
35
|
Abstract
We often think of the lentiginoses, phacomatoses and other neurocutaneous syndromes as conditions that affect the skin and also predispose to a variety of tumors. However, we rarely think of Peutz-Jeghers syndrome (PJS), Carney complex (CNC), Cowden disease (CD), neurofibromatosis type-1 (NF-1) or tuberous sclerosis (TSC) as conditions that are multiple endocrine neoplasias (MEN). Indeed, all of these conditions predispose to a variety of endocrine tumors, in addition to many other neoplasms. On the other hand, the classic MENs, type 1 and 2 (MEN-1 and MEN-2, respectively) are almost never thought in terms of their skin manifestations. In this review, we present extensively the MEN-1, MEN-2 and PJS syndromes, and briefly refer to CD, NF-1, and TSC. CNC is discussed in another article in this journal issue.
Collapse
Affiliation(s)
- Constantine A Stratakis
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Building 10, Room East 1330, CRC, 10 Center Dr. MSC1862, Bethesda, MD, 20892-1862, USA.
| |
Collapse
|
36
|
Neggers SJCMM, Muhammad A, van der Lely AJ. Pegvisomant Treatment in Acromegaly. Neuroendocrinology 2016; 103:59-65. [PMID: 25792221 DOI: 10.1159/000381644] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 03/04/2015] [Indexed: 11/19/2022]
Abstract
Historically, medical treatment of acromegaly has mainly been used as an adjuvant therapy after surgery. In the last decades, an increased range of medical therapy options has been available. Somatostatin analogues have become the cornerstones of medical treatment in acromegaly and are even seen as a primary treatment in a selected group of acromegaly patients. The most recent medical treatment available for acromegaly patients is pegvisomant, a growth hormone receptor antagonist. To date, it is the most effective medical treatment, but it is costly. Pegvisomant is used as monotherapy and combined with somatostatin analogues. In this article, we review clinical studies and cohorts that have documented the efficacy of pegvisomant monotherapy and combined therapy and give a concise overview of associated side effects.
Collapse
Affiliation(s)
- Sebastian J C M M Neggers
- Section of Endocrinology, Department of Medicine, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | | | | |
Collapse
|
37
|
Abstract
Available disease-specific questionnaires like the Acromegaly Quality of Life questionnaire have confirmed that quality of life (QoL) is impaired in acromegaly, especially in active disease. Successful therapy improves QoL, but it may not normalize completely even after endocrine cure; furthermore, there is not always a correlation between growth hormone (GH) and insulin-like growth factor 1 and subjective health perception of QoL. Appearance is the dimension most affected and has the highest impact on the patient's QoL. Worse QoL is associated with the presence of musculoskeletal pain, headache (if only medical therapy, not surgery, has been provided), having required treatment with radiotherapy, being older, of female gender, with a longer disease duration, coexisting diabetes mellitus, a higher BMI or becoming GH deficient after treatment for acromegaly.
Collapse
Affiliation(s)
- Susan M Webb
- Endocrinology/Medicine Departments, Hospital Sant Pau, Centro de Investigacix00F3;n Biomx00E9;dica en Red de Enfermedades Raras (CIBER-ER, Unidad 747), ISCIII, IIB-Sant Pau, Barcelona, Spain
| | | |
Collapse
|
38
|
McCabe J, Ayuk J, Sherlock M. Treatment Factors That Influence Mortality in Acromegaly. Neuroendocrinology 2016; 103:66-74. [PMID: 25661647 DOI: 10.1159/000375163] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 01/12/2015] [Indexed: 11/19/2022]
Abstract
Acromegaly is a rare condition characterized by excessive secretion of growth hormone (GH), which is almost always due to a pituitary adenoma. Acromegaly is associated with significant morbidity such as hypertension, type 2 diabetes, cardiomyopathy, obstructive sleep apnoea, malignancy and musculoskeletal abnormalities. Acromegaly has also been associated with increased mortality in several retrospective studies. This review will focus on the epidemiological data relating to mortality rates in acromegaly, the relationship between acromegaly and malignancy, the role of GH and insulin-like growth factor-I in assessing the risk of future mortality, and the impact of radiotherapy and hypopituitarism on mortality.
