1
|
Liechty B, Kim S, Dobri G, Schwartz TH, Ivanidze J, Pisapia D. SSTR2 expression in neoplastic and normal anterior pituitary is impacted by age, sex, and hormonal status. J Neuropathol Exp Neurol 2025:nlaf034. [PMID: 40261909 DOI: 10.1093/jnen/nlaf034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025] Open
Abstract
Pituitary neuroendocrine tumors (PitNETs) are among the most common tumors encountered in neurooncology. While the majority of PitNETs demonstrate indolent behavior, a subset of tumors demonstrates aggressive behavior, including invasion into surrounding structures. As traditional imaging has limited capacity to distinguish tumor from post-operative changes, better methods of tumor delineation are needed to guide management. Somatotroph adenomas are known to express high levels of SSTR2, and SSTR2-targeting PET imaging has shown clinical utility in the management of neuroendocrine tumors and meningiomas. In this retrospective study of archival PitNETs (n = 271) and autopsy controls (AC) (n = 20), we show that although significant differences in SSTR2 immunostaining are appreciable between adenoma subtypes and ACs, high-staining cases are encountered in all subtypes. In ACs, females demonstrated significantly stronger SSTR2 staining than males. Weak age-related trends towards increasing labelling in females and decreasing labelling in males were noted but these did not reach statistical significance. Decreasing age-related trends were seen in gonadotrophs in both sexes; this was statistically significant in females. Our findings suggest that SSTR2-targeting imaging modalities may assist clinical management of a subset of PitNETs and that these results may need to be interpreted with consideration of patient age and sex.
Collapse
Affiliation(s)
- Benjamin Liechty
- Division of Neuropathology, Department of Pathology and Laboratory Medicine, Weill-Cornell Medical College, New York, NY, United States
| | - Sean Kim
- Division of Neuroradiology, Department of Radiology, Weill-Cornell Medical College, New York, NY, United States
| | - Georgiana Dobri
- Department of Neurological Surgery, Weill-Cornell Medical College, New York, NY, United States
- Department of Medicine, Weill-Cornell Medical College, New York, NY, United States
| | - Theodore H Schwartz
- Department of Neurological Surgery, Weill-Cornell Medical College, New York, NY, United States
| | - Jana Ivanidze
- Division of Neuroradiology, Department of Radiology, Weill-Cornell Medical College, New York, NY, United States
| | - David Pisapia
- Division of Neuropathology, Department of Pathology and Laboratory Medicine, Weill-Cornell Medical College, New York, NY, United States
| |
Collapse
|
2
|
Akdemir AS, Metin Armagan D, Polat Korkmaz O, Ozkaya HM, Kadioglu P, Gazioglu N, Tanriover N, Dirican A, Ozturk M. Association between β-arrestin-2 and filamin-A gene variations with medical treatment response in acromegaly patients. Minerva Endocrinol (Torino) 2025; 50:32-41. [PMID: 34669321 DOI: 10.23736/s2724-6507.21.03611-3] [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: 11/08/2022]
Abstract
BACKGROUND Acromegaly is a disease that occurs as a result of excessive growth hormone caused by pituitary adenomas. Some acromegaly patients show resistance to somatostatin analog (SSA) treatment. Filamin-A (FLNA) and β-arrestins are thought to play a role in the response to SSAs. We aimed to investigate the relationship between FLNA-rs782079491 and β-arrestin-2-rs34230287 single-nucleotide polymorphisms and disease risk, as well as treatment response in patients with acromegaly in the Turkish population. METHODS The genotypes of 110 acromegaly patients and 99 controls were determined by real-time PCR. The genotype distributions were compared with clinical data on the disease. RESULTS There was no association between the β-arrestin-2 gene polymorphism and the response to SSA treatment in acromegaly patients. For responder patients to SSAs, the β-arrestin-2-rs34230287 CT+TT genotype was associated with higher microadenoma as compared with the CC genotype (P=0.017). The FLNA polymorphism was not observed in the study group. CONCLUSIONS We showed that there was no association between the polymorphic genotypes of FLNA and β-arrestin-2 genes with acromegaly disease and SSAs response in the Turkish population. However, there was a relationship between β-arrestin-2 and some of the clinical characteristics. Furthermore, the CC genotype and the C allele are risk factors associated with tumor growth rate in acromegaly patients.
Collapse
Affiliation(s)
- Ayse S Akdemir
- Department of Medical Biology, Cerrahpasa Faculty of Medicine, University of Istanbul-Cerrahpasa, Istanbul, Türkiye
| | - Derya Metin Armagan
- Department of Medical Biology, Cerrahpasa Faculty of Medicine, University of Istanbul-Cerrahpasa, Istanbul, Türkiye
- Department of Medicine, Cedars - Sinai Medical Center, Los Angeles, CA, USA
| | - Ozge Polat Korkmaz
- Department of Endocrinology and Metabolism, Cerrahpasa Faculty of Medicine, University of Istanbul-Cerrahpasa, Istanbul, Türkiye
| | - Hande M Ozkaya
- Department of Endocrinology and Metabolism, Cerrahpasa Faculty of Medicine, University of Istanbul-Cerrahpasa, Istanbul, Türkiye
| | - Pinar Kadioglu
- Department of Endocrinology and Metabolism, Cerrahpasa Faculty of Medicine, University of Istanbul-Cerrahpasa, Istanbul, Türkiye
- Pituitary Research Center, University of Istanbul-Cerrahpasa, Istanbul, Türkiye
| | - Nurperi Gazioglu
- Department of Neurosurgery, Medicine Faculty, Istinye University, Istanbul, Türkiye
| | - Necmettin Tanriover
- Pituitary Research Center, University of Istanbul-Cerrahpasa, Istanbul, Türkiye
- Department of Neurosurgery, Cerrahpasa Faculty of Medicine, University of Istanbul-Cerrahpasa, Istanbul, Türkiye
| | - Ahmet Dirican
- Department of Biostatistics, Cerrahpasa Faculty of Medicine, University of Istanbul-Cerrahpasa, Istanbul, Türkiye
| | - Melek Ozturk
- Department of Medical Biology, Cerrahpasa Faculty of Medicine, University of Istanbul-Cerrahpasa, Istanbul, Türkiye -
| |
Collapse
|
3
|
Botelho L, Dezonne RS, Wildemberg LE, Miranda RL, Gadelha MR, Andreiuolo F. Somatostatin receptors in pituitary somatotroph adenomas as predictors of response to somatostatin receptor ligands: A pathologist's perspective. Brain Pathol 2025; 35:e13313. [PMID: 39473262 DOI: 10.1111/bpa.13313] [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: 07/20/2024] [Accepted: 10/04/2024] [Indexed: 12/28/2024] Open
Abstract
There are five subtypes of somatostatin receptors (SST1-5), which are expressed in several types of solid neoplasms, neuroendocrine tumors, and pituitary adenomas. Most commonly, SST2 and SST5, are of interest regarding diagnostic, treatment, and prognostic purposes. In this article the basic biological characteristics of SST are briefly reviewed, and focus given to the immunohistochemical evaluation of SST2 and SST5 in growth hormone (GH)-secreting pituitary tumors, and their quantification as predictors of response to treatment with somatostatin receptor ligands (SRL), the mainstay of the pharmacological therapy available for these tumors. Although many different scoring systems for SST2 immunohistochemistry showing correlation with SRL response have been reported, among which the immunoreactivity score (IRS) has been the most consistently used, a universally validated immunohistochemical technique and scoring scheme is lacking. Efforts should be made on collaborative multicenter studies aiming at validating homogeneous immunostaining protocols and a scoring system for SST2 and SST5 expression, to help clinicians to define the optimal therapeutic strategy for the patients with somatotroph tumors.
