1
|
Chinezu L, Gliga MC, Borz MB, Gliga C, Pascanu IM. Clinical Implications of Molecular and Genetic Biomarkers in Cushing's Disease: A Literature Review. J Clin Med 2025; 14:3000. [PMID: 40364036 PMCID: PMC12072580 DOI: 10.3390/jcm14093000] [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: 03/30/2025] [Revised: 04/23/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
Cushing's disease (CD) is a rare disorder caused by adrenocorticotropic hormone (ACTH)-secreting pituitary neuroendocrine tumors, which lead to chronic hypercortisolism and significant complications with increased mortality. These tumors are characterized by a substantial heterogeneity in their biological behavior, prognosis, and therapeutic response, making their management challenging. While transsphenoidal surgery remains the first-line treatment, recurrence rates remain high, and alternative therapeutic approaches, such as pharmacological therapy and radiotherapy, have a variable efficacy and are frequently limited due to side effects. Increasing evidence suggests that molecular biomarkers, both immunohistochemical and genetic, may play an important role in predicting a tumor's aggressiveness, recurrence risk, and response to targeted therapies. The immunohistochemical evaluation of its granulation pattern, Ki-67 proliferation index, and E-cadherin expressions have been linked to a tumor's invasiveness and surgical outcomes, while somatostatin and dopamine receptor expressions may influence its response to Pasireotide and cabergoline therapy. Genetic alterations such as USP8 mutations impact tumor growth and its response to targeted therapies, whereas CABLES1 and TP53 alterations may contribute to more aggressive tumor behavior. Despite these findings, the clinical applicability of many of these markers remains limited by inconsistent validation and lack of standardized cutoff values. This narrative review provides an update on the latest evidence regarding the roles of molecular biomarkers in corticotropinomas, emphasizing their role in prognosis, recurrence risk, and the response to different treatment options. A better understanding and integration of these biomarkers into clinical practice could lead to a better patient stratification, more efficient therapeutic strategies, and personalized treatment approaches for patients with CD.
Collapse
Affiliation(s)
- Laura Chinezu
- Department of Histology, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540142 Targu Mures, Romania; (L.C.); (C.G.)
| | - Maximilian Cosma Gliga
- Department of Endocrinology, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540142 Targu Mures, Romania;
| | - Mihnea Bogdan Borz
- Department of Anatomy, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540142 Targu Mures, Romania;
| | - Camelia Gliga
- Department of Histology, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540142 Targu Mures, Romania; (L.C.); (C.G.)
| | - Ionela Maria Pascanu
- Department of Endocrinology, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540142 Targu Mures, Romania;
| |
Collapse
|
2
|
Tataranu LG. A Brief Overview of Molecular Biology in Pituitary Adenomas with a Focus on Aggressive Lesions. Int J Mol Sci 2025; 26:3717. [PMID: 40332351 PMCID: PMC12027641 DOI: 10.3390/ijms26083717] [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: 11/15/2024] [Revised: 12/16/2024] [Accepted: 12/18/2024] [Indexed: 05/08/2025] Open
Abstract
Arising from the anterior lobe of the hypophysis, pituitary neuroendocrine tumors (PitNETs), previously known as pituitary adenomas, constitute an intricate and heterogeneous entity. Although they are defined as benign pathology, these tumors can often invade neighboring structures and demonstrate aggressive behavior. The continuous advancement of molecular biology has begun to shed light on the genetic mutations behind the development and evolution of this pathology, providing a better understanding. Notwithstanding, the rising occurrence in recent decades can mainly be attributed to advanced diagnostic techniques; however, these tumors continue to increase in prevalence and incidence, creating a heavy burden on healthcare systems. Consequently, the need for further studies is dire, primarily due to a lack of tailored therapeutic approaches. Thus, this article aims to offer a brief overview of the molecular biology behind these complex tumors to contribute, even on a small scale, to more comprehensive care.
Collapse
Affiliation(s)
- Ligia Gabriela Tataranu
- Department of Neurosurgery, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania;
- Department of Neurosurgery, Bagdasar-Arseni Emergency Clinical Hospital, 041915 Bucharest, Romania
| |
Collapse
|
3
|
Pofi R, De Alcubierre D, Dong J, Tomlinson JW. New Approaches to the Treatment of Hypercortisolism. Annu Rev Med 2025; 76:431-445. [PMID: 39485832 DOI: 10.1146/annurev-med-071723-044849] [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] [Indexed: 11/03/2024]
Abstract
This review explores the evolving landscape of treatments for hypercortisolism, highlighting both established and emerging therapies. Although surgery remains the cornerstone of management, medical therapies play a crucial and expanding role, especially in cases of persistent, recurrent, or severe hypercortisolism. We discuss the effectiveness and limitations of steroidogenesis inhibitors, pituitary-directed drugs, glucocorticoid receptor antagonists, and experimental drugs targeting novel molecular pathways that have been implicated in the pathogenesis of hypercortisolism. Despite advancements, significant unmet needs persist, underscoring the importance of personalized treatment approaches and the development of targeted therapies. Ongoing and future clinical trials are crucial for validating the safety and efficacy of these innovative treatments in Cushing disease management.
Collapse
Affiliation(s)
- Riccardo Pofi
- Oxford Centre for Diabetes, Endocrinology and Metabolism and National Institute for Health and Care Research (NIHR) Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford, United Kingdom;
| | - Dario De Alcubierre
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Jiawen Dong
- Oxford Centre for Diabetes, Endocrinology and Metabolism and National Institute for Health and Care Research (NIHR) Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford, United Kingdom;
| | - Jeremy W Tomlinson
- Oxford Centre for Diabetes, Endocrinology and Metabolism and National Institute for Health and Care Research (NIHR) Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford, United Kingdom;
| |
Collapse
|
4
|
Agrawal N, Sarkar D, Chatterjee S, Sinha P, Bhattacharya R, Ranjan P, Chakraborty PP, Bhattacharjee R. Successful management of refractory Cushing's disease with severe hypercortisolemia using etomidate and temozolomide in post-surgical failure. J R Coll Physicians Edinb 2024; 54:304-309. [PMID: 39474897 DOI: 10.1177/14782715241293817] [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] [Indexed: 12/15/2024] Open
Abstract
Corticotropinomas account for 20% of all aggressive pituitary tumors and pituitary carcinomas and are associated with high mortality. These tumors not only cause neurovascular compromise but can also be fatal due to severe hypercortisolemia itself. Although surgery is considered the primary treatment modality, it is often partially successful or unsuccessful. Moreover, these tumors frequently recur and may be resistant to conventional treatments, including surgery and radiotherapy. Therefore, early multimodal treatment and regular follow-up are necessary. We present a case of aggressive Cushing's disease managed with combined temozolomide therapy and radiotherapy following an unsuccessful transsphenoidal surgery, resulting in significant long-term radiological and biochemical remission. In addition, etomidate infusion was administered to achieve rapid cortisol reduction, highlighting its role as a bridging therapy to other modalities in treating life-threatening and severe hypercortisolemia outside an intensive care setting.
