1
|
Vella M, Manfield IW, Seychell BC, Trinh CH, Rambo R, Nasir Khan G, Vassallo J, Hunter T, Hunter GJ. Mutations in the N-domain of aryl hydrocarbon receptor interacting protein affect interactions with heat shock protein 90β and phosphodiesterase 4A5. Biochimie 2025; 228:114-126. [PMID: 39299536 DOI: 10.1016/j.biochi.2024.09.005] [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: 07/04/2024] [Revised: 09/03/2024] [Accepted: 09/13/2024] [Indexed: 09/22/2024]
Abstract
The aryl hydrocarbon receptor interacting protein (AIP) is a cytoplasmic molecular co-chaperone and tumour suppressor that assists in protein stability and complex formation involving the aryl hydrocarbon receptor. Germline mutations in the AIP gene predispose to pituitary tumourigenesis with patients exhibiting an aggressive clinical phenotype. Full length AIP proteins harbouring N-domain mutations (R9Q, R16H, V49 M and K103R) were purified from E.coli utilizing a methodology that maintained structural integrity and monomeric stability. Mutations did not significantly affect the thermal stability of the protein and caused no overall disruptive effect in the protein structure. The mutations studied lowered the binding affinity of AIP towards two of its binding partners; heat shock protein 90β and phosphodiesterase 4A5 (PDE4A5). The inhibition of phosphodiesterase activity by AIP was also greatly reduced by all mutants. While previously published data has mainly concentrated on the tetratricopeptide repeats of the C-domain of AIP, we present clear evidence that AIP N-domain mutations play a significant role in two protein:protein interactions with partner proteins. The complex interactome of AIP suggests that any observable change in one or more of its binding partners cannot be disregarded as it may have repercussions on other biochemical pathways.
Collapse
Affiliation(s)
- Marita Vella
- Department of Physiology & Biochemistry, Faculty of Medicine & Surgery, University of Malta, Msida, MSD2080, Malta
| | - Iain W Manfield
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - Brandon C Seychell
- Department of Physiology & Biochemistry, Faculty of Medicine & Surgery, University of Malta, Msida, MSD2080, Malta
| | - Chi H Trinh
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - Robert Rambo
- Soft Condensed Matter Group, Diamond Light Source Ltd, Diamond House, Harwell Science and Innovation Campus, Didcot, Oxfordshire, OX11 0DE, UK
| | - G Nasir Khan
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - Josanne Vassallo
- Department of Medicine, Faculty of Medicine & Surgery, University of Malta, Msida, MSD2080, Malta
| | - Thérèse Hunter
- Department of Physiology & Biochemistry, Faculty of Medicine & Surgery, University of Malta, Msida, MSD2080, Malta
| | - Gary J Hunter
- Department of Physiology & Biochemistry, Faculty of Medicine & Surgery, University of Malta, Msida, MSD2080, Malta.
| |
Collapse
|
2
|
Hernández-Ramírez LC, Perez-Rivas LG, Theodoropoulou M, Korbonits M. An Update on the Genetic Drivers of Corticotroph Tumorigenesis. Exp Clin Endocrinol Diabetes 2024; 132:678-696. [PMID: 38830604 DOI: 10.1055/a-2337-2265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
The genetic landscape of corticotroph tumours of the pituitary gland has dramatically changed over the last 10 years. Somatic changes in the USP8 gene account for the most common genetic defect in corticotrophinomas, especially in females, while variants in TP53 or ATRX are associated with a subset of aggressive tumours. Germline defects have also been identified in patients with Cushing's disease: some are well-established (MEN1, CDKN1B, DICER1), while others are rare and could represent coincidences. In this review, we summarise the current knowledge on the genetic drivers of corticotroph tumorigenesis, their molecular consequences, and their impact on the clinical presentation and prognosis.
Collapse
Affiliation(s)
- Laura C Hernández-Ramírez
- Red de Apoyo a la Investigación, Coordinación de la Investigación Científica, Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | | | - Marily Theodoropoulou
- Medizinische Klinik und Poliklinik IV, LMU Klinikum, LMU München, Munich 80336, Germany
| | - Márta Korbonits
- Centre for Endocrinology, Barts and The London School of Medicine, Queen Mary University of London, Charterhouse Square, London, UK
| |
Collapse
|
3
|
Bizzi MF, Drummond JB, Pinheiro SVB, Paulino E, Araújo SA, Soares BS, Giannetti AV, Schweizer JRDOL, Barry S, Korbonits M, Ribeiro-Oliveira A. Activated AMP-protein kinase (pAMPK) is overexpressed in human somatotroph pituitary adenomas. Mol Cell Endocrinol 2024; 592:112318. [PMID: 38908427 DOI: 10.1016/j.mce.2024.112318] [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: 04/28/2024] [Revised: 06/17/2024] [Accepted: 06/18/2024] [Indexed: 06/24/2024]
Abstract
INTRODUCTION AMPK (AMP-activated protein kinase) is an enzyme that acts as a metabolic sensor and regulates multiple pathways via phosphorylating proteins in metabolic and proliferative pathways. The aim of this work was to study the activated cellular AMPK (phosphorylated-AMPK at Thr172, pAMPK) levels in pituitary tumor samples from patients with sporadic and familial acromegaly, as well as in samples from normal human pituitary gland. METHODS We studied pituitary adenoma tissue from patients with sporadic somatotroph adenomas, familial acromegaly with heterozygote germline variants in the aryl hydrocarbon receptor interacting protein (AIP) gene (p.Q164*, p.R304* and p.F269_H275dup) and autopsy from normal pituitary glands without structural alterations. RESULTS Cellular levels of pAMPK were significantly higher in patients with sporadic acromegaly compared to normal pituitary glands (p < 0.0001). Tissues samples from patients with germline AIP mutations also showed higher cellular levels of pAMPK compared to normal pituitary glands. We did not observe a significant difference in cellular levels of pAMPK according to the cytokeratin (CAM5.2) pattern (sparsely or densely granulated) for tumor samples of sporadic acromegaly. CONCLUSION Our data show, for the first time in human cells, an increase of cellular levels of pAMPK in sporadic somatotropinomas, regardless of cytokeratin pattern, as well as in GH-secreting adenomas from patients with germline AIP mutations.
Collapse
Affiliation(s)
- Mariana Ferreira Bizzi
- Departments of Internal Medicine of the Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, 30130-100, Brazil
| | - Juliana Beaudette Drummond
- Departments of Internal Medicine of the Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, 30130-100, Brazil
| | - Sergio Veloso Brant Pinheiro
- Departments of Pediatrics of the Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, 30130-100, Brazil
| | - Eduardo Paulino
- Departments of Pathology of the Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, 30130-100, Brazil
| | - Stanley Almeida Araújo
- Departments of Pathology of the Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, 30130-100, Brazil
| | - Beatriz Santana Soares
- Departments of Internal Medicine of the Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, 30130-100, Brazil
| | - Alexandre V Giannetti
- Departments of Surgery of the Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, 30130-100, Brazil
| | | | - Sayka Barry
- Centre for Endocrinology of Queen Mary University of London, London, EC1M 6BQ, UK
| | - Márta Korbonits
- Centre for Endocrinology of Queen Mary University of London, London, EC1M 6BQ, UK
| | - Antonio Ribeiro-Oliveira
- Departments of Internal Medicine of the Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, 30130-100, Brazil.
| |
Collapse
|
4
|
Maiter D, Chanson P, Constantinescu SM, Linglart A. Diagnosis and management of pituitary adenomas in children and adolescents. Eur J Endocrinol 2024; 191:R55-R69. [PMID: 39374844 DOI: 10.1093/ejendo/lvae120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/11/2024] [Accepted: 09/27/2024] [Indexed: 10/09/2024]
Abstract
BACKGROUND Pituitary adenomas (PAs)-also now called pituitary neuroendocrine tumours or Pit-NETS-are rare in children and adolescents and exceptional below the age of 10. Most evidence-based high-quality data are derived from larger studies in adult patients. AIMS We will review recent knowledge on the epidemiology, clinical features, diagnosis, and treatment modalities of the different types of pituitary adenomas diagnosed in children and adolescents, emphasizing the many reasons why these cases should be discussed within pituitary-specific multidisciplinary teams with experts from both paediatric and adult practice. CONCLUSIONS Paediatric PA presents multiple peculiarities that may challenge their adequate management. They are overall proportionally larger and more aggressive than in adults, with potential mass effects including hypopituitarism. Hormonal hypersecretion is frequent, resulting in clinical syndromes affecting normal growth and pubertal development. Prolactinomas represent the most frequent subtype of PA found during childhood, followed by adrenocorticotropin (ACTH) and growth hormone (GH)-secreting adenomas, while clinically non-functioning adenomas are exceptionally diagnosed before the age of 16. The occurrence of a pituitary tumour in a young individual should also prompt genetic testing in each case, searching for either germline mutations in one of the known genes that may drive inherited/familial PA (such as the multiple endocrine neoplasia type 1 or MEN1 gene, or the aryl hydrocarbon receptor interacting protein or AIP gene), or for a mosaic activating mutation of GNAS as found in the McCune-Albright syndrome.
Collapse
Affiliation(s)
- Dominique Maiter
- Department of Endocrinology and Nutrition, UCLouvain Cliniques universitaires Saint Luc, 1200 Brussels, Belgium
| | - Philippe Chanson
- Université Paris-Saclay, Inserm, Physiologie et Physiopathologie Endocriniennes, Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Service d'Endocrinologie et des Maladies de la Reproduction et Centre de Référence des Maladies Rares de l'Hypophyse HYPO, 94270 Le Kremlin-Bicêtre, France
| | - Stefan Matei Constantinescu
- Department of Endocrinology and Nutrition, UCLouvain Cliniques universitaires Saint Luc, 1200 Brussels, Belgium
| | - Agnès Linglart
- INSERM-U1185, Paris Sud Paris-Saclay University and AP-HP Division of Endocrinology and Diabetes for Children, Bicêtre Paris Sud Hospital, 64 Gabriel Péri Street, 94270 Le Kremlin Bicêtre, France
| |
Collapse
|
5
|
Daly AF, Beckers A. The Genetic Pathophysiology and Clinical Management of the TADopathy, X-Linked Acrogigantism. Endocr Rev 2024; 45:737-754. [PMID: 38696651 DOI: 10.1210/endrev/bnae014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 03/21/2024] [Accepted: 04/29/2024] [Indexed: 05/04/2024]
Abstract
Pituitary gigantism is a rare manifestation of chronic growth hormone (GH) excess that begins before closure of the growth plates. Nearly half of patients with pituitary gigantism have an identifiable genetic cause. X-linked acrogigantism (X-LAG; 10% of pituitary gigantism) typically begins during infancy and can lead to the tallest individuals described. In the 10 years since its discovery, about 40 patients have been identified. Patients with X-LAG usually develop mixed GH and prolactin macroadenomas with occasional hyperplasia that secrete copious amounts of GH, and frequently prolactin. Circulating GH-releasing hormone is also elevated in a proportion of patients. X-LAG is caused by constitutive or sporadic mosaic duplications at chromosome Xq26.3 that disrupt the normal chromatin architecture of a topologically associating domain (TAD) around the orphan G-protein-coupled receptor, GPR101. This leads to the formation of a neo-TAD in which GPR101 overexpression is driven by ectopic enhancers ("TADopathy"). X-LAG has been seen in 3 families due to transmission of the duplication from affected mothers to sons. GPR101 is a constitutively active receptor with an unknown natural ligand that signals via multiple G proteins and protein kinases A and C to promote GH/prolactin hypersecretion. Treatment of X-LAG is challenging due to the young patient population and resistance to somatostatin analogs; the GH receptor antagonist pegvisomant is often an effective option. GH, insulin-like growth factor 1, and prolactin hypersecretion and physical overgrowth can be controlled before definitive adult gigantism occurs, often at the cost of permanent hypopituitarism.
Collapse
Affiliation(s)
- Adrian F Daly
- Department of Endocrinology, Centre Hospitalier Universitaire (CHU) de Liège, University of Liège, Domaine Universitaire Sart Tilman, 4000 Liège, Belgium
| | - Albert Beckers
- Department of Endocrinology, Centre Hospitalier Universitaire (CHU) de Liège, University of Liège, Domaine Universitaire Sart Tilman, 4000 Liège, Belgium
| |
Collapse
|
6
|
Xiang B, Zhang X, Liu W, Mao B, Zhao Y, Wang Y, Gong W, Ye H, Li Y, Zhang Z, Yu Y, He M. Germline AIP variants in sporadic young acromegaly and pituitary gigantism: clinical and genetic insights from a Han Chinese cohort. Endocrine 2024; 85:1346-1356. [PMID: 38851643 DOI: 10.1007/s12020-024-03898-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 05/27/2024] [Indexed: 06/10/2024]
Abstract
PURPOSE Variants in the Aryl hydrocarbon receptor-interacting protein (AIP) gene have been identified in sporadic acromegaly and pituitary gigantism, especially in young patients, with a predisposition to aggressive clinical phenotype and poor treatment efficacy. The clinical characteristics of patients with sporadic acromegaly and pituitary gigantism as well as AIP variants in Han Chinese have been rarely reported. We aimed to identify AIP gene variants and analyze the clinical characteristics of patients with sporadic acromegaly and pituitary gigantism in Han Chinese. METHODS The study included 181 sporadic acromegaly (N = 163) and pituitary gigantism (N = 18) patients with an onset age of no more than 45 years old, who were diagnosed, treated, and followed up in Huashan Hospital. All 6 exons and their flanking regions of the AIP gene were analyzed with Sanger sequencing or NGS. The clinical characteristics were compared between groups with and without AIP variants. RESULTS Germline AIP variants were found in 15/181 (8.29%) cases. In patients with an onset age ≤30 years old, AIP variants were identified in 12/133 (9.02%). Overall, 13 variants were detected. The pathogenic (P) variants p.R304X and p.R81X were identified in four cases, with two instances of each variant. Six exon variants (p.C254R, p.K103fs, p.Q228fs, p.Y38X, p.Q213*, and p.1115 fs) have not been reported before, which were likely pathogenic (LP). Patients with P/LP variants had younger onset ages, a higher prevalence of pituitary gigantism, larger tumor volumes, and a higher percentage of Ki-67-positive cells in tumors. In addition, the group with P/LP variants showed a less significant reduction of GH levels in an acute octreotide suppression test (OST) [17.7% (0, 65.0%) vs. 80.5% (63.9%, 90.2%), P = 0.001], and a trend of less GH decrease after the 3-month treatment with long-acting somatostatin analogs (SSAs). CONCLUSION Germline AIP variants existed in sporadic Chinese Han acromegaly and pituitary gigantism patients and were more likely to be detected in young patients. AIP variants were associated with more aggressive tumor phenotypes and less response to SSA treatment.
Collapse
Affiliation(s)
- Boni Xiang
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, No. 12 Wulumuqi Middle Road, Shanghai, China
| | - Xintong Zhang
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, No. 12 Wulumuqi Middle Road, Shanghai, China
- Department of General Practice, Zhongshan Hospital, Fudan University, No.180 Fenglin Road, Shanghai, China
| | - Wenjuan Liu
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, No. 12 Wulumuqi Middle Road, Shanghai, China
| | - Bei Mao
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, No. 12 Wulumuqi Middle Road, Shanghai, China
| | - Yao Zhao
- Department of Neurosurgery, Huashan Hospital, Fudan University, No. 12 Wulumuqi Middle Road, Shanghai, China
- Shanghai Pituitary Tumor Center, Shanghai, China
| | - Yongfei Wang
- Department of Neurosurgery, Huashan Hospital, Fudan University, No. 12 Wulumuqi Middle Road, Shanghai, China
- Shanghai Pituitary Tumor Center, Shanghai, China
| | - Wei Gong
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, No. 12 Wulumuqi Middle Road, Shanghai, China
| | - Hongying Ye
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, No. 12 Wulumuqi Middle Road, Shanghai, China
- Shanghai Pituitary Tumor Center, Shanghai, China
- Huashan Rare Disease Center, Huashan Hospital Fudan University, Shanghai, China
| | - Yiming Li
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, No. 12 Wulumuqi Middle Road, Shanghai, China
- Shanghai Pituitary Tumor Center, Shanghai, China
- Huashan Rare Disease Center, Huashan Hospital Fudan University, Shanghai, China
| | - Zhaoyun Zhang
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, No. 12 Wulumuqi Middle Road, Shanghai, China
- Shanghai Pituitary Tumor Center, Shanghai, China
- Huashan Rare Disease Center, Huashan Hospital Fudan University, Shanghai, China
| | - Yifei Yu
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, No. 12 Wulumuqi Middle Road, Shanghai, China.
| | - Min He
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, No. 12 Wulumuqi Middle Road, Shanghai, China.
- Shanghai Pituitary Tumor Center, Shanghai, China.
- Huashan Rare Disease Center, Huashan Hospital Fudan University, Shanghai, China.
| |
Collapse
|
7
|
Veleno M, Giampietro A, Raia S, Menotti S, Tartaglione T, Gaudino S, Doglietto F, DE Marinis L, Pontecorvi A, Chiloiro S, Bianchi A. Clinical implications of the 2022 WHO classification on the multidisciplinary management of PitNETS patients. Minerva Endocrinol (Torino) 2024; 49:269-282. [PMID: 38963295 DOI: 10.23736/s2724-6507.24.04126-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/05/2024]
Abstract
The review explores the 2022 update to the World Health Organization (WHO) classification of pituitary adenomas, now referred to as pituitary neuroendocrine tumors (PitNETs), and his possible impact on the clinical management of PitNET patients. The review highlights the differences and the evolution from the 2017 to 2022 version, with the current classification considering the lineage of the tumor cells, cell type, hormones produced, and other auxiliary characteristics for a comprehensive histological classification. The revision in terminology reflects a broader perspective on neuroendocrine neoplasia. The new approach based on transcription factors, hormone expression and other biomarkers has allowed a major revision of the nomenclature and a more accurate classification of pituitary adenomas. Furthermore, in some cases this approach is also assuming a prognostic value, useful in clinical practice. However, despite this elaborate classification and stratification, the review points out the lack of a robust grading or staging system and suggests the need for further research and validation of diagnostic methods. Despite these limitations, the revised classification presents a significant step towards understanding and managing PitNETs patients.