Collapse
Affiliation(s)
- Laura Botelho
- Neuropathology and Molecular Genetics Laboratory, Instituto Estadual do Cérebro Paulo Niemeyer, Rio de Janeiro, Brazil
- Department of Pathology, Rede D'Or, Rio de Janeiro, Brazil
| | - Rômulo Sperduto Dezonne
- Neuropathology and Molecular Genetics Laboratory, Instituto Estadual do Cérebro Paulo Niemeyer, Rio de Janeiro, Brazil
| | - Luiz Eduardo Wildemberg
- 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, Rio de Janeiro, Brazil
| | - Renan Lyra Miranda
- Neuropathology and Molecular Genetics Laboratory, Instituto Estadual do Cérebro Paulo Niemeyer, Rio de Janeiro, Brazil
| | - Mônica R Gadelha
- Neuropathology and Molecular Genetics Laboratory, Instituto Estadual do Cérebro Paulo Niemeyer, Rio de Janeiro, Brazil
- 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, Rio de Janeiro, Brazil
| | - Felipe Andreiuolo
- Neuropathology and Molecular Genetics Laboratory, Instituto Estadual do Cérebro Paulo Niemeyer, Rio de Janeiro, Brazil
- Department of Pathology, Rede D'Or, Rio de Janeiro, Brazil
- D'Or Institute for Research and Education, Rio de Janeiro, Brazil
| |
Collapse
|
4
|
Wang Y, Xu Y, Wang Y, Zhang J, Chen L, He X, Fan W, Wu K, Hu W, Cheng X, Yang G, Xu HE, Zhuang Y, Sun S. Selective ligand recognition and activation of somatostatin receptors SSTR1 and SSTR3. Proc Natl Acad Sci U S A 2024; 121:e2400298121. [PMID: 39361640 PMCID: PMC11474030 DOI: 10.1073/pnas.2400298121] [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: 01/07/2024] [Accepted: 07/31/2024] [Indexed: 10/05/2024] Open
Abstract
Somatostatin receptors (SSTRs) exert critical biological functions such as negatively regulating hormone release and cell proliferation, making them popular targets for developing therapeutics to treat endocrine disorders, especially neuroendocrine tumors. Although several panagonists mimicking the endogenous ligand somatostatin are available, the development of more effective and safer somatostatinergic therapies is limited due to a lack of molecular understanding of the ligand recognition and regulation of divergent SSTR subtypes. Here, we report four cryoelectron microscopy structures of Gi-coupled SSTR1 and SSTR3 activated by distinct agonists, including the FDA-approved panagonist pasireotide as well as their selective small molecule agonists L-797591 and L-796778. Our structures reveal a conserved recognition pattern of pasireotide in SSTRs attributed to the binding with a conserved extended binding pocket, distinct from SST14, octreotide, and lanreotide. Together with mutagenesis analyses, our structures further reveal the dynamic feature of ligand binding pockets in SSTR1 and SSTR3 to accommodate divergent agonists, the key determinants of ligand selectivity lying across the orthosteric pocket of different SSTR subtypes, as well as the molecular mechanism underlying diversity and conservation of receptor activation. Our work provides a framework for rational design of subtype-selective SSTR ligands and may facilitate drug development efforts targeting SSTRs with improved therapeutic efficacy and reduced side effects.
Collapse
Affiliation(s)
- Yujue Wang
- Department of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200011, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai200011, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai200011, China
| | - Youwei Xu
- The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai201203, China
| | - Yue Wang
- The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai201203, China
| | - Jie Zhang
- Department of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200011, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai200011, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai200011, China
| | - Lan Chen
- Department of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200011, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai200011, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai200011, China
| | - Xinheng He
- The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai201203, China
- University of Chinese Academy of Sciences, Beijing100049, China
| | - Wenjia Fan
- The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai201203, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing210046, China
| | - Kai Wu
- The Shanghai Advanced Electron Microscope Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai201203, China
| | - Wen Hu
- The Shanghai Advanced Electron Microscope Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai201203, China
| | - Xi Cheng
- The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai201203, China
| | - Guizhu Yang
- Department of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200011, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai200011, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai200011, China
| | - H. Eric Xu
- The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai201203, China
| | - Youwen Zhuang
- The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai201203, China
- Medicinal Bioinformatics Center, School of Medicine, Shanghai Jiao Tong University, Shanghai200025, China
| | - Shuyang Sun
- Department of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200011, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai200011, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai200011, China
| |
Collapse
|
5
|
Marazuela M, Martínez-Hernandez R, Marques-Pamies M, Biagetti B, Araujo-Castro M, Puig-Domingo M. Predictors of biochemical response to somatostatin receptor ligands in acromegaly. Best Pract Res Clin Endocrinol Metab 2024; 38:101893. [PMID: 38575404 DOI: 10.1016/j.beem.2024.101893] [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: 04/06/2024]
Abstract
Although predictors of response to first-generation somatostatin receptor ligands (fg-SRLs), and to a lesser extent to pasireotide, have been studied in acromegaly for many years, their use is still not recommended in clinical guidelines. Is there insufficient evidence to use them? Numerous biomarkers including various clinical, functional, radiological and molecular markers have been identified. The first ones are applicable pre-surgery, while the molecular predictors are utilized for patients not cured after surgery. In this regard, factors predicting a good response to fg-SRLs are specifically: low basal GH, a low GH nadir in the acute octreotide test, T2 MRI hypointensity, a densely granulated pattern, high immunohistochemistry staining for somatostatin receptor 2 (SSTR2), and E-cadherin. However, there is still a lack of consensus regarding which of these biomarkers is more useful or how to integrate them into clinical practice. With classical statistical methods, it is complex to define reliable and generalizable cut-off values for a single biomarker. The potential solution to the limitations of traditional methods involves combining systems biology with artificial intelligence, which is currently providing answers to such long-standing questions that may eventually be finally included into the clinical guidelines and make personalized medicine a reality. The aim of this review is to describe the current knowledge of the main fg-SRLs and pasireotide response predictors, discuss their current usefulness, and point to future directions in the research of this field.