Collapse
Affiliation(s)
- Neeti Agrawal
- Department of Endocrinology and Metabolism, Medical College and Hospital, Kolkata, India
| | - Debalina Sarkar
- Department of Endocrinology and Metabolism, Medical College and Hospital, Kolkata, India
| | - Subhankar Chatterjee
- Department of Endocrinology and Metabolism, Medical College and Hospital, Kolkata, India
| | - Pallab Sinha
- Department of Endocrinology and Metabolism, Medical College and Hospital, Kolkata, India
| | - Ranita Bhattacharya
- Department of Endocrinology and Metabolism, Medical College and Hospital, Kolkata, India
| | - Prabhat Ranjan
- Department of Endocrinology and Metabolism, Medical College and Hospital, Kolkata, India
| | | | - Rana Bhattacharjee
- Department of Endocrinology and Metabolism, Medical College and Hospital, Kolkata, India
| |
Collapse
|
5
|
Facundo AN, Magalhães M, Nascimento GC, Azulay RS, Santos RM, Freitas LA, Nascimento AGPAC, Rodrigues VP, Santos WC, Beckman AMGS, Abreu JMF, Silva RP, Carneiro EL, Oliveira Neto CP, Gil da Costa RM, Corcoy R, Mato E, Faria MS. The expression of VDACs and Bcl2 family genes in pituitary adenomas: clinical correlations and postsurgical outcomes. Front Endocrinol (Lausanne) 2024; 15:1481050. [PMID: 39449743 PMCID: PMC11499145 DOI: 10.3389/fendo.2024.1481050] [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: 08/15/2024] [Accepted: 09/13/2024] [Indexed: 10/26/2024] Open
Abstract
Introduction Pituitary adenomas (PAs) are benign tumors with high prevalence and, occasionally, aggressive course. The tumorigenesis of these lesions is not completely understood at the molecular level. BAK1 and BAX proteins play fundamental roles in apoptosis and seem to interact with VDAC proteins, whose expressions have been markedly altered in cancer, impacting their prognosis. Objective to evaluate the gene expression of VDAC1, VDAC2, BAK1 and BAX and their association with clinical and imaging characteristics in PA. Methods Clinical-epidemiological data were collected from 117 tumor samples from patients affected by PA. Invasiveness was assessed by the Knosp scale. Gene expression was examined by real-time PCR. Relative expression analysis was performed by 2^(-DDCt) method. Results The sample was mainly composed of women (69/117 - 57.2%). Tumor subtypes observed were Non-Functioning (NF) (73/117 - 62.4%), Acromegaly (24/117 - 20.5%) and Cushing's Disease (CD) (20/117 - 17.1%). Compared to normal tissue, there was a significant reduction in VDAC1 expression in the Acromegaly (p=0.029) and NF (p=0.002) groups. BAX expression was lower in all groups (p <0.001; p=0.007; P =0.005). No difference was found in VDAC2 and BAK1 expression, compared to normal pituitary. Overexpression of VDAC2 occurred in PAs with post-surgical regrowth (p=0.042). A strongly negative correlation was observed in BAX and BAK1 expression in CD. Conclusion The results indicated that downregulations of VDAC1 and BAX may be related to resistance to apoptosis. In contrast, overexpression of VDAC2 in regrowing PAs suggests an antiapoptotic role for this gene. In summary, the genes evaluated might be involved in the biopathology of PAs.
Collapse
Affiliation(s)
- AN Facundo
- Post-Graduate Program in Adult Health (PPGSAD), Federal University of Maranhão (UFMA), São Luis, Brazil
- Service of Endocrinology, University Hospital of the Federal University of Maranhao (HUUFMA), São Luis, Brazil
- Research Group in Clinical and Molecular Endocrinology and Metabology (ENDOCLIM), Federal University of Maranhão (UFMA), São Luis, Brazil
| | - M Magalhães
- Research Group in Clinical and Molecular Endocrinology and Metabology (ENDOCLIM), Federal University of Maranhão (UFMA), São Luis, Brazil
| | - GC Nascimento
- Service of Endocrinology, University Hospital of the Federal University of Maranhao (HUUFMA), São Luis, Brazil
- Research Group in Clinical and Molecular Endocrinology and Metabology (ENDOCLIM), Federal University of Maranhão (UFMA), São Luis, Brazil
| | - RS Azulay
- Service of Endocrinology, University Hospital of the Federal University of Maranhao (HUUFMA), São Luis, Brazil
- Research Group in Clinical and Molecular Endocrinology and Metabology (ENDOCLIM), Federal University of Maranhão (UFMA), São Luis, Brazil
| | - RM Santos
- Service of Radiology, University Hospital of the Federal University of Maranhao (HUUFMA), São Luis, Brazil
| | - LA Freitas
- Service of Radiology, University Hospital of the Federal University of Maranhao (HUUFMA), São Luis, Brazil
| | - AGPAC Nascimento
- Research Group in Clinical and Molecular Endocrinology and Metabology (ENDOCLIM), Federal University of Maranhão (UFMA), São Luis, Brazil
- Service of Pathology, University Hospital of the Federal University of Maranhao (HUUFMA), São Luis, Brazil
| | - VP Rodrigues
- Research Group in Clinical and Molecular Endocrinology and Metabology (ENDOCLIM), Federal University of Maranhão (UFMA), São Luis, Brazil
- Department of Morphology, Federal University of Maranhao (UFMA), São Luis, Brazil
| | - WC Santos
- Post-Graduate Program in Adult Health (PPGSAD), Federal University of Maranhão (UFMA), São Luis, Brazil
- Research Group in Clinical and Molecular Endocrinology and Metabology (ENDOCLIM), Federal University of Maranhão (UFMA), São Luis, Brazil
| | - AMGS Beckman
- Research Group in Clinical and Molecular Endocrinology and Metabology (ENDOCLIM), Federal University of Maranhão (UFMA), São Luis, Brazil
| | - JMF Abreu
- Service of Endocrinology, University Hospital of the Federal University of Maranhao (HUUFMA), São Luis, Brazil
- Research Group in Clinical and Molecular Endocrinology and Metabology (ENDOCLIM), Federal University of Maranhão (UFMA), São Luis, Brazil
| | - RP Silva
- Research Group in Clinical and Molecular Endocrinology and Metabology (ENDOCLIM), Federal University of Maranhão (UFMA), São Luis, Brazil
| | - EL Carneiro
- Service of Neurosurgery, University Hospital of the Federal University of Maranhao (HUUFMA), São Luis, Brazil
| | - CP Oliveira Neto
- Service of Endocrinology, University Hospital of the Federal University of Maranhao (HUUFMA), São Luis, Brazil
- Research Group in Clinical and Molecular Endocrinology and Metabology (ENDOCLIM), Federal University of Maranhão (UFMA), São Luis, Brazil
| | - RM Gil da Costa
- Post-Graduate Program in Adult Health (PPGSAD), Federal University of Maranhão (UFMA), São Luis, Brazil
- Research Group in Clinical and Molecular Endocrinology and Metabology (ENDOCLIM), Federal University of Maranhão (UFMA), São Luis, Brazil
- Department of Morphology, Federal University of Maranhao (UFMA), São Luis, Brazil
| | - R Corcoy
- CIBER Bioengineering, Biomaterials and Nanotechnology (CIBER-BBN), Instituto de Salud III, Madrid, Spain