Collapse
Affiliation(s)
- Miriam Veleno
- Pituitary Unit, Department of Internal Medicine, Endocrinology and Diabetes, IRCCS A. Gemelli University Polyclinic Foundation, Rome, Italy
- Department of Translational Medicine and Surgery, Sacred Heart Catholic University, Rome, Italy
| | - Antonella Giampietro
- Pituitary Unit, Department of Internal Medicine, Endocrinology and Diabetes, IRCCS A. Gemelli University Polyclinic Foundation, Rome, Italy
- Department of Translational Medicine and Surgery, Sacred Heart Catholic University, Rome, Italy
| | - Salvatore Raia
- Pituitary Unit, Department of Internal Medicine, Endocrinology and Diabetes, IRCCS A. Gemelli University Polyclinic Foundation, Rome, Italy
- Department of Translational Medicine and Surgery, Sacred Heart Catholic University, Rome, Italy
| | - Sara Menotti
- Pituitary Unit, Department of Internal Medicine, Endocrinology and Diabetes, IRCCS A. Gemelli University Polyclinic Foundation, Rome, Italy
- Department of Translational Medicine and Surgery, Sacred Heart Catholic University, Rome, Italy
| | - Tommaso Tartaglione
- Department of Translational Medicine and Surgery, Sacred Heart Catholic University, Rome, Italy
- Department of Radiodiagnostic, IRCCS A. Gemelli University Polyclinic Foundation, Rome, Italy
| | - Simona Gaudino
- Department of Translational Medicine and Surgery, Sacred Heart Catholic University, Rome, Italy
- Department of Radiodiagnostic, IRCCS A. Gemelli University Polyclinic Foundation, Rome, Italy
| | - Francesco Doglietto
- Department of Translational Medicine and Surgery, Sacred Heart Catholic University, Rome, Italy
- Department of Neurosurgery, IRCCS A. Gemelli University Polyclinic Foundation, Rome, Italy
| | - Laura DE Marinis
- Pituitary Unit, Department of Internal Medicine, Endocrinology and Diabetes, IRCCS A. Gemelli University Polyclinic Foundation, Rome, Italy
- Department of Translational Medicine and Surgery, Sacred Heart Catholic University, Rome, Italy
| | - Alfredo Pontecorvi
- Pituitary Unit, Department of Internal Medicine, Endocrinology and Diabetes, IRCCS A. Gemelli University Polyclinic Foundation, Rome, Italy
- Department of Translational Medicine and Surgery, Sacred Heart Catholic University, Rome, Italy
| | - Sabrina Chiloiro
- Pituitary Unit, Department of Internal Medicine, Endocrinology and Diabetes, IRCCS A. Gemelli University Polyclinic Foundation, Rome, Italy -
- Department of Translational Medicine and Surgery, Sacred Heart Catholic University, Rome, Italy
| | - Antonio Bianchi
- Pituitary Unit, Department of Internal Medicine, Endocrinology and Diabetes, IRCCS A. Gemelli University Polyclinic Foundation, Rome, Italy
- Department of Translational Medicine and Surgery, Sacred Heart Catholic University, Rome, Italy
| |
Collapse
|
8
|
Zhang X, Chen Y, Yu Y, Li J. Diagnosis and Management of Aggressive/Refractory Growth Hormone-Secreting Pituitary Neuroendocrine Tumors. Int J Endocrinol 2024; 2024:5085905. [PMID: 39224564 PMCID: PMC11368557 DOI: 10.1155/2024/5085905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 07/16/2024] [Accepted: 08/07/2024] [Indexed: 09/04/2024] Open
Abstract
The majority of acromegaly and gigantism are caused by growth hormone-secreting pituitary neuroendocrine tumors (PitNETs). Most cases can be cured or controlled by surgery, medical therapy, and/or radiotherapy. However, a few of these tumors are resistant to traditional therapy and always have a poor prognosis. The title aggressive/refractory is used to differentiate them from pituitary carcinomas. To date, there is no definitive conclusion on how to diagnose aggressive/refractory growth hormone-secreting PitNETs, which may have slowed the process of exploring new therapeutical strategies. We summarized the literature described diagnosis and treatment of the disease. Potential disease markers and prospective therapies were also included.
Collapse
Affiliation(s)
- Xiaojuan Zhang
- Department of Endocrinology and MetabolismWest China Hospital of Sichuan University, Chengdu, China
| | - Yu Chen
- Department of Endocrinology and MetabolismWest China Hospital of Sichuan University, Chengdu, China
| | - Yerong Yu
- Department of Endocrinology and MetabolismWest China Hospital of Sichuan University, Chengdu, China
| | - Jianwei Li
- Department of Endocrinology and MetabolismWest China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
9
|
Balinisteanu I, Caba L, Florea A, Popescu R, Florea L, Ungureanu MC, Leustean L, Gorduza EV, Preda C. Unlocking the Genetic Secrets of Acromegaly: Exploring the Role of Genetics in a Rare Disorder. Curr Issues Mol Biol 2024; 46:9093-9121. [PMID: 39194755 DOI: 10.3390/cimb46080538] [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: 07/16/2024] [Revised: 08/14/2024] [Accepted: 08/18/2024] [Indexed: 08/29/2024] Open
Abstract
Acromegaly is a rare endocrine disorder characterized by the excessive production of growth hormone (GH) in adulthood. Currently, it is understood that certain pituitary neuroendocrine tumors (PitNETs) exhibit a hereditary predisposition. These tumors' genetic patterns fall into two categories: isolated and syndromic tumors. The isolated forms are characterized by molecular defects that predispose exclusively to PitNETs, including familial isolated pituitary adenomas (FIPAs) and sporadic genetic defects not characterized by hereditary predisposition. All the categories involve either germline or somatic mutations, or both, each associated with varying levels of penetrance and different phenotypes. This highlights the importance of genetic testing and the need for a more comprehensive view of the whole disease. Despite the availability of multiple treatment options, diagnosis often occurs after several years, and management is still difficult. Early detection and intervention are crucial for preventing complications and enhancing the quality of life for affected individuals. This review aims to elucidate the molecular, clinical, and histological characteristics of GH-secreting PitNETs, providing insights into their prevalence, treatment nuances, and the benefits of genetic testing for each type of genetic disorder associated with acromegaly.
Collapse
Affiliation(s)
- Ioana Balinisteanu
- Endocrinology Department, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Lavinia Caba
- Medical Genetics Department, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Andreea Florea
- Medical Genetics Department, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Roxana Popescu
- Medical Genetics Department, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Laura Florea
- Nephrology-Internal Medicine Department, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Maria-Christina Ungureanu
- Endocrinology Department, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Letitia Leustean
- Endocrinology Department, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Eusebiu Vlad Gorduza
- Medical Genetics Department, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Cristina Preda
- Endocrinology Department, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| |
Collapse
|
10
|
Poiană IR, Burcea IF, Pițuru SM, Bucur A. Cone Beam Computed Tomography Panoramic Mandibular Indices in the Screening of Postmenopausal Women with Low Bone Mass: Correlations with Bone Quantity and Quality. Dent J (Basel) 2024; 12:256. [PMID: 39195100 DOI: 10.3390/dj12080256] [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: 06/17/2024] [Revised: 08/03/2024] [Accepted: 08/09/2024] [Indexed: 08/29/2024] Open
Abstract
OBJECTIVE This study examined the potential use of computed tomography panoramic mandibular indices on cone beam CT (CBCT) for assessing bone density in postmenopausal women with low bone mass. STUDY DESIGN The study enrolled 104 postmenopausal women who underwent dual-energy X-ray absorptiometry (DXA) using a DXA scanner and mental foramen region CBCT alongside the NewTom VGi EVO Cone Beam 3D system. We assessed the relationship between the following DXA parameters: lumbar, femoral neck, and total hip T score, bone mineral density (BMD), and lumbar trabecular bone score (TBS). The following panoramic mandibular indices were also considered: the computed tomography mandibular index superior (CTI(S)), computed tomography mandibular index inferior (CTI(I)), and computed tomography mental index (CTMI). RESULTS The study revealed moderate correlations between CBCT indices and BMD/TBS scores: CTMI showed the highest correlation with the femoral neck T-score (r = 0.551, p < 0.0001). TBS scores were also moderately correlated with CBCT indices: CTMI showed a moderate positive correlation with TBS (r = 0.431, p < 0.0001); CTI(S) had a similar moderate positive correlation with TBS (r = 0.421, p < 0.0001). AUC values ranged from 0.697 to 0.733 for osteoporosis versus the osteopenia/normal group and from 0.734 to 0.744 for low versus normal bone quality groups, p < 0.0001. The comparison of the values of the studied indices between low versus normal bone quality (quantified with TBS) groups showed high sensitivity but low specificity. CONCLUSIONS CBCT-measured indices CTI(S), CTI(I), and CTMI are useful in assessing patients with low bone mass to improve, by specific treatment, the prognosis of dental implants.
Collapse
Affiliation(s)
- Ioana Ruxandra Poiană
- Faculty of Dentistry, "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Iulia Florentina Burcea
- Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Endocrinology, National Institute of Endocrinology C. I. Parhon, 011853 Bucharest, Romania
| | - Silviu-Mirel Pițuru
- Faculty of Dentistry, "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Alexandru Bucur
- Faculty of Dentistry, "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
| |
Collapse
|
11
|
Bhat SZ, Salvatori R. Current role of pasireotide in the treatment of acromegaly. Best Pract Res Clin Endocrinol Metab 2024; 38:101875. [PMID: 38290866 DOI: 10.1016/j.beem.2024.101875] [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: 02/01/2024]
Abstract
"First-generation" somatostatin receptor agonists (SSTRAs) octreotide and lanreotide are the most commonly used first-line pharmacological therapy for patients with acromegaly. A subset of patients respond only partially or not at all to the first-generation SSTRA, necessitating the use of additional pharmacological agents or other modes of therapy. Pasireotide is a "second-generation" SSTRA that has multi-receptor activity. Prospective studies have shown promise in the use of pasireotide in patients with poor response to first-generation SSTRA. Here we elucidate the molecular pathways of resistance to first-generation SSTRA, the mechanism of action, pre-clinical and clinical evidence of the use of pasireotide in patients having incomplete / lack of response to first-generation SSTRA. We also discuss the clinical, pathological, and radiological markers predicting response to pasireotide, and the difference in side-effect profiles of pasireotide, compared to first-generation SSTRA.
Collapse
Affiliation(s)
- Salman Zahoor Bhat
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; Tidal Health Endocrinology, Salisbury, MD, USA.
| | - Roberto Salvatori
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
12
|
Remba-Shapiro I, Nachtigall LB. Treatment of acromegaly with oral octreotide. Best Pract Res Clin Endocrinol Metab 2024; 38:101888. [PMID: 38443224 DOI: 10.1016/j.beem.2024.101888] [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: 03/07/2024]
Abstract
Acromegaly is a rare disease caused by a growth hormone excess, usually due to a secreting pituitary adenoma. Somatostatin receptor ligands (SRL) are the mainstay of medical therapy for patients with acromegaly who fail to achieve biochemical control post-operatively or are not eligible for surgical treatment. SRLs are typically administered as monthly injections and have shown to be effective in maintaining biochemical and radiological control of acromegaly. However, these injections may cause local adverse events and are associated with increased psychological burden in some patients. Oral octreotide provides a new alternative for patients responding to injectable SRLs. This new formulation has shown to have similar safety and efficacy profiles compared to injectable SRLs and may be a preferable option for some patients with acromegaly. The aim of this review is to provide an overview of the role of oral octreotide in the management of acromegaly.
Collapse
Affiliation(s)
- Ilan Remba-Shapiro
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Lisa B Nachtigall
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
13
|
Korbonits M, Blair JC, Boguslawska A, Ayuk J, Davies JH, Druce MR, Evanson J, Flanagan D, Glynn N, Higham CE, Jacques TS, Sinha S, Simmons I, Thorp N, Swords FM, Storr HL, Spoudeas HA. Consensus guideline for the diagnosis and management of pituitary adenomas in childhood and adolescence: Part 2, specific diseases. Nat Rev Endocrinol 2024; 20:290-309. [PMID: 38336898 DOI: 10.1038/s41574-023-00949-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/19/2023] [Indexed: 02/12/2024]
Abstract
Pituitary adenomas are rare in children and young people under the age of 19 (hereafter referred to as CYP) but they pose some different diagnostic and management challenges in this age group than in adults. These rare neoplasms can disrupt maturational, visual, intellectual and developmental processes and, in CYP, they tend to have more occult presentation, aggressive behaviour and are more likely to have a genetic basis than in adults. Through standardized AGREE II methodology, literature review and Delphi consensus, a multidisciplinary expert group developed 74 pragmatic management recommendations aimed at optimizing care for CYP in the first-ever comprehensive consensus guideline to cover the care of CYP with pituitary adenoma. Part 2 of this consensus guideline details 57 recommendations for paediatric patients with prolactinomas, Cushing disease, growth hormone excess causing gigantism and acromegaly, clinically non-functioning adenomas, and the rare TSHomas. Compared with adult patients with pituitary adenomas, we highlight that, in the CYP group, there is a greater proportion of functioning tumours, including macroprolactinomas, greater likelihood of underlying genetic disease, more corticotrophinomas in boys aged under 10 years than in girls and difficulty of peri-pubertal diagnosis of growth hormone excess. Collaboration with pituitary specialists caring for adult patients, as part of commissioned and centralized multidisciplinary teams, is key for optimizing management, transition and lifelong care and facilitates the collection of health-related quality of survival outcomes of novel medical, surgical and radiotherapeutic treatments, which are currently largely missing.
Collapse
Affiliation(s)
- 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.
| | | | - Anna Boguslawska
- Department of Endocrinology, Jagiellonian University Medical College, Krakow, Poland
| | - John Ayuk
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Justin H Davies
- University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Maralyn R Druce
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Jane Evanson
- Neuroradiology, Barts Health NHS Trust, London, UK
| | | | - Nigel Glynn
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | | | - Thomas S Jacques
- Great Ormond Street Institute of Child Health, University College London, London, UK
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Saurabh Sinha
- Sheffield Children's and Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Ian Simmons
- The Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Nicky Thorp
- The Christie NHS Foundation Trust, Manchester, UK
| | | | - Helen L Storr
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Helen A Spoudeas
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
- University College London Hospitals NHS Foundation Trust, London, UK
| |
Collapse
|
14
|
Korbonits M, Blair JC, Boguslawska A, Ayuk J, Davies JH, Druce MR, Evanson J, Flanagan D, Glynn N, Higham CE, Jacques TS, Sinha S, Simmons I, Thorp N, Swords FM, Storr HL, Spoudeas HA. Consensus guideline for the diagnosis and management of pituitary adenomas in childhood and adolescence: Part 1, general recommendations. Nat Rev Endocrinol 2024; 20:278-289. [PMID: 38336897 DOI: 10.1038/s41574-023-00948-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/19/2023] [Indexed: 02/12/2024]
Abstract
Tumours of the anterior part of the pituitary gland represent just 1% of all childhood (aged <15 years) intracranial neoplasms, yet they can confer high morbidity and little evidence and guidance is in place for their management. Between 2014 and 2022, a multidisciplinary expert group systematically developed the first comprehensive clinical practice consensus guideline for children and young people under the age 19 years (hereafter referred to as CYP) presenting with a suspected pituitary adenoma to inform specialist care and improve health outcomes. Through robust literature searches and a Delphi consensus exercise with an international Delphi consensus panel of experts, the available scientific evidence and expert opinions were consolidated into 74 recommendations. Part 1 of this consensus guideline includes 17 pragmatic management recommendations related to clinical care, neuroimaging, visual assessment, histopathology, genetics, pituitary surgery and radiotherapy. While in many aspects the care for CYP is similar to that of adults, key differences exist, particularly in aetiology and presentation. CYP with suspected pituitary adenomas require careful clinical examination, appropriate hormonal work-up, dedicated pituitary imaging and visual assessment. Consideration should be given to the potential for syndromic disease and genetic assessment. Multidisciplinary discussion at both the local and national levels can be key for management. Surgery should be performed in specialist centres. The collection of outcome data on novel modalities of medical treatment, surgical intervention and radiotherapy is essential for optimal future treatment.
Collapse
Affiliation(s)
- 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.
| | | | - Anna Boguslawska
- Department of Endocrinology, Jagiellonian University Medical College, Krakow, Poland
| | - John Ayuk
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Justin H Davies
- University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Maralyn R Druce
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Jane Evanson
- Neuroradiology, Barts Health NHS Trust, London, UK
| | | | - Nigel Glynn
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | | | - Thomas S Jacques
- Great Ormond Street Institute of Child Health, University College London, London, UK
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Saurabh Sinha
- Sheffield Children's and Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Ian Simmons
- The Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Nicky Thorp
- The Christie NHS Foundation Trust, Manchester, UK
| | | | - Helen L Storr
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Helen A Spoudeas
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
- University College London Hospitals NHS Foundation Trust, London, UK
| |
Collapse
|
15
|
Ramírez-Rentería C, Hernández-Ramírez LC. Genetic diagnosis in acromegaly and gigantism: From research to clinical practice. Best Pract Res Clin Endocrinol Metab 2024; 38:101892. [PMID: 38521632 DOI: 10.1016/j.beem.2024.101892] [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: 03/25/2024]
Abstract
It is usually considered that only 5% of all pituitary neuroendocrine tumours are due to inheritable causes. Since this estimate was reported, however, multiple genetic defects driving syndromic and nonsyndromic somatotrophinomas have been unveiled. This heterogeneous genetic background results in overlapping phenotypes of GH excess. Genetic tests should be part of the approach to patients with acromegaly and gigantism because they can refine the clinical diagnoses, opening the possibility to tailor the clinical conduct to each patient. Even more, genetic testing and clinical screening of at-risk individuals have a positive impact on disease outcomes, by allowing for the timely detection and treatment of somatotrophinomas at early stages. Future research should focus on determining the actual frequency of novel genetic drivers of somatotrophinomas in the general population, developing up-to-date disease-specific multi-gene panels for clinical use, and finding strategies to improve access to modern genetic testing worldwide.