Collapse
Affiliation(s)
- Mónica Marazuela
- Department of Endocrinology and Nutrition Hospital Universitario La Princesa, Universidad Autónoma de Madrid,Instituto de Investigación Princesa, and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER GCV14/ER/12), Madrid, Spain.
| | | | | | - Betina Biagetti
- Endocrinology & Nutrition Service, Vall d'Hebron University Hospital and Vall d'Hebron Research Institute (VHIR), Department of Medicine, Autonomous University of Barcelona, Reference Networks (ERN), 08035 Barcelona, Spain
| | - Marta Araujo-Castro
- Endocrinology & Nutrition Department. Hospital Universitario Ramón y Cajal, Spain & Instituto de Investigación Biomédica Ramón y Cajal (IRYCIS), Madrid, Spain
| | - Manel Puig-Domingo
- Department of Endocrinology and Nutrition, Department of Medicine, Germans Trias i Pujol Research Institute and Hospital, Universitat Autònoma de Barcelona, Spain and Centro de Investigación Biomédica en Red de Enfermedades Raras CIBERER G747, Badalona, Spain
| |
Collapse
|
6
|
Sampedro-Nuñez M, Herrera-Martínez AD, Ibáñez-Costa A, Rivero-Cortés E, Venegas E, Robledo M, Martínez-Hernández R, García-Martínez A, Gil J, Jordà M, López-Fernández J, Gavilán I, Maraver S, Marqués-Pamies M, Cámara R, Fajardo-Montañana C, Valassi E, Dios E, Aulinas A, Biagetti B, Álvarez Escola C, Araujo-Castro M, Blanco C, Paz DM, Villar-Taibo R, Álvarez CV, Gaztambide S, Webb SM, Castaño L, Bernabéu I, Picó A, Gálvez MÁ, Soto-Moreno A, Puig-Domingo M, Castaño JP, Marazuela M, Luque RM. Integrative clinical, hormonal, and molecular data associate with invasiveness in acromegaly: REMAH study. Eur J Endocrinol 2024; 190:421-433. [PMID: 38701338 DOI: 10.1093/ejendo/lvae045] [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: 09/28/2023] [Revised: 02/18/2024] [Accepted: 03/04/2024] [Indexed: 05/05/2024]
Abstract
INTRODUCTION Growth hormone (GH)-secreting pituitary tumors (GHomas) are the most common acromegaly cause. At diagnosis, most of them are macroadenomas, and up to 56% display cavernous sinus invasion. Biomarker assessment associated with tumor growth and invasion is important to optimize their management. OBJECTIVES The study aims to identify clinical/hormonal/molecular biomarkers associated with tumor size and invasiveness in GHomas and to analyze the influence of pre-treatment with somatostatin analogs (SSAs) or dopamine agonists (DAs) in key molecular biomarker expression. METHODS Clinical/analytical/radiological variables were evaluated in 192 patients from the REMAH study (ambispective multicenter post-surgery study of the Spanish Society of Endocrinology and Nutrition). The expression of somatostatin/ghrelin/dopamine system components and key pituitary/proliferation markers was evaluated in GHomas after the first surgery. Univariate/multivariate regression studies were performed to identify association between variables. RESULTS Eighty percent of patients harbor macroadenomas (63.8% with extrasellar growth). Associations between larger and more invasive GHomas with younger age, visual abnormalities, higher IGF1 levels, extrasellar/suprasellar growth, and/or cavernous sinus invasion were found. Higher GH1 and lower PRL/POMC/CGA/AVPR1B/DRD2T/DRD2L expression levels (P < .05) were associated with tumor invasiveness. Least Absolute Shrinkage and Selection Operator's penalized regression identified combinations of clinical and molecular features with areas under the curve between 0.67 and 0.82. Pre-operative therapy with DA or SSAs did not alter the expression of any of the markers analyzed except for DRD1/AVPR1B (up-regulated with DA) and FSHB/CRHR1 (down-regulated with SSAs). CONCLUSIONS A specific combination of clinical/analytical/molecular variables was found to be associated with tumor invasiveness and growth capacity in GHomas. Pre-treatment with first-line drugs for acromegaly did not significantly modify the expression of the most relevant biomarkers in our association model. These findings provide valuable insights for risk stratification and personalized management of GHomas.