- Department of Nutricion and Endocrinology of Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - E Mato
- CIBER Bioengineering, Biomaterials and Nanotechnology (CIBER-BBN), Instituto de Salud III, Madrid, Spain
- Department of Nutricion and Endocrinology of Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - MS Faria
- Post-Graduate Program in Adult Health (PPGSAD), Federal University of Maranhão (UFMA), São Luis, Brazil
- Service of Endocrinology, University Hospital of the Federal University of Maranhao (HUUFMA), São Luis, Brazil
- Research Group in Clinical and Molecular Endocrinology and Metabology (ENDOCLIM), Federal University of Maranhão (UFMA), São Luis, Brazil
| |
Collapse
|
6
|
Sahakian N, Goetz L, Appay R, Graillon T, Raingeard I, Piazzola C, Regis J, Castinetti F, Brue T, Dufour H, Cuny T. Outcome of non-functioning ACTH pituitary tumors: silent does not mean indolent. Pituitary 2024; 27:644-653. [PMID: 38995473 DOI: 10.1007/s11102-024-01428-6] [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: 07/03/2024] [Indexed: 07/13/2024]
Abstract
INTRODUCTION Silent corticotroph tumors (siACTH) represent a rare entity of pituitary tumors (PT), usually more aggressive than other PT. Few predictor factors of recurrence in the post-operative period have been proposed until now. This study aimed (1) to evaluate the clinical outcome of siACTH after surgery according to a five-tiered clinicopathological classification (2) to compare siACTH characteristics to ACTH-secreting macroadenomas (macroCD), and silent gonadotropinomas (siLH/FSH). PATIENTS AND METHODS Between 2008 and 2022, 29 siACTH out of 865 PT cases operated in one tertiary center were included. Clinical, paraclinical, histological, and surgical data were collected and compared to 25 macroCD and 143 siLH/FSH cases, respectively. The tumor grading was established according to both invasion (no = 1; yes = 2) and proliferation (no = a; yes = b). Progression-free survival was estimated using Kaplan-Meier method and log-rank test. RESULTS We identified 15 (51.7%) grade 1a, 11 (37.9%) grade 2a and 3 (10.3%) grade 2b siACTH with a trend for a 7-fold-time higher risk of progression/recurrence in grade 2b as compared to 1a (p = 0.06). The repartition of tumor grades was similar between the three subgroups, however a 5.7-fold-higher risk of progression was observed in grade 1a siACTH than in grade 1a siLH/FSH (p = 0.02). Compared to siLH/FSH, higher ACTH levels may help to preoperatively identify siACTH. CONCLUSION The five-tiered clinicopathological classification contribute to predict the risk of recurrence of operated siACTH tumors. Noteworthy, non-invasive and non-proliferative siACTH exhibit a less favorable outcomes than their siLH/FSH counterparts, which should prompt for a personalized follow up.
Collapse
Affiliation(s)
- Nicolas Sahakian
- Aix Marseille Univ, APHM, INSERM, MMG, Marmara Institute, La Conception University Hospital, Department of Endocrinology, CRMR HYPO, Marseille, France
| | - Lise Goetz
- Department of Endocrinology, University Hospital of Montpellier, Montpellier, France
| | - Romain Appay
- Department of Pathological Anatomy and Neuropathology, Timone Hospital, APHM, Marseille, France
| | - Thomas Graillon
- Aix Marseille Univ, APHM, INSERM, MMG, Marmara Institute, Timone Hospital, Department of Neurosurgery, CRMR HYPO, Marseille, France
| | - Isabelle Raingeard
- Department of Endocrinology, University Hospital of Montpellier, Montpellier, France
| | - Cécilia Piazzola
- Aix Marseille Univ, APHM, INSERM, MMG, Marmara Institute, La Conception University Hospital, Department of Endocrinology, CRMR HYPO, Marseille, France
| | - Jean Regis
- Department of Functional and Stereotactic Neurosurgery, CHU Timone, Aix Marseille University, INS-UMR1106, Marseille, 13385, France
| | - Frédéric Castinetti
- Aix Marseille Univ, APHM, INSERM, MMG, Marmara Institute, La Conception University Hospital, Department of Endocrinology, CRMR HYPO, Marseille, France
| | - Thierry Brue
- Aix Marseille Univ, APHM, INSERM, MMG, Marmara Institute, La Conception University Hospital, Department of Endocrinology, CRMR HYPO, Marseille, France
| | - Henry Dufour
- Aix Marseille Univ, APHM, INSERM, MMG, Marmara Institute, Timone Hospital, Department of Neurosurgery, CRMR HYPO, Marseille, France
| | - Thomas Cuny
- Aix Marseille Univ, APHM, INSERM, MMG, Marmara Institute, La Conception University Hospital, Department of Endocrinology, CRMR HYPO, Marseille, France.
- Aix Marseille Univ, Hôpital de la Conception, Clinical Investigation Centre 1409, Inserm, Marseille, France.
- Service d'Endocrinologie, Hôpital de la Conception, APHM, 147 Boulevard Baille, Marseille, 13005, France.
| |
Collapse
|
7
|
Li Y, Ren X, Gao W, Cai R, Wu J, Liu T, Chen X, Jiang D, Chen C, Cheng Q, Wu A, Cheng W. The biological behavior and clinical outcome of pituitary adenoma are affected by the microenvironment. CNS Neurosci Ther 2024; 30:e14729. [PMID: 38738958 PMCID: PMC11090080 DOI: 10.1111/cns.14729] [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: 07/22/2023] [Revised: 02/25/2024] [Accepted: 03/31/2024] [Indexed: 05/14/2024] Open
Abstract
BACKGROUND Pituitary adenoma is one of the most common brain tumors. Most pituitary adenomas are benign and can be cured by surgery and/or medication. However, some pituitary adenomas show aggressive growth with a fast growth rate and are resistant to conventional treatments such as surgery, drug therapy, and radiation therapy. These tumors, referred to as refractory pituitary adenomas, often relapse or regrow in the early postoperative period. The tumor microenvironment (TME) has recently been identified as an important factor affecting the biological manifestations of tumors and acts as the main battlefield between the tumor and the host immune system. MAIN BODY In this review, we focus on describing TME in pituitary adenomas and refractory pituitary adenomas. Research on the immune microenvironment of pituitary adenomas is currently focused on immune cells such as macrophages and lymphocytes, and extensive research and experimental verifications are still required regarding other components of the TME. In particular, studies are needed to determine the role of the TME in the specific biological behaviors of refractory pituitary adenomas, such as high invasion, fast recurrence rate, and high tolerance to traditional treatments and to identify the mechanisms involved. CONCLUSION Overall, we summarize the similarities and differences between the TME of pituitary adenomas and refractory pituitary adenomas as well as the changes in the biological behavior of pituitary adenomas that may be caused by the microenvironment. These changes greatly affect the outcome of patients.