Collapse
Affiliation(s)
- John McCabe
- Department of Endocrinology, Adelaide and Meath Hospital incorporating the National Children's Hospital, Tallaght, Ireland
| | | | | |
Collapse
|
39
|
Claessen KMJA, Mazziotti G, Biermasz NR, Giustina A. Bone and Joint Disorders in Acromegaly. Neuroendocrinology 2016; 103:86-95. [PMID: 25633971 DOI: 10.1159/000375450] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 01/22/2015] [Indexed: 11/19/2022]
Abstract
Acromegaly is a chronic, progressive disease caused by a growth hormone (GH)-producing pituitary adenoma, resulting in elevated GH and insulin-like growth factor 1 concentrations. Following appropriate therapy (surgery, radiotherapy and/or medical treatment), many systemic GH-induced comorbid conditions improve considerably. Unfortunately, despite biochemical control, acromegaly patients suffer from a high prevalence of late manifestations of transient GH excess, significantly impairing their quality of life. In this overview article, we summarize the pathophysiology, diagnosis, clinical picture, disease course and management of skeletal complications of acromegaly, focusing on vertebral fractures and arthropathy.
Collapse
Affiliation(s)
- Kim M J A Claessen
- Department of Endocrinology and Metabolism, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | |
Collapse
|
40
|
Powlson AS, Gurnell M. Cardiovascular Disease and Sleep-Disordered Breathing in Acromegaly. Neuroendocrinology 2016; 103:75-85. [PMID: 26227953 DOI: 10.1159/000438903] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 07/15/2015] [Indexed: 11/19/2022]
Abstract
Treatment goals in acromegaly include symptom relief, tumour control and reversal of the excess morbidity and mortality associated with the disorder. Cardiovascular complications include concentric biventricular hypertrophy and cardiomyopathy, hypertension, valvular heart disease and arrhythmias, while metabolic disturbance (insulin resistance/diabetes mellitus, dyslipidaemia) further increases the risk of cardiovascular and cerebrovascular events. Sleep-disordered breathing (in the form of sleep apnoea) is also common in patients with acromegaly and may exacerbate cardiovascular dysfunction, in addition to contributing to impaired quality of life. Accordingly, and in keeping with evidence that cardiorespiratory complications in acromegaly are not automatically reversed/ameliorated simply through the attainment of 'safe' growth hormone and insulin-like growth factor 1 levels, recent guidelines have emphasised the need not only to achieve stringent biochemical control, but also to identify and independently treat these comorbidities. It is important, therefore, that patients with acromegaly are systematically screened at diagnosis, and periodically thereafter, for the common cardiovascular and respiratory manifestations and that biochemical targets do not become the only treatment goal.
Collapse
Affiliation(s)
- Andrew S Powlson
- Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge and National Institute for Health Research Cambridge Biomedical Research Centre, Addenbrooke's Hospital, Cambridge, UK
| | | |
Collapse
|
41
|
Chanson P. Medical Treatment of Acromegaly with Dopamine Agonists or Somatostatin Analogs. Neuroendocrinology 2016; 103:50-8. [PMID: 25677539 DOI: 10.1159/000377704] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 02/04/2015] [Indexed: 11/19/2022]
Abstract
Treatment of acromegaly aims to correct (or prevent) tumor compression of surrounding tissues by excising the disease-causing lesion and reduce growth hormone (GH) and IGF-1 levels to normal values. When surgery (the usual first-line treatment) fails to correct GH/IGF-1 hypersecretion, medical treatment with dopamine agonists (DAs; particularly cabergoline) or somatostatin analogs (SAs) can be used. The GH receptor antagonist pegvisomant is helpful in patients who are totally or partially resistant to SAs and can be given in association with both SAs and/or DAs. Thanks to this multistep therapeutic strategy, adequate hormonal disease control is achieved in most patients, giving them normal life expectancy. Comorbidities associated with acromegaly generally improve after treatment, but persistent sequelae may nonetheless impair quality of life.