Collapse
Affiliation(s)
- Miguel Sampedro-Nuñez
- Department of Endocrinology and Nutrition Hospital Universitario de la Princesa, Instituto de Investigación Sanitaria Princesa, Universidad Autónoma de Madrid, and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER GCV14/ER/12), Madrid, Spain
| | - Aura Dulcinea Herrera-Martínez
- Endocrinology and Nutrition Service, Reina Sofia University Hospital, Córboba, Spain
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Córboba, Spain
| | - Alejandro Ibáñez-Costa
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Córboba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain
- Reina Sofia University Hospital, Córdoba, Spain
| | - Esther Rivero-Cortés
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Córboba, Spain
- Reina Sofia University Hospital, Córdoba, Spain
| | - Eva Venegas
- Unidad de Gestión de Endocrinología y Nutrición, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, Sevilla, Spain
| | - Mercedes Robledo
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Rebeca Martínez-Hernández
- Department of Endocrinology and Nutrition Hospital Universitario de la Princesa, Instituto de Investigación Sanitaria Princesa, Universidad Autónoma de Madrid, and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER GCV14/ER/12), Madrid, Spain
| | - Araceli García-Martínez
- Alicante General University Hospital-Institute for Health and Biomedical Research (ISABIAL), Alicante, Spain
| | - Joan Gil
- Fundació Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Department of Endocrinology and Nutrition, Barcelona, Spain
| | - Mireia Jordà
- Fundació Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Department of Endocrinology and Nutrition, Barcelona, Spain
| | - Judith López-Fernández
- Servicio de Endocrinología y Nutrición, Hospital Universitario de Canarias, La Laguna, Santa Cruz de Tenerife, Spain
| | - Inmaculada Gavilán
- Hospital Universitario Puerta del Mar de Cádiz, Department of Endocrinology, Cádiz, Spain
| | - Silvia Maraver
- Servicio de Endocrinología y Nutrición, Hospital Universitario Virgen de la Victoria, Málaga, Spain
| | | | - Rosa Cámara
- Hospital Universitari i Politecnic La Fe, Department of Endocrinology, Valencia, Spain
| | | | - Elena Valassi
- Hospital Universitari Germans Trias i Pujol, Department of Endocrinology and Nutrition, Barcelona, Spain
| | - Elena Dios
- Virgen del Rocio University Hospital, Department of Endocrinology, Sevilla, Spain
| | - Anna Aulinas
- Hospital de la Santa Creu i Sant Pau, Department of Endocrinology, IIB-Sant Pau, CIBER de Enfermedades Raras (CIBER-ER), University of Vic-Central University of Catalonia, Barcelona, Spain
| | - Betina Biagetti
- Hospital Vall d'Hebron, Department of Endocrinology, Barcelona, Spain
| | | | | | - Concepción Blanco
- Hospital Universitario Principe de Asturias, Department of Endocrinology, Alcalá de Henares, Madrid, Spain
| | - de Miguel Paz
- Hospital Clinico San Carlos, Department of Endocrinology, Madrid, Spain
| | - Rocío Villar-Taibo
- Complejo Hospitalario Universitario de Santiago de Compostela, Department of Endocrinology, La Coruña, Spain
| | - Clara V Álvarez
- Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), University of Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - Sonia Gaztambide
- Biobizkaia Health Research Institute, Hospital Universitario Cruces, University of the Basque Country (UPV/EHU), CIBERDEM, CIBERER, EndoERN, Barakaldo, Bizkaia, Spain
| | - Susan M Webb
- Hospital de la Santa Creu i Sant Pau, Department of Endocrinology, IIB-Sant Pau, Research Center for Pituitary Diseases, CIBERER, Univ Autonoma Barcelona, Barcelona, Spain
| | - Luis Castaño
- Biobizkaia Health Research Institute, Hospital Universitario Cruces, University of the Basque Country (UPV/EHU), CIBERDEM, CIBERER, EndoERN, Barakaldo, Bizkaia, Spain
| | - Ignacio Bernabéu
- Complejo Hospitalario Universitario de Santiago de Compostela, Department of Endocrinology, Santiago de Compostela, A Coruña, Spain
| | - Antonio Picó
- Department of Endocrinology and Nutrition, Alicante General University Hospital, Alicante, Spain
- Institute for Health and Biomedical Research (ISABIAL), Alicante, Spain
- University Miguel Hernandez, CIBERER, Alicante, Spain
| | - María-Ángeles Gálvez
- Endocrinology and Nutrition Service, Reina Sofia University Hospital, Córboba, Spain
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Córboba, Spain
| | - Alfonso Soto-Moreno
- Unidad de Gestión de Endocrinología y Nutrición, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, Sevilla, Spain
| | - Manel Puig-Domingo
- Department of Endocrinology and Nutrition, Department of Medicine, Germans Trias i Pujol Research Institute and Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Justo P Castaño
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Córboba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain
- Reina Sofia University Hospital, Córdoba, Spain
| | - Mónica Marazuela
- Department of Endocrinology and Nutrition Hospital Universitario de la Princesa, Instituto de Investigación Sanitaria Princesa, Universidad Autónoma de Madrid, and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER GCV14/ER/12), Madrid, Spain
| | - Raúl M Luque
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Córboba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain
- Reina Sofia University Hospital, Córdoba, Spain
| |
Collapse
|
7
|
Chiloiro S, Moroni R, Giampietro A, Angelini F, Gessi M, Lauretti L, Mattogno PP, Calandrelli R, Tartaglione T, Carlino A, Gaudino S, Olivi A, Rindi G, De Marinis L, Pontecorvi A, Doglietto F, Bianchi A. The Multibiomarker Acro-TIME Score Predicts fg-SRLs Response: Preliminary Results of a Retrospective Acromegaly Cohort. J Clin Endocrinol Metab 2024; 109:1341-1350. [PMID: 37975821 DOI: 10.1210/clinem/dgad673] [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: 08/04/2023] [Revised: 10/15/2023] [Accepted: 11/15/2023] [Indexed: 11/19/2023]
Abstract
CONTEXT The prompt control of acromegaly is a primary treatment aim for reducing related disease morbidity and mortality. First-generation somatostatin receptor ligands (fg-SRLs) are the cornerstone of medical therapies. A non-negligible number of patients do not respond to this treatment. Several predictors of fg-SRL response were identified, but a comprehensive prognostic model is lacking. OBJECTIVE We aimed to design a prognostic model based on clinical and biochemical parameters, and pathological features, including data on immune tumor microenvironment. METHODS A retrospective, monocenter, cohort study was performed on 67 medically naïve patients with acromegaly. Fifteen clinical, pathological, and radiological features were collected and analyzed as independent risk factors of fg-SRLs response, using univariable and multivariable logistic regression analyses. A stepwise selection method was applied to identify the final regression model. A nomogram was then obtained. RESULTS Thirty-seven patients were fg-SRLs responders. An increased risk to poor response to fg-SRLs were observed in somatotropinomas with absent/cytoplasmatic SSTR2 expression (OR 5.493 95% CI 1.19-25.16, P = .028), with low CD68+/CD8+ ratio (OR 1.162, 95% CI 1.01-1.33, P = .032). Radical surgical resection was associated with a low risk of poor fg-SRLs response (OR 0.106, 95% CI 0.025-0.447 P = .002). The nomogram obtained from the stepwise regression model was based on the CD68+/CD8+ ratio, SSTR2 score, and the persistence of postsurgery residual tumor and was able to predict the response to fg-SRLs with good accuracy (area under the curve 0.85). CONCLUSION Although our predictive model should be validated in prospective studies, our data suggest that this nomogram may represent an easy to use tool for predicting the fg-SRL outcome early.