Collapse
Affiliation(s)
- Yuhe Li
- Department of NeurosurgeryShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Xiufang Ren
- Department of PathologyShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Wei Gao
- Department of NeurosurgeryShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Ruikai Cai
- Department of NeurosurgeryShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Jianqi Wu
- Department of NeurosurgeryShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Tianqi Liu
- Department of NeurosurgeryShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Xin Chen
- Department of NeurosurgeryShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Daoming Jiang
- Shenyang ShenDa Endoscopy Co., Ltd.ShenyangLiaoningChina
| | - Chong Chen
- Shenyang ShenDa Endoscopy Co., Ltd.ShenyangLiaoningChina
| | - Quan Cheng
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaHunanChina
- National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Anhua Wu
- Department of NeurosurgeryShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Wen Cheng
- Department of NeurosurgeryShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| |
Collapse
|
8
|
Kanzawa M, Shichi H, Kanie K, Yamamoto M, Yamamoto N, Tsujimoto Y, Bando H, Iguchi G, Kitano S, Inoshita N, Yamada S, Ogawa W, Itoh T, Fukuoka H. Effects of the Cortisol Milieu on Tumor-Infiltrating Immune Cells in Corticotroph Tumors. Endocrinology 2024; 165:bqae016. [PMID: 38340329 DOI: 10.1210/endocr/bqae016] [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: 08/01/2023] [Revised: 01/31/2024] [Accepted: 02/05/2024] [Indexed: 02/12/2024]
Abstract
CONTEXT Corticotrophs are susceptible to lymphocyte cytotoxicity, as seen in hypophysitis, suggesting that an immunological approach may be a potential strategy for corticotroph-derived tumors. OBJECTIVE We aimed to clarify whether corticotroph tumors that induce hypercortisolemia (ACTHomas) could be targets for immunotherapy. METHODS Tumor-infiltrating immune cells were immunohistochemically analyzed. ACTHomas were compared with other pituitary tumors, and further divided into 3 different cortisol-exposed milieus: Naïve (ACTHomas without preoperative treatment), Met (ACTHomas with preoperative metyrapone), and SCA (silent corticotroph adenomas). A 3-dimensional cell culture of resected tumors was used to analyze the effects of immune checkpoint inhibitors. RESULTS The number of tumor-infiltrating lymphocytes (TILs) was low in ACTHomas. Among these, the number of CD8+ cells was lower in ACTHomas than in both somatotroph and gonadotroph tumors (both P < .01). Then we compared the differences in TILs among Naïve, Met, and SCA. The number of CD4+ cells, but not CD8+ cells, was higher in both Met and SCA than in Naïve. Next, we investigated tumor-associated macrophages, which could negatively affect T cell infiltration. The numbers of CD163+ and CD204+ cells were positively associated with cortisol levels. Moreover, tumor size was positively correlated with the number of CD204+ cells. CONCLUSION We found the possibility that ACTHomas were immunologically cold in a cortisol-independent manner. In contrast, the tumor infiltration of CD4+ cells and M2-macrophages were associated with the cortisol milieu. Future studies are needed to validate these results and develop effective immunotherapy while considering the cortisol milieu.
Collapse
Affiliation(s)
- Maki Kanzawa
- Department of Diagnostic Pathology, Kobe University Hospital, Kobe, 650-0017, Japan
| | - Hiroki Shichi
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Hospital, Kobe, 650-0017, Japan
| | - Keitaro Kanie
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Hospital, Kobe, 650-0017, Japan
| | - Masaaki Yamamoto
- Division of Diabetes and Endocrinology, Kobe University Graduate School of Medicine, Kobe, 650-0017, Japan
| | - Naoki Yamamoto
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Hospital, Kobe, 650-0017, Japan
| | - Yasutaka Tsujimoto
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Hospital, Kobe, 650-0017, Japan
| | - Hironori Bando
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Hospital, Kobe, 650-0017, Japan
| | - Genzo Iguchi
- Medical Center for Student Health, Kobe University, Kobe, 657-8501, Japan
| | - Shigehisa Kitano
- Division of Cancer Immunotherapy Development, Department of Advanced Medical Development, The Cancer Institute Hospital of Japanese Foundation for Cancer Research (JFCR), Koto-ku, Tokyo, 135-8550, Japan
| | - Naoko Inoshita
- Department of Diagnostic Pathology, Moriyama Memorial Hospital, Tokyo, 134-0088, Japan
| | - Shozo Yamada
- Pituitary Center, Moriyama Memorial Hospital, Tokyo, 134-0088, Japan
- Hypothalamic and Pituitary Center, Toranomon Hospital, Tokyo, 105-8470, Japan
| | - Wataru Ogawa
- Division of Diabetes and Endocrinology, Kobe University Graduate School of Medicine, Kobe, 650-0017, Japan
| | - Tomoo Itoh
- Department of Diagnostic Pathology, Kobe University Hospital, Kobe, 650-0017, Japan
| | - Hidenori Fukuoka
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Hospital, Kobe, 650-0017, Japan
| |
Collapse
|
9
|
Iglesias P. Aggressive and Metastatic Pituitary Neuroendocrine Tumors: Therapeutic Management and Off-Label Drug Use. J Clin Med 2023; 13:116. [PMID: 38202123 PMCID: PMC10779494 DOI: 10.3390/jcm13010116] [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: 11/11/2023] [Revised: 12/17/2023] [Accepted: 12/22/2023] [Indexed: 01/12/2024] Open
Abstract
Pituitary neuroendocrine tumors (PitNETs) are the most common pituitary tumors and the second most common brain tumors. Although the vast majority (>90%) are benign, a small percentage (<2%) are aggressive. These aggressive PitNETs (AgPitNETs) are defined by the presence of radiological invasion, a high rate of cell proliferation, resistance to conventional treatments, and/or a high propensity for recurrence. Lastly, there are the rare pituitary carcinomas, also known as metastatic PitNETs (MetPitNETs), which account for only 0.2% of cases and are defined by the presence of craniospinal or distant metastases. At present, there are no definitive factors that allow us to predict with certainty the aggressive behavior of PitNETs, making the therapeutic management of AgPitNETs a real challenge. Surgery is considered the first-line treatment for AgPitNETs and MetPitNETs. Radiation therapy can be effective in controlling tumor growth and regulating hormone hypersecretion. Currently, there are no approved non-endocrine medical therapies for the management of AgPitNETs/MetPitNETs, mainly due to the lack of randomized controlled clinical trials. As a result, many of the medical therapies used are off-label drugs, and several are under investigation. Temozolomide (TMZ) is now recognized as the primary medical treatment following the failure of standard therapy (medical treatment, surgery, and radiotherapy) in AgPitNETs/MetPitNETs due to its ability to improve overall and progression-free survival rates in responding patients over 5 years. Other therapeutic options include pituitary-targeted therapies (dopamine agonists and somatostatin analogs), hormonal antisecretory drugs, non-hormonal targeted therapies, radionuclide treatments, and immunotherapy. However, the number of patients who have undergone these treatments is limited, and the results obtained to date have been inconsistent. As a result, it is imperative to expand the cohort of patients undergoing treatment to better determine the therapeutic efficacy and safety of these drugs for individuals with AgPitNETs/MetPitNETs.