Collapse
Affiliation(s)
- Claudia Ramírez-Rentería
- Unidad de Investigación Médica en Enfermedades Endocrinas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Laura C Hernández-Ramírez
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México, e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.
| |
Collapse
|
16
|
Shen AL, Moran SM, Glover EN, Lin BC, Carney PR, Bradfield CA. Familial isolated pituitary adenoma is independent of Ahr genotype in a novel mouse model of disease. Heliyon 2024; 10:e28231. [PMID: 38590848 PMCID: PMC10999881 DOI: 10.1016/j.heliyon.2024.e28231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 03/13/2024] [Accepted: 03/13/2024] [Indexed: 04/10/2024] Open
Abstract
Human familial isolated pituitary adenoma (FIPA) has been linked to germline heterozygous mutations in the gene encoding the aryl hydrocarbon receptor-interacting protein (AIP, also known as ARA9, XAP2, FKBP16, or FKBP37). To investigate the hypothesis that AIP is a pituitary adenoma tumor suppressor via its role in aryl hydrocarbon receptor (AHR) signaling, we have compared the pituitary phenotype of our global null Aip (AipΔC) mouse model with that of a conditional null Aip model (Aipfx/fx) carrying the same deletion, as well as pituitary phenotypes of Ahr global null and Arnt conditional null animals. We demonstrate that germline AipΔC heterozygosity results in a high incidence of pituitary tumors in both sexes, primarily somatotropinomas, at 16 months of age. Biallelic deletion of Aip in Pit-1 cells (Aipfx/fx:rGHRHRcre) increased pituitary tumor incidence and also accelerated tumor progression, supporting a loss-of-function/loss-of-heterozygosity model of tumorigenesis. Tumor development exhibited sexual dimorphism in wildtype and Aipfx/fx:rGHRHRcre animals. Despite the role of AHR as a tumor suppressor in other cancers, the observation that animals lacking AHR in all tissues, or ARNT in Pit-1 cells, do not develop somatotropinomas argues against the hypothesis that pituitary tumorigenesis in AIP-associated FIPA is related to decreased activities of either the Ahr or Arnt gene products.
Collapse
Affiliation(s)
- Anna L Shen
- The McArdle Laboratory for Cancer Research, Department of Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53706, United States
| | - Susan M Moran
- The McArdle Laboratory for Cancer Research, Department of Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53706, United States
| | - Edward N Glover
- The McArdle Laboratory for Cancer Research, Department of Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53706, United States
| | - Bernice C Lin
- The McArdle Laboratory for Cancer Research, Department of Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53706, United States
- Current address, Lin-Zhi International, 2945, Oakmead Village Court, Santa Clara, CA, 95051, United States
| | - Patrick R Carney
- The McArdle Laboratory for Cancer Research, Department of Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53706, United States
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53706, United States
| | - Christopher A Bradfield
- The McArdle Laboratory for Cancer Research, Department of Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53706, United States
| |
Collapse
|
17
|
Alzahrani AS, Bin Nafisah A, Alswailem M, Alghamdi B, Alsaihati B, Aljafar H, Baz B, Alhindi H, Moria Y, Butt MI, Alkabbani AG, Alshaikh OM, Alnassar A, Bin Afeef A, AlQuraa R, Alsuhaibani R, Alhadlaq O, Abothenain F, Altwaijry YA. Germline Variants in Sporadic Pituitary Adenomas. J Endocr Soc 2024; 8:bvae085. [PMID: 38745824 PMCID: PMC11091836 DOI: 10.1210/jendso/bvae085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Indexed: 05/16/2024] Open
Abstract
Context Data on germline genetics of pituitary adenomas (PAs) using whole-exome sequencing (WES) are limited. Objective This study investigated the germline genetic variants in patients with PAs using WES. Methods We studied 134 consecutive functioning (80.6%) and nonfunctioning (19.4%) PAs in 61 female (45.5%) and 73 male patients (54.5%). Their median age was 34 years (range, 11-85 years) and 31 patients had microadenomas (23.0%) and 103 macroadenomas (77%). None of these patients had family history of PA or a known PA-associated syndrome. Peripheral blood DNA was isolated and whole-exome sequenced. We used American College of Medical Genetics and Genomics (ACMG) criteria and a number of in silico analysis tools to characterize genetic variant pathogenicity levels and focused on previously reported PA-associated genes. Results We identified 35 variants of unknown significance (VUS) in 17 PA-associated genes occurring in 40 patients (29.8%). Although designated VUS by the strict ACGM criteria, they are predicted to be pathogenic by in silico analyses and their extremely low frequencies in 1000 genome, gnomAD, and the Saudi Genome Project databases. Further analysis of these variants by the Alpha Missense analysis tool yielded 8 likely pathogenic variants in 9 patients in the following genes: AIP:c.767C>T (p.S256F), CDH23:c.906G>C (p.E302D), CDH23:c.1096G>A (p.A366T), DICER1:c.620C>T (p.A207V), MLH1:c.955G>A (p.E319K), MSH2:c.148G>A (p.A50T), SDHA:c.869T>C (p.L290P) and USP48 (2 patients): c.2233G>A (p.V745M). Conclusion This study suggests that about 6.7% of patients with apparently sporadic PAs carry likely pathogenic variants in PA-associated genes. These findings need further studies to confirm them.
Collapse
Affiliation(s)
- Ali S Alzahrani
- Department of Medicine, King Faisal Specialist Hospital & Research Center, P.O. Box 3354, Riyadh 11211, Saudi Arabia
- Department of Molecular Oncology, King Faisal Specialist Hospital & Research Center, P.O. Box 3354, Riyadh 11211, Saudi Arabia
| | - Abdulghani Bin Nafisah
- Department of Molecular Oncology, King Faisal Specialist Hospital & Research Center, P.O. Box 3354, Riyadh 11211, Saudi Arabia
| | - Meshael Alswailem
- Department of Molecular Oncology, King Faisal Specialist Hospital & Research Center, P.O. Box 3354, Riyadh 11211, Saudi Arabia
| | - Balgees Alghamdi
- Department of Molecular Oncology, King Faisal Specialist Hospital & Research Center, P.O. Box 3354, Riyadh 11211, Saudi Arabia
| | - Burair Alsaihati
- Applied Genomic Technologies Institute, King Abdulaziz City for Science and Technology, P.O. Box 6086, Riyadh 11442, Saudi Arabia
| | - Hussain Aljafar
- Applied Genomic Technologies Institute, King Abdulaziz City for Science and Technology, P.O. Box 6086, Riyadh 11442, Saudi Arabia
| | - Batoul Baz
- Health and Wellness Sector, King Abdulaziz City for Science and Technology, P.O. Box 6086, Riyadh 11442, Saudi Arabia
| | - Hindi Alhindi
- Department of Pathology and Laboratory Medicine, King Faisal Specialist Hospital & Research Center, P.O. Box 3354, Riyadh 11211, Saudi Arabia
| | - Yosra Moria
- Department of Medicine, King Faisal Specialist Hospital & Research Center, P.O. Box 3354, Riyadh 11211, Saudi Arabia
| | - Muhammad Imran Butt
- Department of Medicine, King Faisal Specialist Hospital & Research Center, P.O. Box 3354, Riyadh 11211, Saudi Arabia
| | | | | | - Anhar Alnassar
- Department of Medicine, King Faisal Specialist Hospital & Research Center, P.O. Box 3354, Riyadh 11211, Saudi Arabia
| | - Ahmed Bin Afeef
- Department of Medicine, King Faisal Specialist Hospital & Research Center, P.O. Box 3354, Riyadh 11211, Saudi Arabia
| | - Reem AlQuraa
- Department of Medicine, King Faisal Specialist Hospital & Research Center, P.O. Box 3354, Riyadh 11211, Saudi Arabia
| | - Rawan Alsuhaibani
- Department of Medicine, King Faisal Specialist Hospital & Research Center, P.O. Box 3354, Riyadh 11211, Saudi Arabia
| | - Omar Alhadlaq
- Department of Medicine, King Faisal Specialist Hospital & Research Center, P.O. Box 3354, Riyadh 11211, Saudi Arabia
| | - Fayha Abothenain
- Department of Medicine, King Faisal Specialist Hospital & Research Center, P.O. Box 3354, Riyadh 11211, Saudi Arabia
| | - Yasser A Altwaijry
- Department of Medicine, King Faisal Specialist Hospital & Research Center, P.O. Box 3354, Riyadh 11211, Saudi Arabia
| |
Collapse
|
18
|
Kazzaz SA, Tawil J, Harhaj EW. The aryl hydrocarbon receptor-interacting protein in cancer and immunity: Beyond a chaperone protein for the dioxin receptor. J Biol Chem 2024; 300:107157. [PMID: 38479600 PMCID: PMC11002312 DOI: 10.1016/j.jbc.2024.107157] [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: 01/09/2024] [Revised: 03/04/2024] [Accepted: 03/06/2024] [Indexed: 04/04/2024] Open
Abstract
The aryl hydrocarbon receptor (AhR)-interacting protein (AIP) is a ubiquitously expressed, immunophilin-like protein best known for its role as a co-chaperone in the AhR-AIP-Hsp90 cytoplasmic complex. In addition to regulating AhR and the xenobiotic response, AIP has been linked to various aspects of cancer and immunity that will be the focus of this review article. Loss-of-function AIP mutations are associated with pituitary adenomas, suggesting that AIP acts as a tumor suppressor in the pituitary gland. However, the tumor suppressor mechanisms of AIP remain unclear, and AIP can exert oncogenic functions in other tissues. While global deletion of AIP in mice yields embryonically lethal cardiac malformations, heterozygote, and tissue-specific conditional AIP knockout mice have revealed various physiological roles of AIP. Emerging studies have established the regulatory roles of AIP in both innate and adaptive immunity. AIP interacts with and inhibits the nuclear translocation of the transcription factor IRF7 to inhibit type I interferon production. AIP also interacts with the CARMA1-BCL10-MALT1 complex in T cells to enhance IKK/NF-κB signaling and T cell activation. Taken together, AIP has diverse functions that vary considerably depending on the client protein, the tissue, and the species.
Collapse
Affiliation(s)
- Sarah A Kazzaz
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, Pennsylvania, USA; Medical Scientist Training Program, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - John Tawil
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Edward W Harhaj
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, Pennsylvania, USA.
| |
Collapse
|
19
|
Sada V, Puliani G, Feola T, Pirchio R, Pofi R, Sesti F, De Alcubierre D, Amodeo ME, D'Aniello F, Vincenzi L, Gianfrilli D, Isidori AM, Grossman AB, Sbardella E. Tall stature and gigantism in transition age: clinical and genetic aspects-a literature review and recommendations. J Endocrinol Invest 2024; 47:777-793. [PMID: 37891382 DOI: 10.1007/s40618-023-02223-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023]
Abstract
PURPOSE Tall stature is defined as height greater than the threshold of more than 2 standard deviations above the average population height for age, sex, and ethnicity. Many studies have described the main aspects of this condition during puberty, but an analysis of the characteristics that the physician should consider in the differential diagnosis of gigantism-tall stature secondary to a pituitary tumour-during the transition age (15-25 years) is still lacking. METHODS A comprehensive search of English-language original articles was conducted in the MEDLINE database (December 2021-March 2022). We selected all studies regarding epidemiology, genetic aspects, and the diagnosis of tall stature and gigantism during the transition age. RESULTS Generally, referrals for tall stature are not as frequent as expected because most cases are familial and are usually unreported by parents and patients to endocrinologists. For this reason, lacking such experience of tall stature, familiarity with many rarer overgrowth syndromes is essential. In the transition age, it is important but challenging to distinguish adolescents with high constitutional stature from those with gigantism. Pituitary gigantism is a rare disease in the transition age, but its systemic complications are very relevant for future health. Endocrine evaluation is crucial for identifying conditions that require hormonal treatment so that they can be treated early to improve the quality of life and prevent comorbidities of individual patient in this age range. CONCLUSION The aim of our review is to provide a practical clinical approach to recognise adolescents, potentially affected by gigantism, as early as possible.
Collapse
Affiliation(s)
- V Sada
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - G Puliani
- Oncological Endocrinology Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - T Feola
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
- Neuroendocrinology, Neuromed Institute, IRCCS, Pozzilli, Italy
| | - R Pirchio
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Degli Studi di Napoli "Federico II", Naples, Italy
| | - R Pofi
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, Churchill Hospital, Oxford University Hospitals, NHS Trust, Oxford, UK
| | - F Sesti
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - D De Alcubierre
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - M E Amodeo
- Dipartimento Pediatrico Universitario Ospedaliero, Bambino Gesù Children Hospital, Rome, Italy
| | - F D'Aniello
- Dipartimento Pediatrico Universitario Ospedaliero, Bambino Gesù Children Hospital, Rome, Italy
| | - L Vincenzi
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - D Gianfrilli
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - A M Isidori
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
- Centre for Rare Diseases (ENDO-ERN Accredited), Policlinico Umberto I, Rome, Italy
| | - A B Grossman
- Green Templeton College, University of Oxford, Oxford, UK
- Centre for Endocrinology, Barts and the London School of Medicine, London, UK
| | - E Sbardella
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy.
| |
Collapse
|
20
|
Nguyen JTT, Ferrière A, Tabarin A. Case report: Complete restoration of the HPA axis function in Cushing's disease with drug treatment. Front Endocrinol (Lausanne) 2024; 15:1337741. [PMID: 38390203 PMCID: PMC10882091 DOI: 10.3389/fendo.2024.1337741] [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: 11/13/2023] [Accepted: 01/19/2024] [Indexed: 02/24/2024] Open
Abstract
This report describes a rare case of a 20-year-old man with an ACTH- and prolactin-secreting invasive pituitary macroadenoma causing hyperprolactinemia and Cushing's disease. He was later found to have an AIP mutation. Treatment with cabergoline (1.5 mg weekly) normalized prolactin concentrations and induced a major shrinkage of the adenoma. Not only was urinary free cortisol normalized for more than 14 years, but also the treatment induced normal hypothalamo-pituitary-adrenal (HPA) axis function as illustrated by the reappearance of a normal cortisol/ACTH circadian rhythm, cortisol suppression to dexamethasone, and disappearance of the excessive and aberrant responses to CRH and desmopressin, respectively. This case is the first description of complete restoration of the physiological characteristics of the HPA axis by a medication during the treatment of Cushing's disease. Although exceptional, it illustrates that drugs targeting the pituitary adenoma can bring true complete remission of Cushing's disease.
Collapse
Affiliation(s)
- Joanne Thanh-Tâm Nguyen
- Service d’Endocrinologie, Diabétologie et Maladies Métaboliques, Centre Hospitalier Universitaire (CHU) de Bordeaux, Pessac, France
| | - Amandine Ferrière
- Service d’Endocrinologie, Diabétologie et Maladies Métaboliques, Centre Hospitalier Universitaire (CHU) de Bordeaux, Pessac, France
| | - Antoine Tabarin
- INSERM (Institut National de la Santé et de la Recherche Médicale) and University of Bordeaux, Neurocentre Magendie, Service d’Endocrinologie, Diabétologie et Maladies Métaboliques, Centre Hospitalier Universitaire (CHU) de Bordeaux, Pessac, France
| |
Collapse
|
21
|
Dzialach L, Sobolewska J, Zak Z, Respondek W, Witek P. Prolactin-secreting pituitary adenomas: male-specific differences in pathogenesis, clinical presentation and treatment. Front Endocrinol (Lausanne) 2024; 15:1338345. [PMID: 38370355 PMCID: PMC10870150 DOI: 10.3389/fendo.2024.1338345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 01/22/2024] [Indexed: 02/20/2024] Open
Abstract
Prolactinomas (PRLomas) constitute approximately half of all pituitary adenomas and approximately one-fifth of them are diagnosed in males. The clinical presentation of PRLomas results from direct prolactin (PRL) action, duration and severity of hyperprolactinemia, and tumor mass effect. Male PRLomas, compared to females, tend to be larger and more invasive, are associated with higher PRL concentration at diagnosis, present higher proliferative potential, are more frequently resistant to standard pharmacotherapy, and thus may require multimodal approach, including surgical resection, radiotherapy, and alternative medical agents. Therefore, the management of PRLomas in men is challenging in many cases. Additionally, hyperprolactinemia is associated with a significant negative impact on men's health, including sexual function and fertility potential, bone health, cardiovascular and metabolic complications, leading to decreased quality of life. In this review, we highlight the differences in pathogenesis, clinical presentation and treatment of PRLomas concerning the male sex.
Collapse
Affiliation(s)
- Lukasz Dzialach
- Department of Internal Medicine, Endocrinology and Diabetes, Medical University of Warsaw, Warsaw, Poland
| | - Joanna Sobolewska
- Department of Internal Medicine, Endocrinology and Diabetes, Medical University of Warsaw, Warsaw, Poland
| | - Zuzanna Zak
- Department of Internal Medicine, Endocrinology and Diabetes, Medical University of Warsaw, Warsaw, Poland
| | - Wioleta Respondek
- Department of Internal Medicine, Endocrinology and Diabetes, Mazovian Brodnowski Hospital, Warsaw, Poland
| | - Przemysław Witek
- Department of Internal Medicine, Endocrinology and Diabetes, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
22
|
Urai S, Yamamoto M, Yamamoto N, Suzuki M, Shichi H, Kanie K, Fujita Y, Bando H, Fukuoka H, Takahashi M, Iguchi G, Takahashi Y, Ogawa W. Newer parameters of the octreotide test in patients with acromegaly. Pituitary 2024; 27:33-43. [PMID: 37999819 DOI: 10.1007/s11102-023-01362-z] [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] [Accepted: 10/23/2023] [Indexed: 11/25/2023]
Abstract
PURPOSE Predicting the therapeutic effects of first-generation somatostatin receptor ligands (fg-SRLs) is important when assessing or planning effective treatment strategies in patients with acromegaly. The oft-used maximum growth hormone (GH) suppression rate parameter of the octreotide test has a suboptimal predictive value. Therefore, this study explored newer parameters of the octreotide test for predicting the therapeutic effect of long-acting fg-SRLs. METHODS In this single-center retrospective study, the octreotide test parameters and the therapeutic effects of fg-SRL at 3 months were investigated in 45 consecutive treatment-naïve patients with acromegaly between April 2008 and March 2023. Additionally, the relationship between the octreotide test parameters and the therapeutic effects of fg-SRLs was investigated. Tumor shrinkage was evaluated based on changes in the longitudinal diameter of the macroadenomas. The area GH suppression rate-time under the curve (AUC) and the time to nadir GH level were calculated and compared with the maximum GH suppression rate. RESULTS The AUC estimated reductions in serum insulin-like growth factor I, and tumor shrinkage. The time to nadir GH level predicted tumor shrinkage more robustly than the maximum GH suppression rate in patients with macroadenoma. CONCLUSION The AUC and time to nadir GH level may potentially be newer parameters of the octreotide test for estimating the therapeutic effect of fg-SRLs.