Collapse
Affiliation(s)
- Philippe Chanson
- Service d'Endocrinologie et des Maladies de la Reproduction and Centre de Référence des Maladies Endocriniennes Rares de la Croissance, Hôpital Bicêtre, Assistance Publique-Hôpitaux de Paris, and Inserm 1185, Fac Med Paris Sud, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| |
Collapse
|
42
|
Dal J, List EO, Jørgensen JOL, Berryman DE. Glucose and Fat Metabolism in Acromegaly: From Mice Models to Patient Care. Neuroendocrinology 2016; 103:96-105. [PMID: 25925240 DOI: 10.1159/000430819] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 04/20/2015] [Indexed: 11/19/2022]
Abstract
Patients with active acromegaly are frequently insulin resistant, glucose intolerant, and at risk for developing overt type 2 diabetes. At the same time, these patients have a relatively lean phenotype associated with mobilization and oxidation of free fatty acids. These features are reversed by curative surgical removal of the growth hormone (GH)-producing adenoma. Mouse models of acromegaly share many of these characteristics, including a lean phenotype and proneness to type 2 diabetes. There are, however, also species differences with respect to oxidation rates of glucose and fat as well as the specific mechanisms underlying GH-induced insulin resistance. The impact of acromegaly treatment on insulin sensitivity and glucose tolerance depends on the treatment modality (e.g. somatostatin analogs also suppress insulin secretion, whereas the GH antagonist restores insulin sensitivity). The interplay between animal research and clinical studies has proven useful in the field of acromegaly and should be continued in order to understand the metabolic actions of GH.
Collapse
Affiliation(s)
- Jakob Dal
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | | | | | | |
Collapse
|
43
|
Hannon MJ, Barkan AL, Drake WM. The Role of Radiotherapy in Acromegaly. Neuroendocrinology 2016; 103:42-9. [PMID: 26088716 DOI: 10.1159/000435776] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 06/04/2015] [Indexed: 11/19/2022]
Abstract
Radiotherapy has, historically, played a central role in the management of acromegaly, and the last 30 years have seen substantial improvements in the technology used in the delivery of radiation therapy. More recently, the introduction of highly targeted radiotherapy, or 'radiosurgery', has further increased the therapeutic options available in the management of secretory pituitary tumors. Despite these developments, improvements in primary surgical outcomes, an increase in the range and effectiveness of medical therapy options, and long-term safety concerns have combined to dictate that, although still deployed in selected cases, the use of radiotherapy in the management of acromegaly has declined steadily over the past 2 decades. In this article, we review some of the main studies that have documented the efficacy of pituitary radiotherapy on growth hormone hypersecretion and summarize the data around its potential deleterious effects, including hypopituitarism, cranial nerve damage, and the development of radiation-related intracerebral tumors. We also give practical recommendations to guide its future use in patients with acromegaly, generally, as a third-line intervention after neurosurgical intervention in combination with various medical therapy options.