Collapse
Affiliation(s)
- Pedro Iglesias
- Department of Endocrinology, Hospital Universitario Puerta de Hierro Majadahonda, Instituto de Investigación Sanitaria Puerta de Hierro Segovia de Arana (IDIPHISA), 28222 Madrid, Spain
| |
Collapse
|
10
|
Gentilin E, Borges De Souza P, Ambrosio MR, Bondanelli M, Gagliardi I, Zatelli MC. Protein kinase C delta mediates Pasireotide effects in an ACTH-secreting pituitary tumor cell line. J Endocrinol Invest 2023; 46:2609-2616. [PMID: 37233978 PMCID: PMC10632222 DOI: 10.1007/s40618-023-02117-0] [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: 10/12/2022] [Accepted: 05/16/2023] [Indexed: 05/27/2023]
Abstract
PURPOSE Clinical control of corticotroph tumors is difficult to achieve since they usually persist or relapse after surgery. Pasireotide is approved to treat patients with Cushing's disease for whom surgical therapy is not an option. However, Pasireotide seems to be effective only in a sub-set of patients, highlighting the importance to find a response marker to this approach. Recent studies demonstrated that the delta isoform of protein kinase C (PRKCD) controls viability and cell cycle progression of an in vitro model of ACTH-secreting pituitary tumor, the AtT-20/D16v-F2 cells. This study aims at exploring the possible PRKCD role in mediating Pasireotide effects. METHODS It was assessed cell viability, POMC expression and ACTH secretion in AtT20/D16v-F2 cells over- or under-expressing PRKCD. RESULTS We found that Pasireotide significantly reduces AtT20/D16v-F2 cell viability, POMC expression and ACTH secretion. In addition, Pasireotide reduces miR-26a expression. PRKCD silencing decreases AtT20/D16v-F2 cell sensitivity to Pasireotide treatment; on the contrary, PRKCD overexpression increases the inhibitory effects of Pasireotide on cell viability and ACTH secretion. CONCLUSION Our results provide new insights into potential PRKCD contribution in Pasireotide mechanism of action and suggest that PRKCD might be a possible marker of therapeutic response in ACTH-secreting pituitary tumors.
Collapse
Affiliation(s)
- E Gentilin
- Bioacoustics Research Laboratory, Department of Neurosciences, University of Padua, Padua, Italy.
- Section of Endocrinology, Geriatrics & Internal Medicine, Dept. of Medical Sciences, University of Ferrara, Ferrara, Italy.
| | - P Borges De Souza
- Section of Endocrinology, Geriatrics & Internal Medicine, Dept. of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - M R Ambrosio
- Section of Endocrinology, Geriatrics & Internal Medicine, Dept. of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - M Bondanelli
- Section of Endocrinology, Geriatrics & Internal Medicine, Dept. of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - I Gagliardi
- Section of Endocrinology, Geriatrics & Internal Medicine, Dept. of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - M C Zatelli
- Section of Endocrinology, Geriatrics & Internal Medicine, Dept. of Medical Sciences, University of Ferrara, Ferrara, Italy.
| |
Collapse
|
11
|
Li X, Deng K, Zhang Y, Feng M, Xing B, Lian W, Yao Y. Pediatric pituitary neuroendocrine tumors-a 13-year experience in a tertiary center. Front Oncol 2023; 13:1270958. [PMID: 38023185 PMCID: PMC10661939 DOI: 10.3389/fonc.2023.1270958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction Pediatric pituitary neuroendocrine tumor is a rare condition, and despite previous research focusing on this specific group, the main factors influencing the surgical cure rate have not been identified. Methods We conducted a single-center retrospective study on pediatric pituitary neuroendocrine tumor patients who visited Peking Union Medical College Hospital between 2010 and 2023. We collected data on their clinical characteristics, imaging features, surgical outcomes, and follow-up information. Additionally, we used multiple-factor logistic regression to investigate the factors affecting the surgical cure rate of pediatric pituitary neuroendocrine tumor. Results 232 patients were diagnosed with pediatric pituitary neuroendocrine tumors, with a higher incidence in females. The most common type was ACTH-secreting adenoma (90/232), followed by prolactin-secreting adenoma (63/232), and growth hormone-secreting adenoma (41/232). The majority of pediatric adenomas were macroadenomas (139/232), and some tumors were associated with cystic changes or hemorrhage (58/232), while a few exhibited invasion of the cavernous sinus (33/232). The results of the multivariate analysis indicated that the different hormone secretion types, macroadenoma or the presence of cystic changes or hemorrhage were not significant risk factors for the cure rate after the first surgery. However, the invasion of the cavernous sinus was found to be an important factor influencing the postoperative cure rate. Most pediatric pituitary neuroendocrine tumors with cavernous sinus invasion were macroadenomas, and some displayed characteristics of refractory pituitary neuroendocrine tumors, with some patients experiencing irreversible complications after surgery. Conclusion Pediatric pituitary neuroendocrine tumors are complex, and the postoperative cure rate is particularly poor for tumors with cavernous sinus invasion. Although macroadenoma itself does not significantly impact the postoperative cure rate, it is still recommended to diagnose and treat early to avoid unnecessary surgery or surgical complications.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yong Yao
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
12
|
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
|
13
|
Serioli S, Borghesi I, Regge Gianas N, Fontanella MM, Calbucci F, Draghi R. Petroclival Aggressive Pituitary Adenoma in Nelson's Syndrome: 2-Dimensional Operative Video. Oper Neurosurg (Hagerstown) 2023; 25:e164. [PMID: 37222515 DOI: 10.1227/ons.0000000000000778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 03/29/2023] [Indexed: 05/25/2023] Open
Affiliation(s)
- Simona Serioli
- Division of Neurosurgery, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, Spedali Civili of Brescia, University of Brescia, Brescia, Italy
- Unit of Neurosurgery, Maria Cecilia Hospital, GVM Care & Research, Cotignola, Ravenna, Italy
| | - Ignazio Borghesi
- Unit of Neurosurgery, Maria Cecilia Hospital, GVM Care & Research, Cotignola, Ravenna, Italy
| | - Nicolò Regge Gianas
- Unit of Neurosurgery, Maria Cecilia Hospital, GVM Care & Research, Cotignola, Ravenna, Italy
| | - Marco Maria Fontanella
- Division of Neurosurgery, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, Spedali Civili of Brescia, University of Brescia, Brescia, Italy
| | - Fabio Calbucci
- Unit of Neurosurgery, Maria Cecilia Hospital, GVM Care & Research, Cotignola, Ravenna, Italy
| | - Riccardo Draghi
- Unit of Neurosurgery, Maria Cecilia Hospital, GVM Care & Research, Cotignola, Ravenna, Italy
| |
Collapse
|
14
|
Yami Channaiah C, Memon SS, Sarathi V, Lila AR, Sankhe S, Arya S, Karlekar M, Patil VA, Shah N, Bandgar T. Pediatric Macrocorticotropinoma: Do They Differ from Microcorticotropinoma? Neuroendocrinology 2023; 114:42-50. [PMID: 37634509 DOI: 10.1159/000533770] [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: 10/04/2022] [Accepted: 11/29/2022] [Indexed: 08/29/2023]
Abstract
INTRODUCTION Cushing's disease (CD) due to macrocorticotropinoma (MC) in children and adolescents is a rare entity with limited information regarding its characteristics. The objective of the study is to describe the clinical, biochemical, imaging, management, outcome, and genetic characteristics of children and adolescents with CD due to MC and compare them with those of microcorticotropinoma (mc). METHODS This retrospective study was conducted at a single tertiary care center. Thirty-two patients with CD and MC (maximum tumor dimension ≥10 mm on imaging) and 65 patients with mc (<10 mm on imaging) aged ≤20 years at presentation were enrolled. RESULTS Nineteen girls and 13 boys with MC presented at a median (IQR) age of 14.5 (12.0-17.9) years. Patients with MC had higher body mass index-standard deviation score (BMI-SDS) (3.70 ± 2.60 vs. 2.59 ± 2.01, p = 0.04), more frequent neuro-ophthalmic symptoms (25% vs. 9% p = 0.04) and short stature (59% vs. 34%, p = 0.049) but less frequent livid striae (53% vs. 77%, p = 0.01), hypokalemia (12% vs. 36%, p = 0.04), and lower cortisol (nmol/L) to corticotropin (pmol/L) ratio (41.20 vs. 55.74, p = 0.04) than those with mc. The remission (59% vs. 64%, p = 1.0) and relapse (53% vs. 37%, p = 0.26) rates after first-line surgery and remission rate after radiotherapy (RT) were comparable between the two cohorts, whereas time to remission after RT (27 vs. 13 months, p = 0.05) was longer in the MC group. A patient with MC had a pathogenic germline variant in CDH23. CONCLUSION In this large monocentric series of pediatric CD, frequent mass effect symptoms and short stature, higher BMI-SDS, less frequent livid striae, and hypokalemia with lower effective cortisol secretion characterize the MC cohort. The outcomes of surgery and RT were similar between the groups except for a longer time to remission after RT in the MC cohort. Germline variants are rare (4%) in pediatric MC.