Collapse
Affiliation(s)
- Shin Urai
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan
| | - Masaaki Yamamoto
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan.
| | - Naoki Yamamoto
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan
| | - Masaki Suzuki
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan
| | - Hiroki Shichi
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan
| | - Keitaro Kanie
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan
| | - Yasunori Fujita
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan
| | - Hironori Bando
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Hospital, Kobe, Japan
| | - Hidenori Fukuoka
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Hospital, Kobe, Japan
| | - Michiko Takahashi
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Hospital, Kobe, Japan
- Department of Nutrition, Kobe University Hospital, Kobe, Japan
| | - Genzo Iguchi
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan
- Medical Center for Student Health, Kobe University, Kobe, Japan
- Division of Biosignal Pathophysiology, Kobe University, Kobe, Japan
| | - Yutaka Takahashi
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan
- Department of Diabetes and Endocrinology, Nara Medical University, Kashihara, Japan
| | - Wataru Ogawa
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan
| |
Collapse
|
23
|
Kasuki L, Lamback E, Antunes X, Gadelha MR. Biomarkers of response to treatment in acromegaly. Expert Rev Endocrinol Metab 2024; 19:71-80. [PMID: 38078447 DOI: 10.1080/17446651.2023.2293107] [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: 09/29/2023] [Accepted: 12/06/2023] [Indexed: 01/03/2024]
Abstract
INTRODUCTION Medical treatment of acromegaly is based in a `trial and error` approach. First-generation somatostatin receptor ligands (fg-SRL) are prescribed as first-line medical therapy to the vast majority of patients, despite lack of disease control in approximately 60% of patients. However, other drugs used in acromegaly treatment are available (cabergoline, pasireotide and pegvisomant). AREAS COVERED In this article, we review and discuss the biomarkers of response to medical treatment in acromegaly. EXPERT OPINION Biomarkers for fg-SRL that can already be applied in clinical practice are: gender, age, pretreatment GH and IGF-I levels, cytokeratin granulation pattern, and the expression of somatostatin receptor type 2. Using biomarkers of response could guide treatment towards precision medicine with greater efficacy and lower costs.
Collapse
Affiliation(s)
- Leandro Kasuki
- Neuroendocrinology Research Center/Endocrinology Division, Medical School and Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Neuroendocrinology Division, Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria Estadual de Saúde, Rio de Janeiro, Brazil
- Endocrinology Division, Hospital Federal de Bonsucesso, Rio de Janeiro, Brazil
| | - Elisa Lamback
- Neuroendocrinology Research Center/Endocrinology Division, Medical School and Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Neuroendocrinology Division, Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria Estadual de Saúde, Rio de Janeiro, Brazil
- Neuropathology and Molecular Genetics Laboratory, Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria Estadual de Saúde, Rio de Janeiro, Brazil
| | - Ximene Antunes
- Neuroendocrinology Research Center/Endocrinology Division, Medical School and Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Mônica R Gadelha
- Neuroendocrinology Research Center/Endocrinology Division, Medical School and Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Neuroendocrinology Division, Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria Estadual de Saúde, Rio de Janeiro, Brazil
- Neuropathology and Molecular Genetics Laboratory, Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria Estadual de Saúde, Rio de Janeiro, Brazil
| |
Collapse
|
24
|
Basile M, Valentini I, Attanasio R, Cozzi R, Persichetti A, Samperi I, Scoppola A, Auriemma RS, De Menis E, Esposito F, Ferrante E, Iatì G, Mazzatenta D, Poggi M, Rudà R, Tortora F, Cruciani F, Mitrova Z, Saulle R, Vecchi S, Cappabianca P, Paoletta A, Bozzao A, Caputo M, Doglietto F, Ferraù F, Lania AG, Laureti S, Lello S, Locatelli D, Maffei P, Minniti G, Peri A, Ruini C, Settanni F, Silvani A, Veronese N, Grimaldi F, Papini E, Cicchetti A. A pharmacoeconomic analysis from Italian guidelines for the management of prolactinomas. GLOBAL & REGIONAL HEALTH TECHNOLOGY ASSESSMENT 2024; 11:1-16. [PMID: 38230389 PMCID: PMC10788683 DOI: 10.33393/grhta.2024.2601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 11/22/2023] [Indexed: 01/18/2024] Open
Abstract
Background Prolactinoma, the most common pituitary adenoma, is usually treated with dopamine agonist (DA) therapy like cabergoline. Surgery is second-line therapy, and radiotherapy is used if surgical treatment fails or in relapsing macroprolactinoma. Objective This study aimed to provide economic evidence for the management of prolactinoma in Italy, using a cost-of-illness and cost-utility analysis that considered various treatment options, including cabergoline, bromocriptine, temozolomide, radiation therapy, and surgical strategies. Methods The researchers conducted a systematic literature review for each research question on scientific databases and surveyed a panel of experts for each therapeutic procedure's specific drivers that contributed to its total cost. Results The average cost of the first year of treatment was €2,558.91 and €3,287.40 for subjects with microprolactinoma and macroprolactinoma, respectively. Follow-up costs from the second to the fifth year after initial treatment were €798.13 and €1,084.59 per year in both groups. Cabergoline had an adequate cost-utility profile, with an incremental cost-effectiveness ratio (ICER) of €3,201.15 compared to bromocriptine, based on a willingness-to-pay of €40,000 per quality-adjusted life year (QALY) in the reference economy. Endoscopic surgery was more cost-effective than cabergoline, with an ICER of €44,846.64. Considering a willingness-to-pay of €40,000/QALY, the baseline findings show cabergoline to have high cost utility and endoscopic surgery just a tad above that. Conclusions Due to the favorable cost-utility profile and safety of surgical treatment, pituitary surgery should be considered more frequently as the initial therapeutic approach. This management choice could lead to better outcomes and an appropriate allocation of healthcare resources.
Collapse
Affiliation(s)
- Michele Basile
- High School of Economy and Management of Health Systems, Catholic University of Sacred Heart, Rome
| | - Ilaria Valentini
- High School of Economy and Management of Health Systems, Catholic University of Sacred Heart, Rome
| | | | - Renato Cozzi
- ASST Grande Ospedale Metropolitano Niguarda, Endocrinology Department, Milan
| | - Agnese Persichetti
- Ministry of Interior, Department of Firefighters, Public Rescue and Civil Defense, Rome
| | | | | | - Renata Simona Auriemma
- Department of Clinical Medicine and Surgery, Endocrinology Section, ‘Federico II’ University of Naples, Naples
| | - Ernesto De Menis
- Internal Medicine 2, Treviso Hospital, Treviso and Functional Department of Endocrinology and Metabolism, AULSS 2 Veneto
| | - Felice Esposito
- Neurosurgical Clinic, Department of Neurosciences and Reproductive and Odontostomatological Sciences, Federico II’ University of Naples, Naples
| | - Emanuele Ferrante
- Endocrinology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan
| | - Giuseppe Iatì
- Department of Radiation Oncology, University of Messina, Messina
| | - Diego Mazzatenta
- Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, IRCCS Istituto delle Scienze Neurologiche di Bologna, Programma Neurochirurgia Ipofisi - Pituitary Unit, Bologna
| | - Maurizio Poggi
- Endocrinology, Department of Clinical and Molecular Medicine, S. Andrea Hospital, Sapienza University of Rome, Rome
| | - Roberta Rudà
- Division of Neurology, Castelfranco Veneto and Treviso Hospital, Treviso Division of Neuro-Oncology, Department of Neuroscience “Rita Levi Montalcini”, University of Turin, Turin
| | - Fabio Tortora
- Radiology Unit, Department of Advanced Biomedical Sciences, University “Federico II”, Naples
| | - Fabio Cruciani
- Department of Epidemiology, Lazio Region Health Service, Rome
| | - Zuzana Mitrova
- Department of Epidemiology, Lazio Region Health Service, Rome
| | - Rosella Saulle
- Department of Epidemiology, Lazio Region Health Service, Rome
| | - Simona Vecchi
- Department of Epidemiology, Lazio Region Health Service, Rome
| | - Paolo Cappabianca
- Neurosurgical Clinic, Department of Neurosciences and Reproductive and Odontostomatological Sciences, Federico II’ University of Naples, Naples
| | | | - Alessandro Bozzao
- Neuroradiology, S. Andrea Hospital, NESMOS Department (Neuroscience, Mental Health, Sensorial Organs), Sapienza University of Rome, Rome
| | - Marco Caputo
- Laboratorio Analisi Cliniche e Microbiologia, Synlab SRL, Calenzano (FI)
| | | | - Francesco Ferraù
- Department of Human Pathology of Adulthood and Childhood ‘G. Barresi’, University of Messina, Messina
| | - Andrea Gerardo Lania
- Department of Biomedical Sciences, Endocrinology Unit, Humanitas Clinical and Research Center IRCCS, Humanitas University, Rozzano (MI)
| | | | - Stefano Lello
- Department of Woman and Child Health and Public Health, Institute of Obstetrics and Gynecology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome
| | - Davide Locatelli
- Division of Neurosurgery, Department of Biotechnology and Life Sciences, University of Insubria, ASST Sette Laghi, Varese
| | - Pietro Maffei
- Department of Medicine (DIMED), 3rd Medical Clinic, Padua University, Padua
| | - Giuseppe Minniti
- Department of Medicine, Surgery and Neurosciences, University of Siena, Siena
| | - Alessandro Peri
- Pituitary Diseases and Sodium Alterations Unit, AOU Careggi, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Florence
| | - Chiara Ruini
- Department of Psychology, University of Bologna, Bologna
| | - Fabio Settanni
- Clinical Biochemistry Laboratory, City of Health and Science University Hospital, Turin
| | - Antonio Silvani
- Department of Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano
| | - Nadia Veronese
- ASST Grande Ospedale Metropolitano Niguarda, Endocrinology Department, Milan
| | | | - Enrico Papini
- Endocrinology, Ospedale Regina Apostolorum, Albano Laziale (RM) - Italy
| | - Americo Cicchetti
- High School of Economy and Management of Health Systems, Catholic University of Sacred Heart, Rome
| |
Collapse
|
25
|
Kumar U. Somatostatin and Somatostatin Receptors in Tumour Biology. Int J Mol Sci 2023; 25:436. [PMID: 38203605 PMCID: PMC10779198 DOI: 10.3390/ijms25010436] [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: 11/03/2023] [Revised: 12/24/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024] Open
Abstract
Somatostatin (SST), a growth hormone inhibitory peptide, is expressed in endocrine and non-endocrine tissues, immune cells and the central nervous system (CNS). Post-release from secretory or immune cells, the first most appreciated role that SST exhibits is the antiproliferative effect in target tissue that served as a potential therapeutic intervention in various tumours of different origins. The SST-mediated in vivo and/or in vitro antiproliferative effect in the tumour is considered direct via activation of five different somatostatin receptor subtypes (SSTR1-5), which are well expressed in most tumours and often more than one receptor in a single cell. Second, the indirect effect is associated with the regulation of growth factors. SSTR subtypes are crucial in tumour diagnosis and prognosis. In this review, with the recent development of new SST analogues and receptor-specific agonists with emerging functional consequences of signaling pathways are promising therapeutic avenues in tumours of different origins that are discussed.
Collapse
Affiliation(s)
- Ujendra Kumar
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| |
Collapse
|
26
|
Boukerrouni A, Cuny T, Anjou T, Raingeard I, Ferrière A, Grunenwald S, Maïza JC, Marquant E, Sahakian N, Fodil-Cherif S, Salle L, Niccoli P, Randrianaivo H, Sonnet E, Chevalier N, Thuillier P, Vezzosi D, Reynaud R, Dufour H, Brue T, Tabarin A, Delemer B, Kerlan V, Castinetti F, Barlier A, Romanet P. Genetic testing in prolactinomas: a cohort study. Eur J Endocrinol 2023; 189:567-574. [PMID: 37956455 DOI: 10.1093/ejendo/lvad148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/28/2023] [Accepted: 10/19/2023] [Indexed: 11/15/2023]
Abstract
BACKGROUND Prolactinomas represent 46%-66% of pituitary adenomas, but the prevalence of germline mutations is largely unknown. We present here the first study focusing on hereditary predisposition to prolactinoma. OBJECTIVE We studied the prevalence of germline mutations in a large cohort of patients with isolated prolactinomas. MATERIALS AND METHODS A retrospective study was performed combining genetic and clinical data from patients referred for genetic testing of MEN1, AIP, and CDKN1B between 2003 and 2020. SF3B1 was Sanger sequenced in genetically negative patients. RESULTS About 506 patients with a prolactinoma were included: 80 with microprolactinoma (15.9%), 378 with macroprolactinoma (74.7%), 48 unknown; 49/506 in a familial context (9.7%). Among these, 14 (2.8%) had a (likely) pathogenic variant (LPV) in MEN1 or AIP, and none in CDKN1B. All positive patients had developed a macroprolactinoma before age 30. The prevalence of germline mutations in patients with isolated macroprolactinoma under 30 was 4% (11/258) in a sporadic context and 15% (3/20) in a familial context. Prevalence in sporadic cases younger than 18 was 15% in men (5/33) and 7% in women (4/57). No R625H SF3B1 germline mutation was identified in 264 patients with macroprolactinomas. CONCLUSIONS We did not identify any LPVs in patients over 30 years of age, either in a familial or in a sporadic context, and in a sporadic context in our series or the literature. Special attention should be paid to young patients and to familial context.
Collapse
Affiliation(s)
- Amina Boukerrouni
- Aix Marseille Univ, APHM, INSERM, MMG, Laboratory of Molecular Biology Hospital La Conception, MarMaRa Institute, 13005 Marseille, France
| | - Thomas Cuny
- Aix Marseille Univ, APHM, INSERM, MMG, Department of Endocrinology Hospital La Conception, MarMaRa Institute, 13305 Marseille, France
| | - Thibaut Anjou
- Aix Marseille Univ, APHM, INSERM, MMG, Laboratory of Molecular Biology Hospital La Conception, MarMaRa Institute, 13005 Marseille, France
| | - Isabelle Raingeard
- CHRU de Montpellier, Service d'Endocrinologie, Diabète, Maladies Métaboliques, 34000 Montpellier, France
| | - Amandine Ferrière
- Department of Endocrinology, University Hospital of Bordeaux, Haut Lévêque, 33318 Pessac, France
| | - Solange Grunenwald
- Department of Endocrinology and Metabolic Disease, Hospital Larrey CHU (University Hospital Centre), 31029 Toulouse, France
| | - Jean-Christophe Maïza
- Department of Endocrinology, Diabetes and Nutrition, GHSR, Centre Hospitalo-Universitaire de la Réunion, 97416 Saint-Pierre, La Réunion, France
| | - Emeline Marquant
- Aix Marseille Univ, APHM, INSERM, MMG, Department of pediatrics, hospital La Timone Enfants, MarMaRa Institute, 13005 Marseille, France
| | - Nicolas Sahakian
- Aix Marseille Univ, APHM, INSERM, MMG, Department of Endocrinology Hospital La Conception, MarMaRa Institute, 13305 Marseille, France
| | - Sarah Fodil-Cherif
- CHRU de Montpellier, Service d'Endocrinologie, Diabète, Maladies Métaboliques, 34000 Montpellier, France
| | - Laurence Salle
- Inserm, University Limoges, CHU de Limoges, IRD, U1094 Tropical Neuroepidemiology, Institute of Epidemiology and Tropical Neurology, GEIST, 87000 Limoges, France
| | | | - Hanitra Randrianaivo
- UF de Génétique Médicale, GHSR, CHU de La Réunion, 97416 Saint Pierre, La Réunion, France
| | - Emmanuel Sonnet
- Department of Endocrinology and Diabetes, Brest University Hospital, Boulevard Tanguy Prigent, 29200 Brest, France
| | - Nicolas Chevalier
- Centre Hospitalier Universitaire de Nice, Hôpital de l'Archet 2, Service d'Endocrinologie, Diabétologie et Médecine de la Reproduction, 151 route de Saint-Antoine de Ginestière, CS 23079, Nice 06202 Cedex 3, France
| | - Philippe Thuillier
- Department of Endocrinology and Diabetes, Brest University Hospital, Boulevard Tanguy Prigent, 29200 Brest, France
| | - Delphine Vezzosi
- Institut CardioMet, 31000 Toulouse, France
- Service d'endocrinologie, Hôpital Larrey, 24, Chemin de Pouvourville, Toulouse 31029 Cedex 9, France
| | - Rachel Reynaud
- Aix Marseille Univ, APHM, INSERM, MMG, Department of pediatrics, hospital La Timone Enfants, MarMaRa Institute, 13005 Marseille, France
| | - Henry Dufour
- Aix Marseille Univ, APHM, INSERM, MMG, Department of Neurosurgery Hospital la Timone Adulte, MarMaRa Institute, 13005 Marseille, France
| | - Thierry Brue
- Aix Marseille Univ, APHM, INSERM, MMG, Department of Endocrinology Hospital La Conception, MarMaRa Institute, 13305 Marseille, France
| | - Antoine Tabarin
- Department of Endocrinology, University Hospital of Bordeaux, Haut Lévêque, 33318 Pessac, France
| | - Brigitte Delemer
- Endocrinology, Diabetology and Nutrition Unit, University Hospital of Reims, 51454 Reims, France
| | - Véronique Kerlan
- Department of Endocrinology and Diabetes, Brest University Hospital, Boulevard Tanguy Prigent, 29200 Brest, France
| | - Frédéric Castinetti
- Aix Marseille Univ, APHM, INSERM, MMG, Department of Endocrinology Hospital La Conception, MarMaRa Institute, 13305 Marseille, France
| | - Anne Barlier
- Aix Marseille Univ, APHM, INSERM, MMG, Laboratory of Molecular Biology Hospital La Conception, MarMaRa Institute, 13005 Marseille, France
| | - Pauline Romanet
- Aix Marseille Univ, APHM, INSERM, MMG, Laboratory of Molecular Biology Hospital La Conception, MarMaRa Institute, 13005 Marseille, France
| |
Collapse
|
27
|
Kumar S, Memon SS, Lila AR, Sarathi V, Sehemby M, Karlekar M, Sankhe S, Thakkar H, Patil VA, Shah N, Bandgar T. Giant prolactinoma in Asian-Indians: A single-center experience from Western India. ANNALES D'ENDOCRINOLOGIE 2023; 84:711-718. [PMID: 37866429 DOI: 10.1016/j.ando.2023.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 09/25/2023] [Accepted: 10/04/2023] [Indexed: 10/24/2023]
Abstract
PURPOSE Giant prolactinomas (GP) are rare tumors accounting for 4.3% of prolactinomas, with paucity of literature from India. We aim to describe clinical, biochemical, radiological, and treatment outcomes in a large series of Asian-Indian patients with GP. METHODS A single-center retrospective analysis of GPs (n=84), age-based (adults: 66 versus pediatric: 18) and gender-based (males: 64 versus females: 20) comparison was done. RESULTS The mean age at presentation was 34.1±13years, and 64 (76.2%) were males. Males were younger at presentation (32.1±12.2 versus 40.1±13.8years, P: 0.01). The majority presented with mass-effect-related manifestations (visual disturbances: 91.6%, headache: 84.5%) and/or hypogonadism (98.7%). At baseline, largest tumor dimension was 5.3±1.0cm, and serum prolactin was 8343 (3865.5-12,306) ng/mL; most (94.6%) had gonadal axis involvement. Dopamine-agonist (DA) as first-line therapy (45/67, 67.2%) achieved normoprolactinemia (maximum cabergoline dose: 2.0±1.2mg/week) in 36/45 (80%) and tumor response (≥50% reduction) in 36/37 (97.3%) patients at the last follow-up (median duration: 33 [14.5-53.5]months). Notably, gonadal axis recovery was poor (6/30, 20%) despite normoprolactinemia post-DA monotherapy. At latest follow-up, secondary hypothyroidism (32.5% versus 82.6%, P: 0.001) and central hypocortisolism (5.6% versus 42.9%, P: 0.007) were less frequent in DA monotherapy (n=43) than in multimodal therapy group (n=23). The proportion of males (94.4% versus 71.2%, P: 0.04) was higher in the pediatric age group, with DA-induced (first-line) normoprolactinemia observed in 66.7% of them. CONCLUSION GP has male predominance, DA as first-line therapy normalized prolactin in four-fifths of patients with better preservation of HPT and HPA axes in patients with DA monotherapy.