Collapse
Affiliation(s)
- Sabrina Chiloiro
- UOC Endocrinology and Diabetology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli, 00168 Roma, Italy
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo A. Gemelli, 00168 Roma, Italy
| | | | - Antonella Giampietro
- UOC Endocrinology and Diabetology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli, 00168 Roma, Italy
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo A. Gemelli, 00168 Roma, Italy
| | - Flavia Angelini
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo A. Gemelli, 00168 Roma, Italy
| | - Marco Gessi
- Department of Woman and Child Health Sciences and Public Health, Anatomic Pathology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli, 00168 Roma, Italy
| | - Liverana Lauretti
- Neurosurgery Unit, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Largo A. Gemelli, 00168 Roma, Italy
| | - Pier Paolo Mattogno
- Neurosurgery Unit, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Largo A. Gemelli, 00168 Roma, Italy
| | - Rosalinda Calandrelli
- UOSD Neuroradiologia Diagnostica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del S. Cuore, Largo A. Gemelli, 8, 00168 Rome, Italy
| | - Tommaso Tartaglione
- UOSD Neuroradiologia Diagnostica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del S. Cuore, Largo A. Gemelli, 8, 00168 Rome, Italy
| | - Angela Carlino
- Department of Woman and Child Health Sciences and Public Health, Anatomic Pathology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli, 00168 Roma, Italy
| | - Simona Gaudino
- UOSD Neuroradiologia Diagnostica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del S. Cuore, Largo A. Gemelli, 8, 00168 Rome, Italy
| | - Alessandro Olivi
- Neurosurgery Unit, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Largo A. Gemelli, 00168 Roma, Italy
| | - Guido Rindi
- Department of Woman and Child Health Sciences and Public Health, Anatomic Pathology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli, 00168 Roma, Italy
| | - Laura De Marinis
- UOC Endocrinology and Diabetology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli, 00168 Roma, Italy
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo A. Gemelli, 00168 Roma, Italy
| | - Alfredo Pontecorvi
- UOC Endocrinology and Diabetology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli, 00168 Roma, Italy
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo A. Gemelli, 00168 Roma, Italy
| | - Francesco Doglietto
- Neurosurgery Unit, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Largo A. Gemelli, 00168 Roma, Italy
| | - Antonio Bianchi
- UOC Endocrinology and Diabetology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli, 00168 Roma, Italy
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo A. Gemelli, 00168 Roma, Italy
| |
Collapse
|
8
|
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
|
9
|
Marques-Pamies M, Gil J, Jordà M, Puig-Domingo M. Predictors of Response to Treatment with First-Generation Somatostatin Receptor Ligands in Patients with Acromegaly. Arch Med Res 2023; 54:102924. [PMID: 38042683 DOI: 10.1016/j.arcmed.2023.102924] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 10/27/2023] [Accepted: 11/17/2023] [Indexed: 12/04/2023]
Abstract
BACKGROUND AND AIMS Predictors of first-generation somatostatin receptor ligands (fgSRLs) response in acromegaly have been studied for over 30 years, but they are still not recommended in clinical guidelines. Is there not enough evidence to support their use? This systematic review aims to describe the current knowledge of the main predictors of fgSRLs response and discuss their current usefulness, as well as future research directions. METHODS A systematic search was performed in the Scopus and PubMed databases for functional, imaging, and molecular predictive factors. RESULTS A total of 282 articles were detected, of which 64 were included. Most of them are retrospective studies performed between 1990 and 2023 focused on the predictive response to fgSRLs in acromegaly. The usefulness of the predictive factors is confirmed, with good response identified by the most replicated factors, specifically low GH nadir in the acute octreotide test, T2 MRI hypointensity, high Somatostatin receptor 2 (SSTR2) and E-cadherin expression, and a densely granulated pattern. Even if these biomarkers are interrelated, the association is quite heterogeneous. With classical statistical methods, it is complex to define reliable and generalizable cut-off values worth recommending in clinical guidelines. Machine-learning models involving omics are a promising approach to achieve the highest accuracy values to date. CONCLUSIONS This survey confirms a sufficiently robust level of evidence to apply knowledge of predictive factors for greater efficiency in the treatment decision process. The irruption of artificial intelligence in this field is providing definitive answers to such long-standing questions that may change clinical guidelines and make personalized medicine a reality.
Collapse
Affiliation(s)
| | - Joan Gil
- Endocrine Research Unit, Germans Trias i Pujol Research Institute, Badalona, Spain; Network Research Center for Rare Diseases, CIBERER, Unit 747, Instituto de Salud Carlos III, Madrid, Spain; Department of Endocrinology, Research Center for Pituitary Diseases, Hospital Sant Pau, IIB-SPau, Barcelona, Spain
| | - Mireia Jordà
- Endocrine Research Unit, Germans Trias i Pujol Research Institute, Badalona, Spain
| | - Manel Puig-Domingo
- Endocrine Research Unit, Germans Trias i Pujol Research Institute, Badalona, Spain; Network Research Center for Rare Diseases, CIBERER, Unit 747, Instituto de Salud Carlos III, Madrid, Spain; Department of Endocrinology and Nutrition, Germans Trias i Pujol University Hospital, Badalona, Spain; Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain.