Collapse
Affiliation(s)
| | - Saba Samad Memon
- Department of Endocrinology, Seth GS Medical College and KEM Hospital, Mumbai, India
| | - Vijaya Sarathi
- Department of Endocrinology, Vydehi Institute of Medical Sciences and Research Centre, Bengaluru, India
| | - Anurag Ranjan Lila
- Department of Endocrinology, Seth GS Medical College and KEM Hospital, Mumbai, India
| | - Shilpa Sankhe
- Department of Radiology, Seth GS Medical College and KEM Hospital, Mumbai, India
| | - Sneha Arya
- Department of Endocrinology, Seth GS Medical College and KEM Hospital, Mumbai, India
| | - Manjiri Karlekar
- Department of Endocrinology, Seth GS Medical College and KEM Hospital, Mumbai, India
| | | | - Nalini Shah
- Department of Endocrinology, Seth GS Medical College and KEM Hospital, Mumbai, India
| | - Tushar Bandgar
- Department of Endocrinology, Seth GS Medical College and KEM Hospital, Mumbai, India
| |
Collapse
|
15
|
Osamura RY, Inomoto C, Tahara S, Oyama KI, Matsuno A, Teramoto A. Pathology of Crooke Cells in the Human Pituitaries: A Timely Review. Appl Immunohistochem Mol Morphol 2023; 31:485-489. [PMID: 36251979 DOI: 10.1097/pai.0000000000001070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 09/09/2022] [Indexed: 11/02/2022]
Abstract
Crooke cell change was first found in the regressed and suppressed corticotroph (adrenocorticotropic hormone-producing) cells, and now is known to occur in pituitary tumors. The tumor cells of this type can be recognized by morphology with immunohistochemistry, and are well known to predict aggressive behavior such as invasion and rare metastases. This is one of the representative neuroendocrine tumors in the pituitary which is now considered to have malignant potential as proposed in the pancreas and gastrointestinal tracts. It is important to emphasize the pituitary tumor pathology such as Crooke cell change for prognostication and appropriate therapies. This review article describes the evolution from the Crooke cells to Crooke cell tumors which is timely along with the Fifth WHO classification 2022 published online.
Collapse
Affiliation(s)
- Robert Y Osamura
- Nippon Koukan Hospital, Kawasaki
- Keio University School of Medicine
| | | | | | | | | | | |
Collapse
|
16
|
Derwich A, Sykutera M, Bromińska B, Rubiś B, Ruchała M, Sawicka-Gutaj N. The Role of Activation of PI3K/AKT/mTOR and RAF/MEK/ERK Pathways in Aggressive Pituitary Adenomas-New Potential Therapeutic Approach-A Systematic Review. Int J Mol Sci 2023; 24:10952. [PMID: 37446128 PMCID: PMC10341524 DOI: 10.3390/ijms241310952] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/18/2023] [Accepted: 06/23/2023] [Indexed: 07/15/2023] Open
Abstract
Pituitary tumors (PT) are mostly benign, although occasionally they demonstrate aggressive behavior, invasion of surrounding tissues, rapid growth, resistance to conventional treatments, and multiple recurrences. The pathogenesis of PT is still not fully understood, and the factors responsible for its invasiveness, aggressiveness, and potential for metastasis are unknown. RAF/MEK/ERK and mTOR signaling are significant pathways in the regulation of cell growth, proliferation, and survival, its importance in tumorigenesis has been highlighted. The aim of our review is to determine the role of the activation of PI3K/AKT/mTOR and RAF/MEK/ERK pathways in the pathogenesis of pituitary tumors. Additionally, we evaluate their potential in a new therapeutic approach to provide alternative therapies and improved outcomes for patients with aggressive pituitary tumors that do not respond to standard treatment. We perform a systematic literature search using the PubMed, Embase, and Scopus databases (search date was 2012-2023). Out of the 529 screened studies, 13 met the inclusion criteria, 7 related to the PI3K/AKT/mTOR pathway, and 7 to the RAF/MEK/ERK pathway (one study was used in both analyses). Understanding the specific factors involved in PT tumorigenesis provides opportunities for targeted therapies. We also review the possible new targeted therapies and the use of mTOR inhibitors and TKI in PT management. Although the RAF/MEK/ERK and PI3K/AKT/mTOR pathways play a pivotal role in the complex signaling network along with many interactions, further research is urgently needed to clarify the exact functions and the underlying mechanisms of these signaling pathways in the pathogenesis of pituitary adenomas and their role in its invasiveness and aggressive clinical outcome.
Collapse
Affiliation(s)
- Aleksandra Derwich
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, 60-355 Poznan, Poland; (A.D.)
- Doctoral School, Poznan University of Medical Sciences, 60-812 Poznan, Poland
| | - Monika Sykutera
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, 60-355 Poznan, Poland; (A.D.)
| | - Barbara Bromińska
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, 60-355 Poznan, Poland; (A.D.)
| | - Błażej Rubiś
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Marek Ruchała
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, 60-355 Poznan, Poland; (A.D.)
| | - Nadia Sawicka-Gutaj
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, 60-355 Poznan, Poland; (A.D.)
| |
Collapse
|
17
|
Sumal AKS, Zhang D, Heaney AP. Refractory corticotroph adenomas. Pituitary 2023; 26:269-272. [PMID: 36917358 PMCID: PMC10333410 DOI: 10.1007/s11102-023-01308-5] [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] [Accepted: 03/04/2023] [Indexed: 03/16/2023]
Abstract
The majority of corticotroph adenomas are benign but some are locally invasive, demonstrate high rates of recurrence, and exhibit a relatively poor response to often repeated surgical, medical, and radiation treatment. Herein, we summarize the currently known somatic and genetic mutations and other molecular factors that influence the pathogenesis of these tumors and discuss currently available therapies. Although recent molecular studies have advanced our understanding of the pathogenesis and behavior of these refractory corticotroph adenomas, these insights do not reliably guide treatment choices at present. Development of additional diagnostic tools and novel tumor-directed therapies that offer efficacious treatment choices for patients with refractory corticotroph adenomas are needed.