Collapse
Affiliation(s)
- Mark J Hannon
- Department of Endocrinology, St. Bartholomew's Hospital, London, UK
| | | | | |
Collapse
|
44
|
Buchfelder M, Schlaffer SM. Novel Techniques in the Surgical Treatment of Acromegaly: Applications and Efficacy. Neuroendocrinology 2016; 103:32-41. [PMID: 26536097 DOI: 10.1159/000441980] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2015] [Accepted: 10/27/2015] [Indexed: 11/19/2022]
Abstract
Since the establishment of transsphenoidal microsurgery as the operative treatment of choice in most patients with acromegaly 40 years ago, a few novel technical developments have evolved. Their application, utility and efficacy will be briefly discussed in this review article, based on an analysis of published results and the authors' personal experience. The endoscope was additionally used to search for residual tumours in locations which could not be visualised with the operating microscope. In many centres it has by now fully replaced the operating microscope. Extended endoscopic operations hardly have limits in respect to accessible pathology. Overall, the results and complications reported from microsurgical and endoscopic series are comparable. Intraoperative magnetic resonance imaging allows depicting the completeness of a tumour resection. While in many patients additional tumour resections are performed on the basis of intraoperative imaging, the improvements in hormonal remission rates reported are less impressive. Neuronavigation uses imaging data to improve the surgeon's orientation, and it is certainly a major asset to the inexperienced. In high-caseload centres it is mainly appreciated in anatomical variants and reoperations. While the Doppler probe is a valuable and easily affordable gadget to avoid vascular arterial injury, intraoperative ultrasound imaging of tumour extension has a much poorer resolution than magnetic resonance imaging and is thus not widely implemented. The clinical value of intraoperative growth hormone measurements is controversially discussed. In summary, the application of modern technology has only led to a minor improvement of results, but it has widened the spectrum of accessible pathologies and increased the safety of the procedures for the patient. It is expected that outcomes will continue to improve as novel techniques and concepts are being developed.
Collapse
Affiliation(s)
- Michael Buchfelder
- Department of Neurosurgery, Friedrich-Alexander-Universitx00E4;t Erlangen-Nx00FC;rnberg, Erlangen, Germany
| | | |
Collapse
|
45
|
Mir-Bonafé JM, Vicente-Villa A, Suñol M, Soria X, González-Enseñat MA. Cutaneous Mucinosis of Infancy: A Rare Congenital Case with Coexisting Progressive, Eruptive, and Spontaneously Involuting Lesions. Pediatr Dermatol 2015; 32:e255-8. [PMID: 26446951 DOI: 10.1111/pde.12679] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Cutaneous mucinosis in infancy is rare. We report an infant with multiple congenital papules distributed over the trunk, neck, and extremities. These papules were mainly dispersed, but they also coalesced into plaques. Histopathologic findings showed features of cutaneous mucinosis of infancy (CMI). Over 2 years of follow-up, we observed that the preexisting lesions on the lower back and left trunk progressively increased in size, and a few new scattered papules continued to appear, mainly on the trunk; several lesions spontaneously resolved with no further complications. CMI is considered to be a persistent cutaneous disorder, even though spontaneously regressing cases have rarely been reported. This case demonstrates the broad clinical spectrum of CMI, with progressive, eruptive, and spontaneously involuting lesions all present in the same patient. This condition should be considered in the differential diagnosis of congenital or infantile-onset papules and plaques, especially those yellowish in color.
Collapse
Affiliation(s)
- José M Mir-Bonafé
- Department of Dermatology, Hospital Sant Joan de Déu, University of Barcelona, Barcelona, Spain
| | - Asunción Vicente-Villa
- Department of Dermatology, Hospital Sant Joan de Déu, University of Barcelona, Barcelona, Spain
| | - Mariona Suñol
- Department of Pathology, Hospital Sant Joan de Déu, University of Barcelona, Barcelona, Spain
| | - Xavier Soria
- Department of Dermatology, Hospital Universitari Arnau de Vilanova, University of Lleida, Lleida, Spain
| | | |
Collapse
|
46
|
Roelfsema F, van den Berg G. Diagnosis, treatment and clinical perspectives of acromegaly. Expert Rev Endocrinol Metab 2015; 10:619-644. [PMID: 30289037 DOI: 10.1586/17446651.2015.1096770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Acromegaly is an insidious disease of the pituitary caused by a growth hormone-secreting adenoma. Generally, the diagnosis is made rather late in the course of the disease. Currently, acromegaly can be cured in about half of the patients with the disease by expert surgery. The remainder of non-surgically cured patients often can be effectively treated with somatostatin analogs; either with the new generation of dopaminergic drugs or with Pegvisomant, a GH-receptor blocking agent. However, at the time of diagnosis many patients suffer from serious comorbidities, including hypertension, heart disease, arthrosis, sleep apnea and diabetes mellitus. Recent reports have shown that mortality risk can be normalized. Nevertheless, all efforts should be undertaken to treat comorbidities. New strategies for surgery and medical treatment are discussed.