Collapse
Affiliation(s)
- Sandeep Kumar
- Department of Endocrinology, Seth G.S. Medical College & King Edward Memorial Hospital, Mumbai, India
| | - Saba Samad Memon
- Department of Endocrinology, Seth G.S. Medical College & King Edward Memorial Hospital, Mumbai, India
| | - Anurag Ranjan Lila
- Department of Endocrinology, Seth G.S. Medical College & King Edward Memorial Hospital, Mumbai, India
| | - Vijaya Sarathi
- Department of Endocrinology, Vydehi Institute of Medical Sciences, Research Centre, Bangalore, India
| | - Manjeetkaur Sehemby
- Department of Endocrinology, Seth G.S. Medical College & King Edward Memorial Hospital, Mumbai, India
| | - Manjiri Karlekar
- Department of Endocrinology, Seth G.S. Medical College & King Edward Memorial Hospital, Mumbai, India
| | - Shilpa Sankhe
- Department of Radiology, Seth G.S. Medical College & King Edward Memorial Hospital, Mumbai, India
| | - Hemangini Thakkar
- Department of Radiology, Seth G.S. Medical College & King Edward Memorial Hospital, Mumbai, India
| | - Virendra A Patil
- Department of Endocrinology, Seth G.S. Medical College & King Edward Memorial Hospital, Mumbai, India
| | - Nalini Shah
- Department of Endocrinology, Seth G.S. Medical College & King Edward Memorial Hospital, Mumbai, India
| | - Tushar Bandgar
- Department of Endocrinology, Seth G.S. Medical College & King Edward Memorial Hospital, Mumbai, India.
| |
Collapse
|
28
|
Brue T, Rahabi H, Barry A, Barlier A, Bertherat J, Borson-Chazot F, Castinetti F, Cazabat L, Chabre O, Chevalier N, Christin-Maitre S, Cortet C, Drui D, Kamenicky P, Lançon C, Lioté F, Pellegrini I, Reynaud R, Salenave S, Tauveron I, Touraine P, Vantyghem MC, Vergès B, Vezzosi D, Villa C, Raverot G, Coutant R, Chanson P, Albarel F. Position statement on the diagnosis and management of acromegaly: The French National Diagnosis and Treatment Protocol (NDTP). ANNALES D'ENDOCRINOLOGIE 2023; 84:697-710. [PMID: 37579837 DOI: 10.1016/j.ando.2023.08.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/16/2023]
Abstract
Acromegaly is a rare disease with prevalence of approximately 60 cases per million, slight female predominance and peak onset in adults in the fourth decade. Clinical diagnosis is often delayed by several years due to the slowly progressive onset of symptoms. There are multiple clinical criteria that define acromegaly: dysmorphic syndrome of insidious onset, symptoms related to the pituitary tumor (headaches, visual disorders), general signs (sweating, carpal tunnel syndrome, joint pain, etc.), complications of the disease (musculoskeletal, cardiovascular, pneumological, dental, metabolic comorbidities, thyroid nodules, colonic polyps, etc.) or sometimes clinical signs of associated prolactin hypersecretion (erectile dysfunction in men or cycle disorder in women) or concomitant mass-induced hypopituitarism (fatigue and other symptoms related to pituitary hormone deficiencies). Biological confirmation is based initially on elevated IGF-I and lack of GH suppression on oral glucose tolerance test or an elevated mean GH on repeated measurements. In confirmed cases, imaging by pituitary MRI identifies the causal tumor, to best determine management. In a minority of cases, acromegaly can be linked to a genetic predisposition, especially when it occurs at a young age or in a familial context. The first-line treatment is most often surgical removal of the somatotroph pituitary tumor, either immediately or after transient medical treatment. Medical treatments are most often proposed in patients not controlled by surgical removal. Conformal or stereotactic radiotherapy may be discussed on a case-by-case basis, especially in case of drug inefficacy or poor tolerance. Acromegaly should be managed by a multidisciplinary team, preferably within an expert center such as a reference or skill center for rare pituitary diseases.
Collapse
Affiliation(s)
- Thierry Brue
- Service d'endocrinologie, centre de référence des maladies rares de l'hypophyse HYPO, assistance publique-hôpitaux de Marseille (AP-HM), hôpital de la Conception, 147, boulevard Baille, 13005 Marseille, France; Aix Marseille université, INSERM, MMG, Marseille Medical Genetics, Marseille, France.
| | - Haïfa Rahabi
- Service d'endocrinologie, centre de référence des maladies rares de l'hypophyse HYPO, assistance publique-hôpitaux de Marseille (AP-HM), hôpital de la Conception, 147, boulevard Baille, 13005 Marseille, France
| | - Abdoulaye Barry
- Service d'endocrinologie, centre de référence des maladies rares de l'hypophyse HYPO, assistance publique-hôpitaux de Marseille (AP-HM), hôpital de la Conception, 147, boulevard Baille, 13005 Marseille, France
| | - Anne Barlier
- Service d'endocrinologie, centre de référence des maladies rares de l'hypophyse HYPO, assistance publique-hôpitaux de Marseille (AP-HM), hôpital de la Conception, 147, boulevard Baille, 13005 Marseille, France; Aix Marseille université, INSERM, MMG, Marseille Medical Genetics, Marseille, France
| | - Jérôme Bertherat
- Service d'endocrinologie, hôpital Cochin, AP-HP centre université Paris Cité, France
| | - Françoise Borson-Chazot
- Service d'endocrinologie, centre de référence des maladies rares de l'hypophyse HYPO « groupement hospitalier Est » hospices civils de Lyon, 59, boulevard Pinel, 69677 Bron, France
| | - Frédéric Castinetti
- Service d'endocrinologie, centre de référence des maladies rares de l'hypophyse HYPO, assistance publique-hôpitaux de Marseille (AP-HM), hôpital de la Conception, 147, boulevard Baille, 13005 Marseille, France; Aix Marseille université, INSERM, MMG, Marseille Medical Genetics, Marseille, France
| | - Laure Cazabat
- Hôpital Foch, service de neurochirurgie, UMR 1198 BREED, UFR Simone Veil Santé, UVSQ-Paris Saclay, 40, rue Worth, 92150 Suresnes, France
| | - Olivier Chabre
- University Grenoble Alpes, UMR 1292 Inserm-CEA-UGA, endocrinologie CHU de Grenoble Alpes, 38000 Grenoble, France
| | - Nicolas Chevalier
- Université Côte d'Azur, CHU, Inserm U1065, C3M, équipe 5, Nice, France
| | - Sophie Christin-Maitre
- Service d'endocrinologie, diabétologie et médecine de la reproduction, centre de référence des maladies endocriniennes rares de la croissance et du développement (CMERC) Centre de compétence HYPO, Sorbonne université, hôpital Saint-Antoine, Assistance publique-Hôpitaux de Paris, 184, rue du Faubourg Saint-Antoine, 75012 Paris, France
| | - Christine Cortet
- Service d'endocrinologie, diabétologie et maladies métaboliques, CHRU de Lille, rue Polonowski, Lille cedex, France
| | - Delphine Drui
- Service d'endocrinologie, l'institut du thorax, centre hospitalier universitaire de Nantes, boulevard Jacques-Monod, 44093 Nantes cedex, France
| | - Peter Kamenicky
- Service d'endocrinologie et des maladies de la reproduction, centre de référence des maladies rares de l'hypophyse, université Paris-Saclay, Inserm, physiologie et physiopathologie endocriniennes, AP-HP, hôpital BicêtreLe Kremlin-Bicêtre, France
| | - Catherine Lançon
- « Acromégales, pas seulement… », association nationale de l'acromégalie reconnue d'intérêt général, 59234 Villers-Au-Tertre, France
| | - Frédéric Lioté
- Centre Viggo Petersen, faculté de santé, université Paris Cité, Inserm UMR 1132 Bioscar et service de rhumatologie, DMU Locomotion, AP-HP, hôpital Lariboisière, 75475 Paris cedex 10, France
| | - Isabelle Pellegrini
- Service d'endocrinologie, centre de référence des maladies rares de l'hypophyse HYPO, assistance publique-hôpitaux de Marseille (AP-HM), hôpital de la Conception, 147, boulevard Baille, 13005 Marseille, France
| | - Rachel Reynaud
- Aix Marseille université, INSERM, MMG, Marseille Medical Genetics, Marseille, France; Service de pédiatrie multidisciplinaire, centre de référence des maladies rares de l'hypophyse HYPO, Assistance publique-Hôpitaux de Marseille (AP-HM), hôpital de la Timone enfants, 13005 Marseille, France
| | - Sylvie Salenave
- Service d'endocrinologie et des maladies de la reproduction, centre de référence des maladies rares de l'hypophyse, université Paris-Saclay, Inserm, physiologie et physiopathologie endocriniennes, AP-HP, hôpital BicêtreLe Kremlin-Bicêtre, France
| | - Igor Tauveron
- Service d'endocrinologie diabétologie, institut génétique, reproduction & développement (iGReD), CHU de Clermont-Ferrand, CNRS, Inserm, université Clermont-Auvergne, Clermont-Ferrand, France
| | - Philippe Touraine
- Service d'endocrinologie et médecine de la reproduction, centre de maladies endocrinennes rares de la croissance et du développement, Sorbonne université médecine, hôpital Pitié Salpêtrière, Paris, France
| | - Marie-Christine Vantyghem
- Service d'endocrinologie, diabétologie et maladies métaboliques, CHRU de Lille, rue Polonowski, Lille cedex, France; Service d'endocrinologie, l'institut du thorax, centre hospitalier universitaire de Nantes, boulevard Jacques-Monod, 44093 Nantes cedex, France
| | - Bruno Vergès
- Service d'endocrinologie, CHU de Dijon, centre Inserm LNC-UMR1231, 14, rue Gaffarel, 21000 Dijon, France
| | - Delphine Vezzosi
- Service d'endocrinologie, hôpital Larrey, CHU Toulouse, 24 chemin de Pouvourville, TSA 30030, université Paul Sabatier, 21059 Toulouse cedex 9, France
| | - Chiara Villa
- Département de neuropathologie de la Pitié Salpêtrière, hôpital de la Pitié-Salpêtrière - AP-HP, Sorbonne université, 47-83, boulevard de l'Hôpital, 75651 Paris cedex 13, France
| | - Gérald Raverot
- Service d'endocrinologie, centre de référence des maladies rares de l'hypophyse HYPO « groupement hospitalier Est » hospices civils de Lyon, 59, boulevard Pinel, 69677 Bron, France
| | - Régis Coutant
- Service d'endocrinologie-diabétologie-nutrition, centre de référence des maladies rares de l'hypophyse, université d'Angers, CHU d'Angers, Angers, France
| | - Philippe Chanson
- Service d'endocrinologie et des maladies de la reproduction, centre de référence des maladies rares de l'hypophyse, université Paris-Saclay, Inserm, physiologie et physiopathologie endocriniennes, AP-HP, hôpital BicêtreLe Kremlin-Bicêtre, France
| | - Frédérique Albarel
- Service d'endocrinologie, centre de référence des maladies rares de l'hypophyse HYPO, assistance publique-hôpitaux de Marseille (AP-HM), hôpital de la Conception, 147, boulevard Baille, 13005 Marseille, France
| |
Collapse
|
29
|
Serioli S, Agostini L, Pietrantoni A, Valeri F, Costanza F, Chiloiro S, Buffoli B, Piazza A, Poliani PL, Peris-Celda M, Iavarone F, Gaudino S, Gessi M, Schinzari G, Mattogno PP, Giampietro A, De Marinis L, Pontecorvi A, Fontanella MM, Lauretti L, Rindi G, Olivi A, Bianchi A, Doglietto F. Aggressive PitNETs and Potential Target Therapies: A Systematic Review of Molecular and Genetic Pathways. Int J Mol Sci 2023; 24:15719. [PMID: 37958702 PMCID: PMC10650665 DOI: 10.3390/ijms242115719] [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: 08/25/2023] [Revised: 10/22/2023] [Accepted: 10/24/2023] [Indexed: 11/15/2023] Open
Abstract
Recently, advances in molecular biology and bioinformatics have allowed a more thorough understanding of tumorigenesis in aggressive PitNETs (pituitary neuroendocrine tumors) through the identification of specific essential genes, crucial molecular pathways, regulators, and effects of the tumoral microenvironment. Target therapies have been developed to cure oncology patients refractory to traditional treatments, introducing the concept of precision medicine. Preliminary data on PitNETs are derived from preclinical studies conducted on cell cultures, animal models, and a few case reports or small case series. This study comprehensively reviews the principal pathways involved in aggressive PitNETs, describing the potential target therapies. A search was conducted on Pubmed, Scopus, and Web of Science for English papers published between 1 January 2004, and 15 June 2023. 254 were selected, and the topics related to aggressive PitNETs were recorded and discussed in detail: epigenetic aspects, membrane proteins and receptors, metalloprotease, molecular pathways, PPRK, and the immune microenvironment. A comprehensive comprehension of the molecular mechanisms linked to PitNETs' aggressiveness and invasiveness is crucial. Despite promising preliminary findings, additional research and clinical trials are necessary to confirm the indications and effectiveness of target therapies for PitNETs.
Collapse
Affiliation(s)
- Simona Serioli
- Division of Neurosurgery, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, 25123 Brescia, Italy;
| | - Ludovico Agostini
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 20123 Rome, Italy; (L.A.); (F.V.); (F.C.); (S.G.); (M.G.); (G.S.); (L.D.M.); (A.P.); (L.L.); (G.R.); (A.O.); (A.B.); (F.D.)
- Department of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy;
| | | | - Federico Valeri
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 20123 Rome, Italy; (L.A.); (F.V.); (F.C.); (S.G.); (M.G.); (G.S.); (L.D.M.); (A.P.); (L.L.); (G.R.); (A.O.); (A.B.); (F.D.)
- Department of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy;
| | - Flavia Costanza
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 20123 Rome, Italy; (L.A.); (F.V.); (F.C.); (S.G.); (M.G.); (G.S.); (L.D.M.); (A.P.); (L.L.); (G.R.); (A.O.); (A.B.); (F.D.)
- Pituitary Unit, Division of Endocrinology and Metabolism, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy;
| | - Sabrina Chiloiro
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 20123 Rome, Italy; (L.A.); (F.V.); (F.C.); (S.G.); (M.G.); (G.S.); (L.D.M.); (A.P.); (L.L.); (G.R.); (A.O.); (A.B.); (F.D.)
- Pituitary Unit, Division of Endocrinology and Metabolism, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy;
| | - Barbara Buffoli
- Section of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, 25121 Brescia, Italy;
| | - Amedeo Piazza
- Department of Neuroscience, Neurosurgery Division, “Sapienza” University of Rome, 00185 Rome, Italy;
| | - Pietro Luigi Poliani
- Pathology Unit, Vita-Salute San Raffaele University, IRCCS San Raffaele, 20132 Milan, Italy;
| | - Maria Peris-Celda
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA;
- Department of Otolaryngology/Head and Neck Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Federica Iavarone
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, 20123 Rome, Italy;
- Fondazione Policlinico Universitario IRCCS “A. Gemelli”, 00168 Rome, Italy
| | - Simona Gaudino
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 20123 Rome, Italy; (L.A.); (F.V.); (F.C.); (S.G.); (M.G.); (G.S.); (L.D.M.); (A.P.); (L.L.); (G.R.); (A.O.); (A.B.); (F.D.)
- Department of Radiological Sciences, Institute of Radiology, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Marco Gessi
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 20123 Rome, Italy; (L.A.); (F.V.); (F.C.); (S.G.); (M.G.); (G.S.); (L.D.M.); (A.P.); (L.L.); (G.R.); (A.O.); (A.B.); (F.D.)