Collapse
Affiliation(s)
- Amit K S Sumal
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Dongyun Zhang
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Anthony P Heaney
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
- Department of Neurosurgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
18
|
Advances in Molecular Pathophysiology and Targeted Therapy for Cushing's Disease. Cancers (Basel) 2023; 15:cancers15020496. [PMID: 36672445 PMCID: PMC9857185 DOI: 10.3390/cancers15020496] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/02/2023] [Accepted: 01/10/2023] [Indexed: 01/15/2023] Open
Abstract
Cushing's disease is caused by autonomous secretion of adrenocorticotropic hormone (ACTH) from corticotroph pituitary neuroendocrine tumors. As a result, excess cortisol production leads to the overt manifestation of the clinical features of Cushing's syndrome. Severe complications have been reported in patients with Cushing's disease, including hypertension, menstrual disorders, hyperglycemia, osteoporosis, atherosclerosis, infections, and mental disorders. Cushing's disease presents with a variety of clinical features, ranging from overt to subtle. In this review, we explain recent advances in molecular insights and targeted therapy for Cushing's disease. The pathophysiological characteristics of hormone production and pituitary tumor cells are also explained. Therapies to treat the tumor growth in the pituitary gland and the autonomous hypersecretion of ACTH are discussed. Drugs that target corticotroph pituitary neuroendocrine tumors have been effective, including cabergoline, a dopamine receptor type 2 agonist, and pasireotide, a multi-receptor-targeted somatostatin analog. Some of the drugs that target adrenal hormones have shown potential therapeutic benefits. Advances in potential novel therapies for Cushing's disease are also introduced.
Collapse
|
19
|
Locantore P, Paragliola RM, Cera G, Novizio R, Maggio E, Ramunno V, Corsello A, Corsello SM. Genetic Basis of ACTH-Secreting Adenomas. Int J Mol Sci 2022; 23:ijms23126824. [PMID: 35743266 PMCID: PMC9224284 DOI: 10.3390/ijms23126824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 06/08/2022] [Accepted: 06/17/2022] [Indexed: 12/10/2022] Open
Abstract
Cushing's disease represents 60-70% of all cases of Cushing's syndrome, presenting with a constellation of clinical features associated with sustained hypercortisolism. Molecular alterations in corticotrope cells lead to the formation of ACTH-secreting adenomas, with subsequent excessive production of endogenous glucocorticoids. In the last few years, many authors have contributed to analyzing the etiopathogenesis and pathophysiology of corticotrope adenomas, which still need to be fully clarified. New molecular modifications such as somatic mutations of USP8 and other genes have been identified, and several case series and case reports have been published, highlighting new molecular alterations that need to be explored. To investigate the current knowledge of the genetics of ACTH-secreting adenomas, we performed a bibliographic search of the recent scientific literature to identify all pertinent articles. This review presents the most recent updates on somatic and germline mutations underlying Cushing's disease. The prognostic implications of these mutations, in terms of clinical outcomes and therapeutic scenarios, are still debated. Further research is needed to define the clinical features associated with the different genotypes and potential pharmacological targets.
Collapse
Affiliation(s)
- Pietro Locantore
- Unit of Endocrinology, Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore—Fondazione Policlinico “Gemelli” IRCCS, Largo Gemelli 8, I-00168 Rome, Italy; (P.L.); (G.C.); (R.N.); (E.M.); (V.R.); (A.C.); (S.M.C.)
| | - Rosa Maria Paragliola
- Unit of Endocrinology, Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore—Fondazione Policlinico “Gemelli” IRCCS, Largo Gemelli 8, I-00168 Rome, Italy; (P.L.); (G.C.); (R.N.); (E.M.); (V.R.); (A.C.); (S.M.C.)
- Correspondence:
| | - Gianluca Cera
- Unit of Endocrinology, Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore—Fondazione Policlinico “Gemelli” IRCCS, Largo Gemelli 8, I-00168 Rome, Italy; (P.L.); (G.C.); (R.N.); (E.M.); (V.R.); (A.C.); (S.M.C.)
| | - Roberto Novizio
- Unit of Endocrinology, Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore—Fondazione Policlinico “Gemelli” IRCCS, Largo Gemelli 8, I-00168 Rome, Italy; (P.L.); (G.C.); (R.N.); (E.M.); (V.R.); (A.C.); (S.M.C.)
| | - Ettore Maggio
- Unit of Endocrinology, Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore—Fondazione Policlinico “Gemelli” IRCCS, Largo Gemelli 8, I-00168 Rome, Italy; (P.L.); (G.C.); (R.N.); (E.M.); (V.R.); (A.C.); (S.M.C.)
| | - Vittoria Ramunno
- Unit of Endocrinology, Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore—Fondazione Policlinico “Gemelli” IRCCS, Largo Gemelli 8, I-00168 Rome, Italy; (P.L.); (G.C.); (R.N.); (E.M.); (V.R.); (A.C.); (S.M.C.)
| | - Andrea Corsello
- Unit of Endocrinology, Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore—Fondazione Policlinico “Gemelli” IRCCS, Largo Gemelli 8, I-00168 Rome, Italy; (P.L.); (G.C.); (R.N.); (E.M.); (V.R.); (A.C.); (S.M.C.)
| | - Salvatore Maria Corsello
- Unit of Endocrinology, Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore—Fondazione Policlinico “Gemelli” IRCCS, Largo Gemelli 8, I-00168 Rome, Italy; (P.L.); (G.C.); (R.N.); (E.M.); (V.R.); (A.C.); (S.M.C.)
- Unicamillus, Saint Camillus International University of Medical Sciences, via di S. Alessandro 10, I-00131 Rome, Italy
| |
Collapse
|
20
|
Kuritsyna NV, Tsoy UA, Cherebillo VY, Paltsev AA, Ryzhkov AV, Ryazanov PA, Ryzhkov VK, Grineva EN. A Comprehensive Approach to Predicting the Outcomes of Transsphenoidal Endoscopic Adenomectomy in Patients with Cushing’s Disease. J Pers Med 2022; 12:jpm12050798. [PMID: 35629220 PMCID: PMC9144911 DOI: 10.3390/jpm12050798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/03/2022] [Accepted: 05/11/2022] [Indexed: 12/10/2022] Open
Abstract
Persistent and recurrent hypercortisolism after transsphenoidal endoscopic surgery (TSS) is considered to be an urgent issue prompting the search for Cushing’s disease (CD) remission predictors. The goal was to find a combination of predictors that can forecast the remission of CD after TSS. A total of 101 patients with CD who had undergone TSS were included. One year after surgery, CD remission status was evaluated. Preoperative pituitary magnetic resonance imaging (MRI) data, preoperative results of a high-dose dexamethasone suppression test (HDDST) and morning serum cortisol level collected 24 h after TSS (24 h MSeC) were compared in patients with and without remission of hypercortisolism. Remission one year after TSS was confirmed in 63 patients. CD remission predictors one year after TSS were: adenoma size ≥ 3 mm in the absence of invasive growth and the suppression of serum cortisol ≥ 74% in the HDDST, 24 h MSeC ≤ 388 nmol/L. A total of 38 patients had three favorable values of detected predictors; all of them had CD remission one year after TSS. With long-term follow-up, 36 of them remained in remission. Patients who had no one favorable predictor had no remission of hypercortisolism one year after TSS. Our data confirmed the prospects of using a combination of selected predictors to forecast CD remission after TSS.