| |
Collapse
|
10
|
Zhao J, Fu H, Yu J, Hong W, Tian X, Qi J, Sun S, Zhao C, Wu C, Xu Z, Cheng L, Chai R, Yan W, Wei X, Shao Z. Prospect of acromegaly therapy: molecular mechanism of clinical drugs octreotide and paltusotine. Nat Commun 2023; 14:962. [PMID: 36810324 PMCID: PMC9944328 DOI: 10.1038/s41467-023-36673-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 02/07/2023] [Indexed: 02/23/2023] Open
Abstract
Somatostatin receptor 2 (SSTR2) is highly expressed in neuroendocrine tumors and represents as a therapeutic target. Several peptide analogs mimicking the endogenous ligand somatostatin are available for clinical use, but poor therapeutic effects occur in a subset of patients, which may be correlated with subtype selectivity or cell surface expression. Here, we clarify the signal bias profiles of the first-generation peptide drug octreotide and a new-generation small molecule paltusotine by evaluating their pharmacological characteristics. We then perform cryo-electron microscopy analysis of SSTR2-Gi complexes to determine how the drugs activate SSTR2 in a selective manner. In this work, we decipher the mechanism of ligand recognition, subtype selectivity and signal bias property of SSTR2 sensing octreotide and paltusotine, which may aid in designing therapeutic drugs with specific pharmacological profiles against neuroendocrine tumors.
Collapse
Affiliation(s)
- Jie Zhao
- Division of Nephrology and Kidney Research Institute, Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Hong Fu
- Division of Nephrology and Kidney Research Institute, Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jingjing Yu
- Division of Nephrology and Kidney Research Institute, Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Weiqi Hong
- Division of Nephrology and Kidney Research Institute, Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xiaowen Tian
- Division of Nephrology and Kidney Research Institute, Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jieyu Qi
- State Key Laboratory of Bioelectronics, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Suyue Sun
- Division of Nephrology and Kidney Research Institute, Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Chang Zhao
- Division of Nephrology and Kidney Research Institute, Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Chao Wu
- Division of Nephrology and Kidney Research Institute, Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Zheng Xu
- Division of Nephrology and Kidney Research Institute, Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Lin Cheng
- Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Renjie Chai
- State Key Laboratory of Bioelectronics, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China. .,Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China. .,Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
| | - Wei Yan
- Division of Nephrology and Kidney Research Institute, Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| | - Xiawei Wei
- Division of Nephrology and Kidney Research Institute, Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| | - Zhenhua Shao
- Division of Nephrology and Kidney Research Institute, Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China. .,Department of Nephrology, Hainan General Hospital, Haikou, Hainan, 570311, China.
| |
Collapse
|
11
|
Proteogenomic landscape and clinical characterization of GH-producing pituitary adenomas/somatotroph pituitary neuroendocrine tumors. Commun Biol 2022; 5:1304. [PMID: 36435867 PMCID: PMC9701206 DOI: 10.1038/s42003-022-04272-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 11/16/2022] [Indexed: 11/28/2022] Open
Abstract
The clinical characteristics of growth hormone (GH)-producing pituitary adenomas/somatotroph pituitary neuroendocrine tumors (GHomas/somatotroph PitNETs) vary across patients. In this study, we aimed to integrate the genetic alterations, protein expression profiles, transcriptomes, and clinical characteristics of GHomas/somatotroph PitNETs to identify molecules associated with acromegaly characteristics. Targeted capture sequencing and copy number analysis of 36 genes and nontargeted proteomics analysis were performed on fresh-frozen samples from 121 sporadic GHomas/somatotroph PitNETs. Targeted capture sequencing revealed GNAS as the only driver gene, as previously reported. Classification by consensus clustering using both RNA sequencing and proteomics revealed many similarities between the proteome and the transcriptome. Gene ontology analysis was performed for differentially expressed proteins between wild-type and mutant GNAS samples identified by nontargeted proteomics and involved in G protein-coupled receptor (GPCR) pathways. The results suggested that GNAS mutations impact endocrinological features in acromegaly through GPCR pathway induction. ATP2A2 and ARID5B correlated with the GH change rate in the octreotide loading test, and WWC3, SERINC1, and ZFAND3 correlated with the tumor volume change rate after somatostatin analog treatment. These results identified a biological connection between GNAS mutations and the clinical and biochemical characteristics of acromegaly, revealing molecules associated with acromegaly that may affect medical treatment efficacy.
Collapse
|
12
|
Gatto F, Feelders RA, van Koetsveld PM, Dogan F, Neggers SJCCMM, van der Lelij AJ, Amarù J, Ferone D, Hofland LJ. Dissecting the in vitro efficacy of octreotide and cabergoline in GH- and GH/PRL-secreting pituitary tumors. J Clin Endocrinol Metab 2022; 108:e98-e109. [PMID: 36413489 DOI: 10.1210/clinem/dgac675] [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] [Received: 07/08/2022] [Revised: 11/17/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022]
Abstract
CONTEXT Cabergoline (CAB) is an off-label medical therapy for acromegaly, overshadowed by first-generation somatostatin receptor ligands, e.g. octreotide (OCT). OBJECTIVE Head-to-head comparison between OCT and CAB in inhibiting growth hormone (GH) secretion in primary cultures of GH- and GH/PRL-secreting tumors. To investigate the role of somatostatin (SST) and dopamine type 2 (D2R) receptor expression. DESIGN To evaluate the antisecretory effect of OCT and CAB, together with receptor mRNA expression, in 23 tumor cultures. SETTING AND PATIENTS Acromegaly patients referred to the Erasmus Medical Center (Rotterdam, The Netherlands). INTERVENTIONS 72-hour OCT and CAB treatment (10 nM). MAIN OUTCOME MEASURES GH concentrations in cell culture media. RESULTS OCT showed a slightly higher efficacy compared with CAB (GH decrease -39.5% vs. -32.5%, p = 0.079). The effect of the two drugs was superimposable in GH/PRL co-secreting tumors (-42.1% vs. -44.8%), where SST1 and D2R had a higher expression compared to the pure GH-secreting ones (p = 0.020 and p = 0.026). OCT was more effective than CAB in 8/23 cultures, while CAB was more effective than OCT in 3/23 (CAB + group). In CAB + tumors, SST1 expression was higher compared to the other groups (p = 0.034). At ROC curve analysis, SST1 and D2R discriminated between GH and GH/PRL co-secretion (AUC 0.856, p = 0.013; AUC 0.822, p = 0.024). SST1 was the best predictor of CAB response (≥50% GH reduction, AUC 0.913, p = 0.006; 80% sensitivity, 94% specificity). CONCLUSIONS OCT is 5-10% more effective than CAB in vitro. SST1 mRNA expression can represent a reliable marker of GH/PRL co-secreting tumors showing a preferential response to CAB treatment.