- Neuropathology Unit, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Giovanni Schinzari
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 20123 Rome, Italy; (L.A.); (F.V.); (F.C.); (S.G.); (M.G.); (G.S.); (L.D.M.); (A.P.); (L.L.); (G.R.); (A.O.); (A.B.); (F.D.)
- Department of Oncology, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Pier Paolo Mattogno
- Department of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy;
| | - Antonella Giampietro
- Pituitary Unit, Division of Endocrinology and Metabolism, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy;
| | - Laura De Marinis
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 20123 Rome, Italy; (L.A.); (F.V.); (F.C.); (S.G.); (M.G.); (G.S.); (L.D.M.); (A.P.); (L.L.); (G.R.); (A.O.); (A.B.); (F.D.)
- Pituitary Unit, Division of Endocrinology and Metabolism, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy;
| | - Alfredo Pontecorvi
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 20123 Rome, Italy; (L.A.); (F.V.); (F.C.); (S.G.); (M.G.); (G.S.); (L.D.M.); (A.P.); (L.L.); (G.R.); (A.O.); (A.B.); (F.D.)
- Pituitary Unit, Division of Endocrinology and Metabolism, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy;
| | - Marco Maria Fontanella
- Division of Neurosurgery, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, 25123 Brescia, Italy;
| | - Liverana Lauretti
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 20123 Rome, Italy; (L.A.); (F.V.); (F.C.); (S.G.); (M.G.); (G.S.); (L.D.M.); (A.P.); (L.L.); (G.R.); (A.O.); (A.B.); (F.D.)
- Department of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy;
| | - Guido Rindi
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 20123 Rome, Italy; (L.A.); (F.V.); (F.C.); (S.G.); (M.G.); (G.S.); (L.D.M.); (A.P.); (L.L.); (G.R.); (A.O.); (A.B.); (F.D.)
- Neuropathology Unit, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Alessandro Olivi
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 20123 Rome, Italy; (L.A.); (F.V.); (F.C.); (S.G.); (M.G.); (G.S.); (L.D.M.); (A.P.); (L.L.); (G.R.); (A.O.); (A.B.); (F.D.)
- Department of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy;
| | - Antonio Bianchi
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 20123 Rome, Italy; (L.A.); (F.V.); (F.C.); (S.G.); (M.G.); (G.S.); (L.D.M.); (A.P.); (L.L.); (G.R.); (A.O.); (A.B.); (F.D.)
- Pituitary Unit, Division of Endocrinology and Metabolism, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy;
| | - Francesco Doglietto
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 20123 Rome, Italy; (L.A.); (F.V.); (F.C.); (S.G.); (M.G.); (G.S.); (L.D.M.); (A.P.); (L.L.); (G.R.); (A.O.); (A.B.); (F.D.)
- Department of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy;
| |
Collapse
|
30
|
Vroonen L, Beckers A, Camby S, Cuny T, Beckers P, Jaffrain-Rea ML, Cogne M, Naves L, Ferriere A, Romanet P, Elenkova A, Karhu A, Brue T, Barlier A, Pétrossians P, Daly AF. The clinical and therapeutic profiles of prolactinomas associated with germline pathogenic variants in the aryl hydrocarbon receptor interacting protein (AIP) gene. Front Endocrinol (Lausanne) 2023; 14:1242588. [PMID: 37711900 PMCID: PMC10498111 DOI: 10.3389/fendo.2023.1242588] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 07/17/2023] [Indexed: 09/16/2023] Open
Abstract
Introduction Prolactinomas are the most frequent type of pituitary adenoma encountered in clinical practice. Dopamine agonists (DA) like cabergoline typically provide sign/ symptom control, normalize prolactin levels and decrease tumor size in most patients. DA-resistant prolactinomas are infrequent and can occur in association with some genetic causes like MEN1 and pathogenic germline variants in the AIP gene (AIPvar). Methods We compared the clinical, radiological, and therapeutic characteristics of AIPvar-related prolactinomas (n=13) with unselected hospital-treated prolactinomas ("unselected", n=41) and genetically-negative, DA-resistant prolactinomas (DA-resistant, n=39). Results AIPvar-related prolactinomas occurred at a significantly younger age than the unselected or DA-resistant prolactinomas (p<0.01). Males were more common in the AIPvar (75.0%) and DA- resistant (49.7%) versus unselected prolactinomas (9.8%; p<0.001). AIPvar prolactinomas exhibited significantly more frequent invasion than the other groups (p<0.001) and exhibited a trend to larger tumor diameter. The DA-resistant group had significantly higher prolactin levels at diagnosis than the AIPvar group (p<0.001). Maximum DA doses were significantly higher in the AIPvar and DA-resistant groups versus unselected. DA-induced macroadenoma shrinkage (>50%) occurred in 58.3% in the AIPvar group versus 4.2% in the DA-resistant group (p<0.01). Surgery was more frequent in the AIPvar and DA- resistant groups (43.8% and 61.5%, respectively) versus unselected (19.5%: p<0.01). Radiotherapy was used only in AIPvar (18.8%) and DA-resistant (25.6%) groups. Discussion AIPvar confer an aggressive phenotype in prolactinomas, with invasive tumors occurring at a younger age. These characteristics can help differentiate rare AIPvar related prolactinomas from DA-resistant, genetically-negative tumors.
Collapse
Affiliation(s)
- Laurent Vroonen
- Department of Endocrinology, Centre Hospitalier Universitaire de Liège, University of Liège, Liège, Belgium
| | - Albert Beckers
- Department of Endocrinology, Centre Hospitalier Universitaire de Liège, University of Liège, Liège, Belgium
| | - Severine Camby
- Department of Otorhinolaryngology, Centre Hospitalier Universitaire de Liège, University of Liège, Liège, Belgium
| | - Thomas Cuny
- Department of Endocrinology, Aix Marseille University, Assistance publique Hôpitaux de Marseille (APHM), INSERM, Marseille Medical Genetics (MMG), Hopital La Conception, Institut MarMaRa, Marseille, France
| | - Pablo Beckers
- Department of Human Genetics, Centre Hospitalier Universitaire de Liège, University of Liège, Liège, Belgium
| | - Marie-Lise Jaffrain-Rea
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
- Department of Neuroendocrinology, Neuromed IRCCS, Pozzilli, Italy
| | - Muriel Cogne
- Department of Endocrinology, Diabetes and Nutrition, Centre Hospitalo-Universitaire de la Réunion, Saint-Pierre, France
| | - Luciana Naves
- Department of Endocrinology, University of Brasilia, Brasilia, Brazil
| | - Amandine Ferriere
- Department of Endocrinology, Hopital Haut-Leveque, Centre Hospitalier Universitaire (CHU) de Bordeaux, Pessac, France
| | - Pauline Romanet
- Laboratory of Molecular Biology, Aix Marseille University, Assistance publique Hôpitaux de Marseille (APHM), INSERM, Marseille Medical Genetics (MMG), Hospital La Conception, Institut MarMaRa, Marseille, France
| | - Atanaska Elenkova
- Department of Endocrinology, Medical University of Sofia, Sofia, Bulgaria
| | - Auli Karhu
- Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland
- Applied Tumor Genomics Research Program, Research Programs Unit, University of Helsinki, Helsinki, Finland
| | - Thierry Brue
- Department of Endocrinology, Aix Marseille University, Assistance publique Hôpitaux de Marseille (APHM), INSERM, Marseille Medical Genetics (MMG), Hopital La Conception, Institut MarMaRa, Marseille, France
- Laboratory of Molecular Biology, Aix Marseille University, Assistance publique Hôpitaux de Marseille (APHM), INSERM, Marseille Medical Genetics (MMG), Hospital La Conception, Institut MarMaRa, Marseille, France
| | - Anne Barlier
- Laboratory of Molecular Biology, Aix Marseille University, Assistance publique Hôpitaux de Marseille (APHM), INSERM, Marseille Medical Genetics (MMG), Hospital La Conception, Institut MarMaRa, Marseille, France
| | - Patrick Pétrossians
- Department of Endocrinology, Centre Hospitalier Universitaire de Liège, University of Liège, Liège, Belgium
| | - Adrian F. Daly
- Department of Endocrinology, Centre Hospitalier Universitaire de Liège, University of Liège, Liège, Belgium
| |
Collapse
|
31
|
Auriemma RS, Pirchio R, Pivonello C, Garifalos F, Colao A, Pivonello R. Approach to the Patient With Prolactinoma. J Clin Endocrinol Metab 2023; 108:2400-2423. [PMID: 36974474 PMCID: PMC10438891 DOI: 10.1210/clinem/dgad174] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 03/03/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023]
Abstract
Prolactinomas are the most common pituitary tumor histotype, with microprolactinomas being prevalent in women and macroprolactinomas in men. Hyperprolactinemia is among the most common causes of hypogonadotropic hypogonadism in both sexes, prompting medical advice for hypogonadism (infertility, oligo-amenorrhea, impotence, osteoporosis/osteopenia) in both sexes, and for signs and symptoms of mass effects (hypopituitarism, visual loss, optic chiasm compression, cranial nerve deficits, headaches) predominantly in men. Diagnostic workup involves a single prolactin measurement and pituitary imaging, but some laboratory artifacts (ie, the "hook effect" and macroprolactin) can complicate or delay the diagnosis. The treatment of choice for prolactinomas is represented by dopamine agonists, mainly cabergoline, which are able to induce disease control, restore fertility in both sexes, and definitively cure one-third of patients, thus permitting treatment discontinuation. Pregnancy and menopause may promote spontaneous prolactin decline and anticipate cabergoline discontinuation in women. Surgery and/or radiotherapy are indicated in case of resistance to cabergoline not overcome by the increase in drug dose up to the maximally tolerated or the patient's personal choice of surgery. The evidence of resistance to cabergoline in invasive and proliferative tumors may indicate biological aggressiveness, thus requiring alternative therapeutic approaches mainly based on temozolomide use as monotherapy or combined with radiotherapy. In uncontrolled patients, new medical approaches (alternative hormonal treatments, cytotoxic drugs, peptide receptor radionuclide therapy, mTOR/Akt inhibitors, tyrosine kinase inhibitors, or immunotherapy) may be offered but the experience collected to date is still very scant. This article reviews different facets of prolactinomas and discusses approaches to the condition in more common clinical situations.
Collapse
Affiliation(s)
- Renata S Auriemma
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, 80131 Naples, Italy
| | - Rosa Pirchio
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, 80131 Naples, Italy
| | - Claudia Pivonello
- Dipartimento di Sanità Pubblica, Università Federico II di Napoli, 80131 Naples, Italy
| | - Francesco Garifalos
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, 80131 Naples, Italy
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Unità di Andrologia e Medicina della Riproduzione e Sessualità Maschile e Femminile (FERTISEXCARES), Università Federico II di Napoli, 80131 Naples, Italy
| | - Annamaria Colao
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, 80131 Naples, Italy
- Unesco Chair for Health Education and Sustainable Development, “Federico II” University, 80131 Naples, Italy
| | - Rosario Pivonello
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, 80131 Naples, Italy
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Unità di Andrologia e Medicina della Riproduzione e Sessualità Maschile e Femminile (FERTISEXCARES), Università Federico II di Napoli, 80131 Naples, Italy
- Unesco Chair for Health Education and Sustainable Development, “Federico II” University, 80131 Naples, Italy
| |
Collapse
|
32
|
Stratakis CA. An update on, and genetics of refractory adenomas of childhood. Pituitary 2023:10.1007/s11102-023-01327-2. [PMID: 37318708 DOI: 10.1007/s11102-023-01327-2] [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] [Accepted: 05/16/2023] [Indexed: 06/16/2023]
Abstract
Pituitary adenomas in childhood tend to be more frequently due to germline genetic changes and are often diagnosed at late stages due to delayed recognition by pediatricians and other caretakers who are not familiar with this rare disease in childhood. As a result, often, pediatric pituitary adenomas are aggressive or remain refractory to treatment. In this review, we discuss germline genetic defects that account for the most common pediatric pituitary adenomas that are refractory to treatment. We also discuss some somatic genetic events, such as chromosomal copy number changes that characterize some of the most aggressive pituitary adenomas in childhood that end up being refractory to treatment.
Collapse
Affiliation(s)
- Constantine A Stratakis
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health, 20892, Bethesda, MD, USA.
- Human Genetics & Precision Medicine, IMMB, FORTH, Heraklion, Greece.
- ELPEN Research Institute, Athens, Greece.
- Medical Genetics, H. Dunant Hospital, Athens, Greece.
- Faculty of Medicine, European University of Cyprus, Nicosia, Cyprus.
| |
Collapse
|
33
|
Marques P. The Effects of Peptide Receptor Radionuclide Therapy on the Neoplastic and Normal Pituitary. Cancers (Basel) 2023; 15:2710. [PMID: 37345047 PMCID: PMC10216433 DOI: 10.3390/cancers15102710] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/07/2023] [Accepted: 05/08/2023] [Indexed: 06/23/2023] Open
Abstract
Pituitary neuroendocrine tumours (PitNETs) are usually benign and slow-growing; however, in some cases, they may behave aggressively and become resistant to conventional treatments. Therapeutic options for aggressive or metastatic PitNETs are limited, and currently mainly consist of temozolomide, with little experience of other emerging approaches, including peptide receptor radionuclide therapy (PRRT). Somatostatin receptor expression in PitNETs explains the effectiveness of somatostatin analogues for treating PitNETs, particularly those hypersecreting pituitary hormones, such as growth hormone or adrenocorticotropic hormone. The expression of such receptors in pituitary tumour cells has provided the rationale for using PRRT to treat patients with aggressive or metastatic PitNETs. However, the PRRT efficacy in this setting remains unestablished, as knowledge on this today is based only on few case reports and small series of cases, which are reviewed here. A total of 30 PRRT-treated patients have been thus far reported: 23 aggressive PitNETs, 5 carcinomas, and 2 of malignancy status unspecified. Of the 27 published cases with information regarding the response to PRRT, 5 (18%) showed a partial response, 8 (30%) had stable disease, and 14 (52%) had progressive disease. No major adverse effects have been reported, and there is also no increased risk of clinically relevant hypopituitarism in patients with pituitary or non-pituitary neuroendocrine tumours following PRRT. PRRT may be regarded as a safe option for patients with aggressive or metastatic PitNETs if other treatment approaches are not feasible or have failed in controlling the disease progression, with tumour shrinkage occurring in up to a fifth of cases, while about a third of aggressive pituitary tumours may achieve stable disease. Here, the data on PRRT in the management of patients with aggressive pituitary tumours are reviewed, as well as the effects of PRRT on the pituitary function in other PRRT-treated cancer patients.
Collapse
Affiliation(s)
- Pedro Marques
- Pituitary Tumor Unit, Endocrinology Department, Hospital CUF Descobertas, 1998-018 Lisbon, Portugal;
- Faculdade de Medicina, Universidade Católica Portuguesa, 2635-631 Lisbon, Portugal
| |
Collapse
|
34
|
Vamvoukaki R, Chrysoulaki M, Betsi G, Xekouki P. Pituitary Tumorigenesis-Implications for Management. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:medicina59040812. [PMID: 37109772 PMCID: PMC10145673 DOI: 10.3390/medicina59040812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 04/11/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023]
Abstract
Pituitary neuroendocrine tumors (PitNETs), the third most common intracranial tumor, are mostly benign. However, some of them may display a more aggressive behavior, invading into the surrounding structures. While they may rarely metastasize, they may resist different treatment modalities. Several major advances in molecular biology in the past few years led to the discovery of the possible mechanisms involved in pituitary tumorigenesis with a possible therapeutic implication. The mutations in the different proteins involved in the Gsa/protein kinase A/c AMP signaling pathway are well-known and are responsible for many PitNETS, such as somatotropinomas and, in the context of syndromes, as the McCune-Albright syndrome, Carney complex, familiar isolated pituitary adenoma (FIPA), and X-linked acrogigantism (XLAG). The other pathways involved are the MAPK/ERK, PI3K/Akt, Wnt, and the most recently studied HIPPO pathways. Moreover, the mutations in several other tumor suppressor genes, such as menin and CDKN1B, are responsible for the MEN1 and MEN4 syndromes and succinate dehydrogenase (SDHx) in the context of the 3PAs syndrome. Furthermore, the pituitary stem cells and miRNAs hold an essential role in pituitary tumorigenesis and may represent new molecular targets for their diagnosis and treatment. This review aims to summarize the different cell signaling pathways and genes involved in pituitary tumorigenesis in an attempt to clarify their implications for diagnosis and management.
Collapse
Affiliation(s)
- Rodanthi Vamvoukaki
- Endocrinology and Diabetes Clinic, University Hospital of Heraklion, School of Medicine, University of Crete, 71500 Crete, Greece
| | - Maria Chrysoulaki
- Endocrinology and Diabetes Clinic, University Hospital of Heraklion, School of Medicine, University of Crete, 71500 Crete, Greece
| | - Grigoria Betsi
- Endocrinology and Diabetes Clinic, University Hospital of Heraklion, School of Medicine, University of Crete, 71500 Crete, Greece
| | - Paraskevi Xekouki
- Endocrinology and Diabetes Clinic, University Hospital of Heraklion, School of Medicine, University of Crete, 71500 Crete, Greece
| |
Collapse
|
35
|
Abstract
Hereditary pituitary tumorigenesis is seen in a relatively small proportion (around 5%) of patients with pituitary neuroendocrine tumors (PitNETs). The aim of the current review is to describe the main clinical and molecular features of such pituitary tumors associated with hereditary or familial characteristics, many of which have now been genetically identified. The genetic patterns of inheritance are classified into isolated familial PitNETs and the syndromic tumors. In general, the established genetic causes of familial tumorigenesis tend to present at a younger age, often pursue a more aggressive course, and are more frequently associated with growth hormone hypersecretion compared to sporadic tumors. The mostly studied molecular pathways implicated are the protein kinase A and phosphatidyl-inositol pathways, which are in the main related to mutations in the syndromes of familial isolated pituitary adenoma (FIPA), Carney complex syndrome, and X-linked acrogigantism. Another well-documented mechanism consists of the regulation of p27 or p21 proteins, with further acceleration of the pituitary cell cycle through the check points G1/S and M/G1, mostly documented in multiple endocrine neoplasia type 4. In conclusion, PitNETs may occur in relation to well-established familial germline mutations which may determine the clinical phenotype and the response to treatment, and may require family screening.