Collapse
Affiliation(s)
- Ferdinand Roelfsema
- a Department of Endocrinology and Metabolism , Leiden University Medical Center , Leiden , The Netherlands
| | - Gerrit van den Berg
- b Department of Endocrinology and Metabolic Diseases, University Medical Center of Groningen , University of Groningen , Groningen , The Netherlands
| |
Collapse
|
47
|
Bruno W, Fornarini G, Ghiorzo P. Signs and genetics of rare cancer syndromes with gastroenterological features. World J Gastroenterol 2015; 21:8985-8993. [PMID: 26290627 PMCID: PMC4533032 DOI: 10.3748/wjg.v21.i30.8985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 05/26/2015] [Accepted: 07/15/2015] [Indexed: 02/07/2023] Open
Abstract
Although the genetic bases of most hereditary cancer syndromes are known, and genetic tests are available for them, the incidence of the most rare of these syndromes is likely underestimated, partially because the clinical expression is neither fully understood nor easily diagnosed due to the variable and complex expressivity. The clinical features of a small pool of rare cancer syndromes include gastroenterological signs, though not necessarily tumors, that could require the intervention of a gastroenterologist during any of the phases of the clinical management. Herein we will attempt to spread the knowledge on these rare syndromes by summarizing the phenotype and genetic basis, and revising the peculiar gastroenterological signs whose underlying role in these rare hereditary cancer syndromes is often neglected. Close collaboration between geneticists and gastroenterologists could facilitate both the early identification of patients or relatives at-risk and the planning of multidisciplinary and tailored management of these subjects.
Collapse
|
48
|
Syro LV, Rotondo F, Ramirez A, Di Ieva A, Sav MA, Restrepo LM, Serna CA, Kovacs K. Progress in the Diagnosis and Classification of Pituitary Adenomas. Front Endocrinol (Lausanne) 2015; 6:97. [PMID: 26124750 PMCID: PMC4464221 DOI: 10.3389/fendo.2015.00097] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 05/26/2015] [Indexed: 12/12/2022] Open
Abstract
Pituitary adenomas are common neoplasms. Their classification is based upon size, invasion of adjacent structures, sporadic or familial cases, biochemical activity, clinical manifestations, morphological characteristics, response to treatment and recurrence. Although they are considered benign tumors, some of them are difficult to treat due to their tendency to recur despite standardized treatment. Functional tumors present other challenges for normalizing their biochemical activity. Novel approaches for early diagnosis, as well as different perspectives on classification, may help to identify subgroups of patients with similar characteristics, creating opportunities to match each patient with the best personalized treatment option. In this paper, we present the progress in the diagnosis and classification of different subgroups of patients with pituitary tumors that may be managed with specific considerations according to their tumor subtype.
Collapse
Affiliation(s)
- Luis V. Syro
- Department of Neurosurgery, Hospital Pablo Tobon Uribe and Clinica Medellin, Medellin, Colombia
| | - Fabio Rotondo
- Laboratory Medicine, Division of Pathology, St. Michael’s Hospital, University of Toronto, Toronto, ON, Canada
| | - Alex Ramirez
- Department of Endocrinology, Universidad Pontificia Bolivariana, Medellin, Colombia
| | - Antonio Di Ieva
- Department of Neurosurgery, Australian School of Advanced Medicine, Macquarie University, Sydney, NSW, Australia
| | | | | | - Carlos A. Serna
- Laboratorio de Patologia y Citologia Rodrigo Restrepo, Department of Pathology, Clinica Medellin, Medellin, Colombia
| | - Kalman Kovacs
- Laboratory Medicine, Division of Pathology, St. Michael’s Hospital, University of Toronto, Toronto, ON, Canada
| |
Collapse
|