Collapse
Affiliation(s)
- Federico Gatto
- Endocrinology Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Internal Medicine, Division of Endocrinology
- Pituitary Center Rotterdam, Erasmus MC, Rotterdam, The Netherlands
| | - Richard A Feelders
- Department of Internal Medicine, Division of Endocrinology
- Pituitary Center Rotterdam, Erasmus MC, Rotterdam, The Netherlands
| | | | - Fadime Dogan
- Department of Internal Medicine, Division of Endocrinology
| | - Sebastian J C C M M Neggers
- Department of Internal Medicine, Division of Endocrinology
- Pituitary Center Rotterdam, Erasmus MC, Rotterdam, The Netherlands
| | - Aart-Jan van der Lelij
- Department of Internal Medicine, Division of Endocrinology
- Pituitary Center Rotterdam, Erasmus MC, Rotterdam, The Netherlands
| | - Jessica Amarù
- Endocrinology Unit, Department of Internal Medicine and Medical Specialties, University of Genova, Italy
| | - Diego Ferone
- Endocrinology Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Endocrinology Unit, Department of Internal Medicine and Medical Specialties, University of Genova, Italy
| | - Leo J Hofland
- Department of Internal Medicine, Division of Endocrinology
- Pituitary Center Rotterdam, Erasmus MC, Rotterdam, The Netherlands
| |
Collapse
|
13
|
Moreno-Moreno P, Ibáñez-Costa A, Venegas-Moreno E, Fuentes-Fayos AC, Alhambra-Expósito MR, Fajardo-Montañana C, García-Martínez A, Dios E, Vázquez-Borrego MC, Remón-Ruiz P, Cámara R, Lamas C, Carlos Padillo-Cuenca J, Solivera J, Cano DA, Gahete MD, Herrera-Martínez AD, Picó A, Soto-Moreno A, Gálvez-Moreno MÁ, Castaño JP, Luque RM. Integrative Clinical, Radiological, and Molecular Analysis for Predicting Remission and Recurrence of Cushing Disease. J Clin Endocrinol Metab 2022; 107:e2938-e2951. [PMID: 35312002 DOI: 10.1210/clinem/dgac172] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Indexed: 11/19/2022]
Abstract
CONTEXT Adrenocorticotropin (ACTH)-secreting pituitary tumors (ACTHomas) are associated with severe comorbidities and increased mortality. Current treatments mainly focus on remission and prevention of persistent disease and recurrence. However, there are still no useful biomarkers to accurately predict the clinical outcome after surgery, long-term remission, or disease relapse. OBJECTIVES This work aimed to identify clinical, biochemical, and molecular markers for predicting long-term clinical outcome and remission in ACTHomas. METHODS A retrospective multicenter study was performed with 60 ACTHomas patients diagnosed between 2004 and 2018 with at least 2 years' follow-up. Clinical/biochemical variables were evaluated yearly. Molecular expression profile of the somatostatin/ghrelin/dopamine regulatory systems components and of key pituitary factors and proliferation markers were evaluated in tumor samples after the first surgery. RESULTS Clinical variables including tumor size, time until diagnosis/first surgery, serum prolactin, and postsurgery cortisol levels were associated with tumor remission and relapsed disease. The molecular markers analyzed were distinctly expressed in ACTHomas, with some components (ie, SSTR1, CRHR1, and MKI67) showing instructive associations with recurrence and/or remission. Notably, an integrative model including selected clinical variables (tumor size/postsurgery serum cortisol), and molecular markers (SSTR1/CRHR1) can accurately predict the clinical evolution and remission of patients with ACTHomas, generating a receiver operating characteristic curve with an area under the curve of 1 (P < .001). CONCLUSION This study demonstrates that the combination of a set of clinical and molecular biomarkers in ACTHomas is able to accurately predict the clinical evolution and remission of patients. Consequently, the postsurgery molecular profile represents a valuable tool for clinical evaluation and follow-up of patients with ACTHomas.
Collapse
Affiliation(s)
- Paloma Moreno-Moreno
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain
- Reina Sofia University Hospital (HURS), 14004 Cordoba, Spain
- Service of Endocrinology and Nutrition, IMIBIC, HURS, 14004 Cordoba, Spain
| | - Alejandro Ibáñez-Costa
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain
- Reina Sofia University Hospital (HURS), 14004 Cordoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004 Cordoba, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004 Cordoba, Spain
| | - Eva Venegas-Moreno
- Unidad de Gestión de Endocrinología y Nutrición. Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Sevilla, Spain
| | - Antonio C Fuentes-Fayos
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain
- Reina Sofia University Hospital (HURS), 14004 Cordoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004 Cordoba, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004 Cordoba, Spain
| | - María R Alhambra-Expósito
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain
- Reina Sofia University Hospital (HURS), 14004 Cordoba, Spain
- Service of Endocrinology and Nutrition, IMIBIC, HURS, 14004 Cordoba, Spain
| | - Carmen Fajardo-Montañana
- Department of Endocrinology, Hospital Universitario de La Ribera, Alzira, 46600, Valencia, Spain
| | - Araceli García-Martínez
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain
- Reina Sofia University Hospital (HURS), 14004 Cordoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004 Cordoba, Spain
| | - Elena Dios
- Unidad de Gestión de Endocrinología y Nutrición. Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Sevilla, Spain
| | - Mari C Vázquez-Borrego
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain
- Reina Sofia University Hospital (HURS), 14004 Cordoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004 Cordoba, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004 Cordoba, Spain
| | - Pablo Remón-Ruiz
- Unidad de Gestión de Endocrinología y Nutrición. Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Sevilla, Spain
| | - Rosa Cámara
- Department of Endocrinology and Nutrition, Polytechnic University Hospital La Fe, 46026, Valencia, Spain
| | - Cristina Lamas
- Department of Endocrinology and Nutrition, Albacete University Hospital, 02006, Albacete, Spain
| | - José Carlos Padillo-Cuenca
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain
- Reina Sofia University Hospital (HURS), 14004 Cordoba, Spain
- Service of Endocrinology and Nutrition, IMIBIC, HURS, 14004 Cordoba, Spain
| | | | - David A Cano
- Unidad de Gestión de Endocrinología y Nutrición. Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Sevilla, Spain
| | - Manuel D Gahete
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain
- Reina Sofia University Hospital (HURS), 14004 Cordoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004 Cordoba, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004 Cordoba, Spain