Collapse
Affiliation(s)
- Eleni Armeni
- Dept. of Endocrinology, Royal Free Hospital, London, NW3 2QG, UK.
| | - Ashley Grossman
- Dept. of Endocrinology, Royal Free Hospital, London, NW3 2QG, UK
- Centre for Endocrinology, Barts and the London School of Medicine, London, UK
- Green Templeton College, University of Oxford, Oxford, UK
| |
Collapse
|
36
|
García-de-la-Torre KS, Kerbel J, Cano-Zaragoza A, Mercado M. Atypical Course of a Patient With AIP-Positive Acromegaly: From GH Excess to GH Deficiency and Back to GH Excess. JCEM CASE REPORTS 2023; 1:luad034. [PMID: 37908467 PMCID: PMC10580445 DOI: 10.1210/jcemcr/luad034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Indexed: 11/02/2023]
Abstract
Acromegaly/giantism results from the chronic excess of growth hormone (GH) and insulin-like growth factor-1 (IGF-1), in more than 96% of cases, due to a GH-secreting pituitary adenoma. Primary treatment of choice is transsphenoidal resection of the adenoma. More than 30% to 40% of operated cases require adjunctive forms of treatment, be it pharmacological or radiotherapeutical. The multimodal treatment of acromegaly has resulted in substantial improvements in the quality of life and life expectancy of these patients. We herein present the complex case of a patient with acromegaly due to a mammosomatotrope adenoma, with a germ-line AIP (aryl hydrocarbon receptor-interacting protein) mutation, who had a chronic and protracted course of more than 15 years during which he was treated with surgery, somatostatin receptor ligands, dopamine agonist, and the GH receptor antagonist pegvisomant. At one point, he was able to come off medications and was even found to be transiently GH-deficient, only to develop acromegaly again after a couple of years.
Collapse
Affiliation(s)
- Keren-Sandyn García-de-la-Torre
- Endocrinology Service and Endocrine Research Unit, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico
| | - Jacobo Kerbel
- Endocrinology Service and Endocrine Research Unit, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico
| | - Amayrani Cano-Zaragoza
- Endocrinology Service and Endocrine Research Unit, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico
| | - Moisés Mercado
- Endocrinology Service and Endocrine Research Unit, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico
| |
Collapse
|
37
|
Horiguchi K. The molecular biology of thyrotroph pituitary neuroendocrine tumors. Endocr J 2023; 70:135-139. [PMID: 36653153 DOI: 10.1507/endocrj.ej22-0514] [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: 01/19/2023] Open
Abstract
Pituitary neuroendocrine tumors (PitNETs), which were formerly known as pituitary adenomas, are classified in 5th Edition of the WHO Classification of Endocrine and Neuroendocrine Tumors. Since thyrotroph PitNETs are rare PitNETs, most previous studies about former thyroid stimulating hormone (TSH)-secreting pituitary adenoma have focused on a small number of cases. However, the diagnostic rate of thyrotroph PitNET has increased because of increased sensitivity of serum TSH measurement and widespread recognition that thyrotroph PitNETs are the cause of syndrome of inappropriate secretion of TSH (SITSH). Therefore, knowledge on the molecular mechanism of thyrotroph PitNET is gradually accumulating. Recently, comprehensive chromosomal, genetic, and epigenomic alterations in thyrotroph PitNET have been revealed with the availability of comprehensive gene and protein analyses, and the nature of thyrotroph PitNET is gradually being elucidated. However, further analysis is needed to determine whether the causes of these changes are directly responsible for the development of tumors.
Collapse
Affiliation(s)
- Kazuhiko Horiguchi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Gunma University Graduate School of Medicine, Gunma 371-8511, Japan
| |
Collapse
|
38
|
Thyrotropin-secreting tumor "TSH-PitNET": From diagnosis to treatment. ANNALES D'ENDOCRINOLOGIE 2023:S0003-4266(23)00024-0. [PMID: 36716819 DOI: 10.1016/j.ando.2023.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Thyrotropic adenomas (TSH-PitNET) are the rarest pituitary tumours. Most TSH-PitNETs are secreting adenoma, with a biological picture of inappropriate TSH secretion (moderately elevated TSH, elevated FT3 and FT4). Patients present most often clinical hyperthyroidism, but with more moderate symptoms than in peripheral hyperthyroidism. Biological diagnosis is not always easy. The main differential diagnoses are interfering antibody assay interactions, dysalbuminemia and thyroid hormone resistance syndrome. Misdiagnosis is common. However, the diagnosis is easier when macroadenomas are involved (80% of cases), with symptoms of optic chiasm compression, headache and signs of hypopituitarism. Treatment is initially based on surgery. In case of failure, somatostatin analogues are very effective in controlling tumor volume and secretion, although there is a risk of thyroid insufficiency, which is usually transient.
Collapse
|
39
|
Trofimiuk-Müldner M, Domagała B, Sokołowski G, Skalniak A, Hubalewska-Dydejczyk A. AIP gene germline variants in adult Polish patients with apparently sporadic pituitary macroadenomas. Front Endocrinol (Lausanne) 2023; 14:1098367. [PMID: 36843582 PMCID: PMC9950257 DOI: 10.3389/fendo.2023.1098367] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 01/23/2023] [Indexed: 02/12/2023] Open
Abstract
INTRODUCTION Up to 5% of all pituitary tumors are hereditary e.g. due to MEN1 or aryl hydrocarbon receptor-interacting protein (AIP) genes mutations. OBJECTIVES The study was aimed at the assessment of the frequency and characteristics of AIP-mutation related tumors in patients with apparently sporadic pituitary macroadenomas in the Polish population. MATERIALS AND METHODS The study included 131 patients (57 males, 74 females; median age 42 years) diagnosed with pituitary macroadenomas, and with a negative family history of familial isolated pituitary adenoma (FIPA) or multiple endocrine neoplasia type 1 (MEN1) syndromes. Sanger sequencing was used for the assessment of AIP gene variants. The study was approved by the Ethics Board of JUMC. RESULTS AIP variants were identified in five of the 131 included subjects (3.8%): one diagnosed with Cushing's disease, two with acromegaly, and two with non-secreting adenomas. Patients harboring hereditary AIP gene alterations did not differ from the rest of the study group in median age at diagnosis (41.0 vs. 42.5 years, P=0.8), median largest tumor diameter (25 vs. 24 mm, P=0.6), gender distribution (60.0% vs. 56.3% females, P=0.8), secreting tumor frequency (60.0% vs. 67.5%, P=0.7), or acromegaly diagnosis frequency (40.0% vs.37.3%, P=0.9). CONCLUSIONS In our series of apparently sporadic pituitary macroadenomas, AIP gene variant carriers did not differ substantially from patients with negative genetic testing. A risk factor-centred approach to AIP genetic screening may result in missing germline variants. Considering the clinical impact of such genetic variants and their relatively low penetrance, it is, however, doubtful if general genetic screening benefits the whole cohort of pituitary macroadenoma patients and their families.
Collapse
Affiliation(s)
- Małgorzata Trofimiuk-Müldner
- Chair and Department of Endocrinology, Jagiellonian University Medical College, Kraków, Poland
- *Correspondence: Małgorzata Trofimiuk-Müldner,
| | - Bartosz Domagała
- Department of Endocrinology, Endocrine Oncology and Nuclear Medicine, University Hospital in Kraków, Kraków, Poland
| | - Grzegorz Sokołowski
- Chair and Department of Endocrinology, Jagiellonian University Medical College, Kraków, Poland
| | - Anna Skalniak
- Chair and Department of Endocrinology, Jagiellonian University Medical College, Kraków, Poland
| | | |
Collapse
|
40
|
Angelousi A, Koumarianou A, Chatzellis E, Kaltsas G. Resistance of neuroendocrine tumours to somatostatin analogs. Expert Rev Endocrinol Metab 2023; 18:33-52. [PMID: 36651768 DOI: 10.1080/17446651.2023.2166488] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 01/05/2023] [Indexed: 01/13/2023]
Abstract
INTRODUCTION A common feature shared by most neuroendocrine tumors (NETs) is the expression on their surface of somatostatin receptors (SSTRs) that are essential for their pathophysiological regulation, diagnosis, and management. The first-generation synthetic somatostatin analogs (SSAs), octreotide and lanreotide, constitute the cornerstone of treatment for growth hormone secreting pituitary adenomas and functioning, progressive functioning, and non-functioning gastro-entero-pancreatic (GEP-NETs). SSAs exert their mechanism of action through binding to the SSTRs; however, their therapeutic response is frequently attenuated or diminished by the development of resistance. The phenomenon of resistance is complex implicating the presence of additional epigenetic and genetic mechanisms. AREAS COVERED We aim to analyze the molecular, genetic, and epigenetic mechanisms of resistance to SSA treatment. We also summarize recent clinical data related to the development of resistance on conventional and non-conventional modes of administration of the first-generation SSAs and the second-generation SSA pasireotide. We explore mechanisms used to counteract the resistance to SSAs using higher doses or more frequent mode of administration of SSAs and/or combination treatments. EXPERT OPINION There is considerable heterogeneity in the development of resistance to SSAs that is tumor-specific necessitating the delineation of the underlying pathophysiological processes to further expand their therapeutic applications.
Collapse
Affiliation(s)
- Anna Angelousi
- First Department of Internal Medicine, Unit of Endocrinology, Laikon General hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Anna Koumarianou
- Hematology Oncology Unit, Fourth Department of Internal Medicine, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Eleftherios Chatzellis
- Endocrinology Diabetes and Metabolism Department, 251 Hellenic Air Force and VA General Hospital, Athens, Greece
| | - Gregory Kaltsas
- First Propaedeutic Department of Internal Medicine, Endocrine Unit, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
41
|
Abstract
Acromegaly is a systemic disease associated with increased morbidity and mortality that can be prevented with adequate disease control. Three modalities of treatment (surgery, medical treatment, and radiotherapy) are available; however, a significant proportion of patients still maintain disease activity despite treatment. Therefore, there is a need for innovations in the treatment of acromegaly that include changes in the current trial and error approach and the development of new drugs. In this review, we summarize the recent innovations in the treatment of acromegaly and address the future perspectives in this field.
Collapse
Affiliation(s)
- Leandro Kasuki
- Neuroendocrinology Research Center/Endocrinology Division, Medical School and Hospital Universitário Clementino Fraga Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil; Neuroendocrinology Division, Instituto Estadual Do Cérebro Paulo Niemeyer, Secretaria Estadual de Saúde, Rio de Janeiro, Brazil; Endocrinology Division, Hospital Federal de Bonsucesso, Rio de Janeiro, Brazil
| | - Mônica R Gadelha
- Neuroendocrinology Research Center/Endocrinology Division, Medical School and Hospital Universitário Clementino Fraga Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil; Neuroendocrinology Division, Instituto Estadual Do Cérebro Paulo Niemeyer, Secretaria Estadual de Saúde, Rio de Janeiro, Brazil; Neuropathology and Molecular Genetics Laboratory, Instituto Estadual Do Cérebro Paulo Niemeyer, Secretaria Estadual de Saúde, Rio de Janeiro, Brazil.
| |
Collapse
|
42
|
Kumar S, Sarathi V, Lila AR, Sehemby M, Memon SS, Karlekar M, Sankhe S, Patil VA, Shah N, Bandgar T. Giant prolactinoma in children and adolescents: a single-center experience and systematic review. Pituitary 2022; 25:819-830. [PMID: 35851929 DOI: 10.1007/s11102-022-01250-y] [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] [Accepted: 06/28/2022] [Indexed: 11/29/2022]
Abstract
PURPOSE Giant prolactinoma (GP) in childhood and adolescence is a rare entity with scarce literature. We aimed to describe clinical features, biochemistry, radiology, genetics, management, and outcome in pediatric (≤ 20 years) GP. METHODS Retrospective record review of 18 pediatric GP patients from our center and systematic review including these and 77 from the literature (total cohort: 95). RESULTS GP constituted 20% of our pediatric prolactinoma cohort. In the total cohort (age: 15.4 ± 3.5 years), the majority (77, 82.8%) were males. Mass effect symptoms (88.6%), and pubertal delay/arrest in males (82.1%) were frequent. Median basal prolactin was 8649 (3246-17,532) ng/ml and the maximum tumor dimension was 5.5 ± 1.5 cm. MEN1 and AIP mutations were noted in 7 (21.9%) and 6 (18.8%) patients, respectively. Males with central hypogonadism had baseline bi-testicular volume of 20.2 ± 8.4 cc, lower LH than FSH (-2.04 ± 0.9 vs. -0.7 ± 1.6 SDS, p = 0.0075), and mostly, normal inhibin B. Majority (49/76, 64.5%) received dopamine agonist (DA) as first-line treatment with additional therapy in 35% (17/49). DA monotherapy arm had less frequent central hypothyroidism (42.9% vs 87.1%, p = 0.002) and central adrenal insufficiency (7.1% vs 66.7%, p = 0.0003) than multimodal therapy. A smaller tumor dimension (4.7 vs. 5.7 cm, p = 0.04) was associated with normoprolactinemia on DA monotherapy and AIP mutations (33.3% vs. nil, p = 0.02) with multimodal therapy. CONCLUSION GP is characterized by male predominance with frequent delay/arrest of puberty (82%), but relative sparing of the FSH-inhibin B axis in boys. DA monotherapy may be preferred as the first-line therapy in pediatric GP.
Collapse
Affiliation(s)
- Sandeep Kumar
- Department of Endocrinology and Metabolism, Seth G S Medical College & KEM Hospital, Parel, Mumbai, 400012, Maharashtra, India
| | - Vijaya Sarathi
- Department of Endocrinology, Vydehi Institute of Medical Sciences and Research Centre, Bangalore, India
| | - Anurag Ranjan Lila
- Department of Endocrinology and Metabolism, Seth G S Medical College & KEM Hospital, Parel, Mumbai, 400012, Maharashtra, India.
| | - Manjeetkaur Sehemby
- Department of Endocrinology and Metabolism, Seth G S Medical College & KEM Hospital, Parel, Mumbai, 400012, Maharashtra, India
| | - Saba Samad Memon
- Department of Endocrinology and Metabolism, Seth G S Medical College & KEM Hospital, Parel, Mumbai, 400012, Maharashtra, India
| | - Manjiri Karlekar
- Department of Endocrinology and Metabolism, Seth G S Medical College & KEM Hospital, Parel, Mumbai, 400012, Maharashtra, India
| | - Shilpa Sankhe
- Department of Radiology, Seth G S Medical College & KEM Hospital, Mumbai, India
| | - Virendra A Patil
- Department of Endocrinology and Metabolism, Seth G S Medical College & KEM Hospital, Parel, Mumbai, 400012, Maharashtra, India
| | - Nalini Shah
- Department of Endocrinology and Metabolism, Seth G S Medical College & KEM Hospital, Parel, Mumbai, 400012, Maharashtra, India
| | - Tushar Bandgar
- Department of Endocrinology and Metabolism, Seth G S Medical College & KEM Hospital, Parel, Mumbai, 400012, Maharashtra, India
| |
Collapse
|
43
|
Sahakian N, Appay R, Resseguier N, Graillon T, Piazzola C, Laure C, Figarella-Branger D, Régis J, Castinetti F, Brue T, Dufour H, Cuny T. Real-life clinical impact of a five-tiered classification of pituitary tumors. Eur J Endocrinol 2022; 187:893-904. [PMID: 36315463 DOI: 10.1530/eje-22-0812] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 10/31/2022] [Indexed: 11/08/2022]
Abstract
INTRODUCTION Usually benign, pituitary tumors (PT) can be invasive and aggressive with a propensity to progress and/or recur. Trouillas's clinicopathological classification attempts to predict the evolutionary risk of a PT. In this study, we assessed the prognostic value of this classification in an independent patient cohort and analyzed its impact on treatment strategies. PATIENTS AND METHODS In this study, 607 patients operated on between 2008 and 2018 for a PT were included. Grading was established based on invasion, proliferative activity (Ki-67, mitotic index) and p53 positivity. The therapeutic management following surgery was analyzed. Progression-free survival (PFS) of the graded tumors was estimated (Kaplan-Meier method and log-rank test) and a multivariate analysis was performed (Cox regression model). RESULTS Grading identified non-invasive PT without (grade 1a: 303 cases) or with proliferative activity (grade 1b: 53 cases) and invasive PT without (grade 2a: 202 cases) or with proliferative activity (grade 2b: 49 cases). The mean follow-up was 47 ± 30 months (median: 38 months). Progression/recurrence occurred in 127 cases. Grades were significant and independent predictors of PFS (P < 0.001) with a 4.8-fold higher risk of progression/recurrence in grade 2b as compared to grade 1a. As second-line therapy, gamma knife or conventional radiotherapy controlled tumor growth in 91.6 and 100% of cases, respectively, irrespective of the grade. Proliferative tumors exposed the patient to a 9.5-fold higher risk of having ≥3 adjuvant therapeutic lines as compared to non-proliferative tumors. DISCUSSION Grading of a PT according to Trouillas's classification predicts its risk of progression and should advocate for a personalized therapeutic approach in invasive and proliferative tumors. SIGNIFICANCE STATEMENT This is the first study to assess, on a cohort of 607 well-characterized patients, the real-life therapeutic impact of the five-tiered clinicopathological classification of pituitary tumors. First, we validate that pituitary tumor grades predict the evolutionary risk of the tumor, with a significant higher risk of progression/recurrence in invasive and/or proliferative tumors (mean follow-up: 47 ± 30 months, median: 38 months). Moreover, our study provides evidence that patients with proliferative tumors have a higher risk to be retreated after primary surgery and point toward the fact that radiotherapy can successfully control tumor growth in case of progression or recurrence. Our findings advocate for a personalized therapeutic approach in clinically aggressive pituitary tumors.