Collapse
|
21
|
Pathophysiology of Mild Hypercortisolism: From the Bench to the Bedside. Int J Mol Sci 2022; 23:ijms23020673. [PMID: 35054858 PMCID: PMC8775422 DOI: 10.3390/ijms23020673] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/31/2021] [Accepted: 01/05/2022] [Indexed: 02/07/2023] Open
Abstract
Mild hypercortisolism is defined as biochemical evidence of abnormal cortisol secretion without the classical detectable manifestations of overt Cushing’s syndrome and, above all, lacking catabolic characteristics such as central muscle weakness, adipose tissue redistribution, skin fragility and unusual infections. Mild hypercortisolism is frequently discovered in patients with adrenal incidentalomas, with a prevalence ranging between 5 and 50%. This high variability is mainly due to the different criteria used for defining this condition. This subtle cortisol excess has also been described in patients with incidentally discovered pituitary tumors with an estimated prevalence of 5%. To date, the mechanisms responsible for the pathogenesis of mild hypercortisolism of pituitary origin are still not well clarified. At variance, recent advances have been made in understanding the genetic background of bilateral and unilateral adrenal adenomas causing mild hypercortisolism. Some recent data suggest that the clinical effects of glucocorticoid (GC) exposure on peripheral tissues are determined not only by the amount of the adrenal GC production but also by the peripheral GC metabolism and by the GC sensitivity. Indeed, in subjects with normal cortisol secretion, the combined estimate of cortisol secretion, cortisone-to-cortisol peripheral activation by the 11 beta-hydroxysteroid dehydrogenase enzyme and GC receptor sensitizing variants have been suggested to be associated with the presence of hypertension, diabetes and bone fragility, which are three well-known consequences of hypercortisolism. This review focuses on the pathophysiologic mechanism underlying both the different sources of mild hypercortisolism and their clinical consequences (bone fragility, arterial hypertension, subclinical atherosclerosis, cardiovascular remodeling, dyslipidemia, glucose metabolism impairment, visceral adiposity, infections, muscle damage, mood disorders and coagulation).
Collapse
|
22
|
Jiang S, Chen X, Wu Y, Wang R, Bao X. An Update on Silent Corticotroph Adenomas: Diagnosis, Mechanisms, Clinical Features, and Management. Cancers (Basel) 2021; 13:cancers13236134. [PMID: 34885244 PMCID: PMC8656508 DOI: 10.3390/cancers13236134] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/26/2021] [Accepted: 12/02/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary The 2017 World Health Organization classification of endocrine tumors defines pituitary adenomas based on their cell lineages. T-PIT can serve as a complimentary tool for further identification of silent corticotroph adenomas (SCAs). Unlike functioning corticotroph adenomas in patients with Cushing’s disease, SCAs present no clinical and biochemical features of Cushing’s syndrome. SCAs have been shown to exhibit a more aggressive course characterized by a higher probability of recurrence and resistance to conventional treatment due to their intrinsic histological features. The aim of our review is to offer an update on the diagnosis, mechanisms, clinical features and management of SCAs. Studies of the molecular mechanisms of SCA pathogenesis will provide new directions for the diagnosis and management of SCAs. Abstract With the introduction of 2017 World Health Organization (WHO) classification of endocrine tumors, T-PIT can serve as a complementary tool for identification of silent corticotroph adenomas (SCAs) in some cases if the tumor is not classifiable by pituitary hormone expression in pathological tissue samples. An increase of the proportion of SCAs among the non-functioning pituitary adenomas (NFPAs) has been witnessed under the new rule with the detection of T-PIT-positive ACTH-negative SCAs. Studies of molecular mechanisms related to SCA pathogenesis will provide new directions for the diagnosis and management of SCAs. A precise pathological diagnosis can help clinicians better identify SCAs. Understanding clinical features in the context of the pathophysiology of SCAs is critical for optimal management. It could provide information on appropriate follow-up time and aid in early recognition and treatment of potentially aggressive forms. Management approaches include surgical, radiation, and/or medical therapies.
Collapse
|
23
|
Massman LJ, Pereckas M, Zwagerman NT, Olivier-Van Stichelen S. O-GlcNAcylation Is Essential for Rapid Pomc Expression and Cell Proliferation in Corticotropic Tumor Cells. Endocrinology 2021; 162:6356179. [PMID: 34418053 PMCID: PMC8482966 DOI: 10.1210/endocr/bqab178] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Indexed: 12/13/2022]
Abstract
Pituitary adenomas have a staggering 16.7% lifetime prevalence and can be devastating in many patients because of profound endocrine and neurologic dysfunction. To date, no clear genomic or epigenomic markers correlate with their onset or severity. Herein, we investigate the impact of the O-GlcNAc posttranslational modification in their etiology. Found in more than 7000 human proteins to date, O-GlcNAcylation dynamically regulates proteins in critical signaling pathways, and its deregulation is involved in cancer progression and endocrine diseases such as diabetes. In this study, we demonstrated that O-GlcNAc enzymes were upregulated, particularly in aggressive adrenocorticotropin (ACTH)-secreting tumors, suggesting a role for O-GlcNAcylation in pituitary adenoma etiology. In addition to the demonstration that O-GlcNAcylation was essential for their proliferation, we showed that the endocrine function of pituitary adenoma is also dependent on O-GlcNAcylation. In corticotropic tumors, hypersecretion of the proopiomelanocortin (POMC)-derived hormone ACTH leads to Cushing disease, materialized by severe endocrine disruption and increased mortality. We demonstrated that Pomc messenger RNA is stabilized in an O-GlcNAc-dependent manner in response to corticotrophin-releasing hormone (CRH). By affecting Pomc mRNA splicing and stability, O-GlcNAcylation contributes to this new mechanism of fast hormonal response in corticotropes. Thus, this study stresses the essential role of O-GlcNAcylation in ACTH-secreting adenomas' pathophysiology, including cellular proliferation and hypersecretion.
Collapse
Affiliation(s)
- Logan J Massman
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, 53226, USA
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin, 53226, USA
| | - Michael Pereckas
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, 53226, USA
| | - Nathan T Zwagerman
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin, 53226, USA
| | - Stephanie Olivier-Van Stichelen
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, 53226, USA
- Correspondence: Stephanie Olivier-Van Stichelen, PhD, Department of Biochemistry, Medical College of Wisconsin, BSB355, 8701 Watertown Plank Rd, Milwaukee, WI 53226, USA.
| |
Collapse
|