Collapse
Affiliation(s)
- Nicolas Sahakian
- Aix Marseille Univ, MMG, INSERM U1251, MARMARA Institute, APHM, Hôpital de la Conception, Department of Endocrinology, CRMR HYPO, Marseille, France
| | - Romain Appay
- APHM, CHU Timone, Service d'Anatomie Pathologique et de Neuropathologie, Marseille, France
| | - Noémie Resseguier
- Aix Marseille Univ, APHM, Hôpital de la Timone, Service d'Epidemiologie et d'Economie de la Santé, Unité de Recherche Clinique, Direction de la Recherche en Santé, Marseille, France
- EA3279, CEReSS, Health Service Research and Quality of Life Center, Aix-Marseille University, Marseille, France
| | - Thomas Graillon
- Aix Marseille University, MMG, INSERM U1251, MARMARA Institute, APHM, Hôpital de la Timone Department of Neurosurgery, CRMR HYPO, Marseille, France
| | - Cécilia Piazzola
- Aix Marseille Univ, MMG, INSERM U1251, MARMARA Institute, APHM, Hôpital de la Conception, Department of Endocrinology, CRMR HYPO, Marseille, France
| | - Cécilia Laure
- Department of Endocrinology, University Hospital of Montpellier, France
| | | | - Jean Régis
- Aix Marseille University, INS-UMR1106, Department of Functional and Stereotactic Neurosurgery, CHU Timone, Marseille, France
| | - Frédéric Castinetti
- Aix Marseille Univ, MMG, INSERM U1251, MARMARA Institute, APHM, Hôpital de la Conception, Department of Endocrinology, CRMR HYPO, Marseille, France
| | - Thierry Brue
- Aix Marseille Univ, MMG, INSERM U1251, MARMARA Institute, APHM, Hôpital de la Conception, Department of Endocrinology, CRMR HYPO, Marseille, France
| | - Henry Dufour
- Aix Marseille University, MMG, INSERM U1251, MARMARA Institute, APHM, Hôpital de la Timone Department of Neurosurgery, CRMR HYPO, Marseille, France
| | - Thomas Cuny
- Aix Marseille Univ, MMG, INSERM U1251, MARMARA Institute, APHM, Hôpital de la Conception, Department of Endocrinology, CRMR HYPO, Marseille, France
| |
Collapse
|
44
|
Melmed S, Kaiser UB, Lopes MB, Bertherat J, Syro LV, Raverot G, Reincke M, Johannsson G, Beckers A, Fleseriu M, Giustina A, Wass JAH, Ho KKY. Clinical Biology of the Pituitary Adenoma. Endocr Rev 2022; 43:1003-1037. [PMID: 35395078 PMCID: PMC9695123 DOI: 10.1210/endrev/bnac010] [Citation(s) in RCA: 124] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Indexed: 02/06/2023]
Abstract
All endocrine glands are susceptible to neoplastic growth, yet the health consequences of these neoplasms differ between endocrine tissues. Pituitary neoplasms are highly prevalent and overwhelmingly benign, exhibiting a spectrum of diverse behaviors and impact on health. To understand the clinical biology of these common yet often innocuous neoplasms, we review pituitary physiology and adenoma epidemiology, pathophysiology, behavior, and clinical consequences. The anterior pituitary develops in response to a range of complex brain signals integrating with intrinsic ectodermal cell transcriptional events that together determine gland growth, cell type differentiation, and hormonal production, in turn maintaining optimal endocrine health. Pituitary adenomas occur in 10% of the population; however, the overwhelming majority remain harmless during life. Triggered by somatic or germline mutations, disease-causing adenomas manifest pathogenic mechanisms that disrupt intrapituitary signaling to promote benign cell proliferation associated with chromosomal instability. Cellular senescence acts as a mechanistic buffer protecting against malignant transformation, an extremely rare event. It is estimated that fewer than one-thousandth of all pituitary adenomas cause clinically significant disease. Adenomas variably and adversely affect morbidity and mortality depending on cell type, hormone secretory activity, and growth behavior. For most clinically apparent adenomas, multimodal therapy controlling hormone secretion and adenoma growth lead to improved quality of life and normalized mortality. The clinical biology of pituitary adenomas, and particularly their benign nature, stands in marked contrast to other tumors of the endocrine system, such as thyroid and neuroendocrine tumors.
Collapse
Affiliation(s)
| | - Ursula B Kaiser
- Brigham & Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - M Beatriz Lopes
- University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Jerome Bertherat
- Université de Paris, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Luis V Syro
- Hospital Pablo Tobon Uribe and Clinica Medellin - Grupo Quirónsalud, Medellin, Colombia
| | - Gerald Raverot
- Hospices Civils de Lyon and Lyon 1 University, Lyon, France
| | - Martin Reincke
- University Hospital of LMU, Ludwig-Maximilians-Universität, Munich, Germany
| | - Gudmundur Johannsson
- Sahlgrenska University Hospital & Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | | | | | - Andrea Giustina
- San Raffaele Vita-Salute University and IRCCS Hospital, Milan, Italy
| | | | - Ken K Y Ho
- The Garvan Institute of Medical Research and St. Vincents Hospital, Sydney, Australia
| |
Collapse
|
45
|
Fleseriu M, Langlois F, Lim DST, Varlamov EV, Melmed S. Acromegaly: pathogenesis, diagnosis, and management. Lancet Diabetes Endocrinol 2022; 10:804-826. [PMID: 36209758 DOI: 10.1016/s2213-8587(22)00244-3] [Citation(s) in RCA: 105] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 08/01/2022] [Accepted: 08/09/2022] [Indexed: 11/23/2022]
Abstract
Growth hormone-secreting pituitary adenomas that cause acromegaly arise as monoclonal expansions of differentiated somatotroph cells and are usually sporadic. They are almost invariably benign, yet they can be locally invasive and show progressive growth despite treatment. Persistent excess of both growth hormone and its target hormone insulin-like growth factor 1 (IGF-1) results in a wide array of cardiovascular, respiratory, metabolic, musculoskeletal, neurological, and neoplastic comorbidities that might not be reversible with disease control. Normalisation of IGF-1 and growth hormone are the primary therapeutic aims; additional treatment goals include tumour shrinkage, relieving symptoms, managing complications, reducing excess morbidity, and improving quality of life. A multimodal approach with surgery, medical therapy, and (more rarely) radiation therapy is required to achieve these goals. In this Review, we examine the epidemiology, pathogenesis, diagnosis, complications, and treatment of acromegaly, with an emphasis on the importance of tailoring management strategies to each patient to optimise outcomes.
Collapse
Affiliation(s)
- Maria Fleseriu
- Department of Medicine (Division of Endocrinology, Diabetes and Clinical Nutrition) and Department of Neurological Surgery, and Pituitary Center, Oregon Health & Science University, Portland, OR, USA.
| | - Fabienne Langlois
- Division of Endocrinology, Department of Medicine, Centre Intégré Universitaire de Santé et de Services Sociaux de l'Estrie-Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada
| | | | - Elena V Varlamov
- Department of Medicine (Division of Endocrinology, Diabetes and Clinical Nutrition) and Department of Neurological Surgery, and Pituitary Center, Oregon Health & Science University, Portland, OR, USA
| | - Shlomo Melmed
- Department of Medicine and Pituitary Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
46
|
Zheng AC, Wang EJ, Aghi MK. Recent advancements in the molecular biology of pituitary adenomas. Expert Rev Endocrinol Metab 2022; 17:293-304. [PMID: 35702013 DOI: 10.1080/17446651.2022.2082942] [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: 03/12/2022] [Accepted: 05/24/2022] [Indexed: 10/18/2022]
Abstract
INTRODUCTION Pituitary adenomas are a common and diverse group of intracranial tumors arising from the anterior pituitary that are usually slow-growing and benign, but still pose a significant healthcare burden to patients. Additionally, they are increasing in both incidence and prevalence, leading to a need for better understanding of molecular changes in the development of these tumors. AREAS COVERED A PubMed literature search was conducted using the terms 'pituitary adenoma' in combination with keywords related to secretory subtype: lactotroph, somatotroph, corticotroph, gonadotroph and null cell, in addition to their transcription factor expression: PIT1, TPIT, and SF-1. Articles resulting from this search were analyzed, as well as relevant articles cited as their references. In this review, we highlight recent advances in the genetic and epigenetic characterization of individual pituitary adenoma subtypes and the effect it may have on guiding future clinical treatment of these tumors. EXPERT OPINION Understanding the molecular biology of pituitary adenomas is a fundamental step toward advancing the treatment of these tumors. Yet crucial knowledge gaps exist in our understanding of the underlying molecular biology of pituitary adenomas which can potentially be addressed by turning to differentially activated molecular pathways in tumor relative to normal gland.
Collapse
Affiliation(s)
- Allison C Zheng
- Department of Neurosurgery; University of California at San Francisco (UCSF) San Francisco, CA, USA
| | - Elaina J Wang
- Department of Neurosurgery; Warren Alpert Medical School of Brown University Providence, RI, USA
| | - Manish K Aghi
- Department of Neurosurgery; University of California at San Francisco (UCSF) San Francisco, CA, USA
| |
Collapse
|
47
|
Vasilakis IA, Paltoglou G, Gavra M, Charmandari E. A rare case of a giant prolactinoma with atypical histological features: 5 years of follow-up. Hormones (Athens) 2022; 21:323-327. [PMID: 35143036 DOI: 10.1007/s42000-022-00350-5] [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: 06/14/2021] [Accepted: 01/10/2022] [Indexed: 11/04/2022]
Abstract
BACKGROUND Giant prolactinomas are rare in childhood and adolescence and represent a challenge in diagnosis and management. CASE PRESENTATION A 15.7-year-old male adolescent presented with short stature and delayed puberty. On clinical examination, mild right central VII paresis, gait instability, decreased visual acuity, and impaired visual fields were noted. Investigations showed hyperprolactinemia (2209 ng/mL), secondary hypothyroidism, hypogonadotropic hypogonadism, and growth hormone deficiency. Imaging studies showed an enormous invasive skull base mass. Craniotomy was undertaken to debulk the tumor and perform biopsies. Histology revealed a very large atypical, prolactin-secreting pituitary macroadenoma, i.e., a giant prolactinoma. After commencing cabergoline treatment, prolactin concentrations decreased in 5 months and normalized 18 months later, while significant shrinkage of the tumor was observed. The diagnostic work-up for genetic syndromes often associated with sporadic macroadenomas was negative. CONCLUSION Giant prolactinomas presenting with multiple pituitary hormone deficiencies in childhood or adolescence are rare and require prompt diagnosis and multidisciplinary management.
Collapse
Affiliation(s)
- Ioannis-Anargyros Vasilakis
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, 'Aghia Sophia' Children's Hospital, Thivon & Levadeias Str., 11527, Athens, Greece
| | - George Paltoglou
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, 'Aghia Sophia' Children's Hospital, Thivon & Levadeias Str., 11527, Athens, Greece.
- University Research Institute of Maternal and Child Health & Precision Medicine, Thivon & Levadeias Str., 11527, Athens, Greece.
| | - Maria Gavra
- Division of CT and MRI, Radiology Department, 'Aghia Sophia' Children's Hospital, Thivon & Levadeias Str., 11527, Athens, Greece
| | - Evangelia Charmandari
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, 'Aghia Sophia' Children's Hospital, Thivon & Levadeias Str., 11527, Athens, Greece
- Division of Endocrinology and Metabolism, Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Ephessiou Str., 11527, Athens, Greece
| |
Collapse
|
48
|
Current and Emerging Medical Therapies in Pituitary Tumors. J Clin Med 2022; 11:jcm11040955. [PMID: 35207228 PMCID: PMC8877616 DOI: 10.3390/jcm11040955] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/01/2022] [Accepted: 02/10/2022] [Indexed: 12/04/2022] Open
Abstract
Pituitary tumors (PT) represent in, the majority of cases, benign tumors for which surgical treatment still remains, except for prolactin-secreting PT, the first-line therapeutic option. Nonetheless, the role played by medical therapies for the management of such tumors, before or after surgery, has evolved considerably, due in part to the recent development of well-tolerated and highly efficient molecules. In this review, our aim was to present a state-of-the-art of the current medical therapies used in the field of PT and the benefits and caveats for each of them, and further specify their positioning in the therapeutic algorithm of each phenotype. Finally, we discuss the future of PT medical therapies, based on the most recent studies published in this field.
Collapse
|
49
|
Cozzi R, Ambrosio MR, Attanasio R, Battista C, Bozzao A, Caputo M, Ciccarelli E, De Marinis L, De Menis E, Faustini Fustini M, Grimaldi F, Lania A, Lasio G, Logoluso F, Losa M, Maffei P, Milani D, Poggi M, Zini M, Katznelson L, Luger A, Poiana C. Italian Association of Clinical Endocrinologists (AME) and International Chapter of Clinical Endocrinology (ICCE). Position statement for clinical practice: prolactin-secreting tumors. Eur J Endocrinol 2022; 186:P1-P33. [PMID: 35000899 PMCID: PMC8859924 DOI: 10.1530/eje-21-0977] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 01/06/2022] [Indexed: 12/03/2022]
Abstract
Prolactinomas are the most frequent pituitary adenomas. Prolactinoma may occur in different clinical settings and always require an individually tailored approach. This is the reason why a panel of Italian neuroendocrine experts was charged with the task to provide indications for the diagnostic and therapeutic approaches that can be easily applied in different contexts. The document provides 15 recommendations for diagnosis and 54 recommendations for treatment, issued according to the GRADE system. The level of agreement among panel members was formally evaluated by RAND-UCLA methodology. In the last century, prolactinomas represented the paradigm of pituitary tumors for which the development of highly effective drugs obtained the best results, allowing to avoid neurosurgery in most cases. The impressive improvement of neurosurgical endoscopic techniques allows a far better definition of the tumoral tissue during surgery and the remission of endocrine symptoms in many patients with pituitary tumors. Consequently, this refinement of neurosurgery is changing the therapeutic strategy in prolactinomas, allowing the definitive cure of some patients with permanent discontinuation of medical therapy.
Collapse
Affiliation(s)
- Renato Cozzi
- Division of Endocrinology, Niguarda Hospital, Milan, Italy
- Correspondence should be addressed to R Cozzi;
| | - Maria Rosaria Ambrosio
- Section of Endocrinology and Internal Medicine, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | | | - Claudia Battista
- Endocrinology Unit, IRCCS Casa Sollievo della Sofferenza Hospital, San Giovanni Rotondo (FG), Italy
| | - Alessandro Bozzao
- Neuroradiology, S. Andrea Hospital, NESMOS Department (Neuroscience, Mental Health, Sensorial Organs), Sapienza University of Rome, Rome, Italy
| | - Marco Caputo
- Laboratorio Analisi Cliniche e Microbiologia, Synlab SRL, Calenzano, Florence, Italy
| | | | - Laura De Marinis
- Pituitary Unit, Department of Endocrinology, Catholic University of the Sacred Heart, School of Medicine, Rome, Italy
| | | | | | - Franco Grimaldi
- AME President, Endocrinology and Metabolism Unit, University Hospital S. Maria della Misericordia, Udine, Italy
| | - Andrea Lania
- Department of Biomedical Sciences, Endocrinology Unit, Rozzano, Italy
| | - Giovanni Lasio
- Department of Neurosurgery, Humanitas Clinical and Research Center IRCCS, Rozzano, Italy
| | | | - Marco Losa
- Department of Neurosurgery and Gamma Knife Radiosurgery, IRCCS San Raffaele Scientific Institute, Vita-Salute University, Milan, Italy
| | - Pietro Maffei
- Department of Medicine (DIMED), 3rd Medical Clinic, Padua University Hospital, Padua, Italy
| | - Davide Milani
- Department of Neurosurgery, Humanitas Clinical and Research Center IRCCS, Rozzano, Italy
| | - Maurizio Poggi
- Endocrinology, Department of Clinical and Molecular Medicine, S. Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Michele Zini
- Endocrinology Unit, Azienda Ospedaliera S. Maria Nuova IRCCS, Reggio Emilia, Italy
| | | | - Anton Luger
- Division of Endocrinology and Metabolism, Medical University of Vienna, Vienna, Austria
| | - Catalina Poiana
- ‘Carol Davila’ University of Medicine and Pharmacy – Endocrinology, “C.I. Parhon” National Institute of Endocrinology – Pituitary and Neuroendocrine Disorders, Bucharest, Romania
| | | |
Collapse
|
50
|
Ershadinia N, Tritos NA. Diagnosis and Treatment of Acromegaly: An Update. Mayo Clin Proc 2022; 97:333-346. [PMID: 35120696 DOI: 10.1016/j.mayocp.2021.11.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 10/16/2021] [Accepted: 11/04/2021] [Indexed: 01/01/2023]
Abstract
Acromegaly is typically caused by a growth hormone-secreting pituitary adenoma, driving excess secretion of insulin-like growth factor 1. Acromegaly may result in a variety of cardiovascular, respiratory, endocrine, metabolic, musculoskeletal, and neoplastic comorbidities. Early diagnosis and adequate treatment are essential to mitigate excess mortality associated with acromegaly. PubMed searches were conducted using the keywords growth hormone, acromegaly, pituitary adenoma, diagnosis, treatment, pituitary surgery, medical therapy, and radiation therapy (between 1981 and 2021). The diagnosis of acromegaly is confirmed on biochemical grounds, including elevated serum insulin-like growth factor 1 and lack of growth hormone suppression after glucose administration. Pituitary magnetic resonance imaging is advised in patients with acromegaly to identify an underlying pituitary adenoma. Transsphenoidal pituitary surgery is generally first-line therapy for patients with acromegaly. However, patients with larger and invasive tumors (macroadenomas) are often not in remission postoperatively. Medical therapies, including somatostatin receptor ligands, cabergoline, and pegvisomant, can be recommended to patients with persistent disease after surgery. Select patients may also be candidates for preoperative medical therapy. In addition, primary medical therapy has a role for patients without mass effect on the optic chiasm who are unlikely to be cured by surgery. Clinical, endocrine, imaging, histologic, and molecular markers may help predict the response to medical therapy; however, confirmation in prospective studies is needed. Radiation therapy is usually a third-line option and is increasingly administered by a variety of stereotactic techniques. An improved understanding of the pathogenesis of acromegaly may ultimately lead to the design of novel, efficacious therapies for this serious condition.
Collapse
Affiliation(s)
- Nazanin Ershadinia
- Neuroendocrine Unit and Neuroendocrine and Pituitary Tumor Clinical Center, Massachusetts General Hospital, Boston
| | - Nicholas A Tritos
- Neuroendocrine Unit and Neuroendocrine and Pituitary Tumor Clinical Center, Massachusetts General Hospital, Boston; Harvard Medical School, Boston, MA.
| |
Collapse
|