| | - Aura D Herrera-Martínez
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain
- Reina Sofia University Hospital (HURS), 14004 Cordoba, Spain
- Service of Endocrinology and Nutrition, IMIBIC, HURS, 14004 Cordoba, Spain
| | - Antonio Picó
- Department of Endocrinology and Nutrition, Alicante General University Hospital. Institute for Health and Biomedical Research (ISABIAL). University Miguel Hernandez, CIBER Rare Diseases, 03010, Alicante, Spain
| | - Alfonso Soto-Moreno
- Unidad de Gestión de Endocrinología y Nutrición. Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Sevilla, Spain
| | - María Ángeles Gálvez-Moreno
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain
- Reina Sofia University Hospital (HURS), 14004 Cordoba, Spain
- Service of Endocrinology and Nutrition, IMIBIC, HURS, 14004 Cordoba, Spain
| | - Justo P Castaño
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain
- Reina Sofia University Hospital (HURS), 14004 Cordoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004 Cordoba, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004 Cordoba, Spain
| | - Raúl M Luque
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain
- Reina Sofia University Hospital (HURS), 14004 Cordoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004 Cordoba, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004 Cordoba, Spain
| |
Collapse
|
14
|
Data mining analyses for precision medicine in acromegaly: a proof of concept. Sci Rep 2022; 12:8979. [PMID: 35643771 PMCID: PMC9148300 DOI: 10.1038/s41598-022-12955-2] [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: 03/16/2022] [Accepted: 05/13/2022] [Indexed: 11/21/2022] Open
Abstract
Predicting which acromegaly patients could benefit from somatostatin receptor ligands (SRL) is a must for personalized medicine. Although many biomarkers linked to SRL response have been identified, there is no consensus criterion on how to assign this pharmacologic treatment according to biomarker levels. Our aim is to provide better predictive tools for an accurate acromegaly patient stratification regarding the ability to respond to SRL. We took advantage of a multicenter study of 71 acromegaly patients and we used advanced mathematical modelling to predict SRL response combining molecular and clinical information. Different models of patient stratification were obtained, with a much higher accuracy when the studied cohort is fragmented according to relevant clinical characteristics. Considering all the models, a patient stratification based on the extrasellar growth of the tumor, sex, age and the expression of E-cadherin, GHRL, IN1-GHRL, DRD2, SSTR5 and PEBP1 is proposed, with accuracies that stand between 71 to 95%. In conclusion, the use of data mining could be very useful for implementation of personalized medicine in acromegaly through an interdisciplinary work between computer science, mathematics, biology and medicine. This new methodology opens a door to more precise and personalized medicine for acromegaly patients.
Collapse
|
15
|
Rass L, Rahvar AH, Matschke J, Saeger W, Renné T, Aberle J, Flitsch J, Rotermund R. Differences in somatostatin receptor subtype expression in patients with acromegaly: new directions for targeted therapy? Hormones (Athens) 2022; 21:79-89. [PMID: 34674191 PMCID: PMC8818633 DOI: 10.1007/s42000-021-00327-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 10/04/2021] [Indexed: 11/02/2022]
Abstract
PURPOSE To analyze the expression of somatostatin receptor (SSTR)2a and 5 by immunohistochemistry (IHC) in surgically resected somatotrophic pituitary adenomas and to associate expression rates with tumor size and clinical, biochemical, and histological parameters and response to somatostatin analog (SA) therapy. METHODS Forty-three microsurgically treated patients with histopathologically proven growth hormone (GH)-producing pituitary adenoma were included (WHO 2017). SSTR subtype expression was analyzed in adenoma tissues using monoclonal antibodies (Abcam, SSTR2a-UMB1, SSTR5-UMB4). Expression rates were classified as low (≤ 20% staining positivity), moderate (21-50%), and high (> 50%). Furthermore, biochemical parameters such as human growth hormone (hGH) and insulin-like growth factor-1 (IGF-1) levels were measured and clinical, biochemical, radiological, and histological data were evaluated. RESULTS Of the 43 patients included in this study, 28 were female and 15 were male. The median age was 52 years (range 17-72 years). The median tumor size was 1.2 cm (range: 0.13-3.93 cm). All resected tumors showed positivity for somatotrophic hormone (STH). In all tissue samples, SSTR2a signal expression was detectable in immunohistochemistry, while only 39 samples were positive for SSTR5. Thirty-six samples had a high expression of SSTR2a, while three had a moderate and four a low SSTR2a signal. In comparison, SSTR5 signal was high in 26 out of 43 samples, while seven adenomas showed a moderate and six cases a low expression rate of SSTR5. The median IGF-1 was 714.2 µg/l and the median GH 19.6 mU/l (= 6.53 µg/l). The present study indicates that there is no significant relationship between the expression rates of receptor subtypes and the parameters we analyzed. However, our study revealed that smaller adenomas have a lower baseline GH level (p = 0.015), CONCLUSION: IHC with monoclonal antibodies appears to be a suitable method to determine the expression rates of SSTR2a and 5 at protein levels, as it is not possible to draw conclusions regarding receptor subtypes solely on the basis of the parameters analyzed.
Collapse
Affiliation(s)
- Lena Rass
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Amir-Hossein Rahvar
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jakob Matschke
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Wolfgang Saeger
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thomas Renné
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jens Aberle
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jörg Flitsch
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Roman Rotermund
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany.
| |
Collapse
|
16
|
Robertson MJ, Meyerowitz JG, Panova O, Borrelli K, Skiniotis G. Plasticity in ligand recognition at somatostatin receptors. Nat Struct Mol Biol 2022; 29:210-217. [PMID: 35210615 PMCID: PMC11073612 DOI: 10.1038/s41594-022-00727-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 01/14/2022] [Indexed: 11/10/2022]
Abstract
Somatostatin is a signaling peptide that plays a pivotal role in physiologic processes relating to metabolism and growth through its actions at somatostatin receptors (SSTRs). Members of the SSTR subfamily, particularly SSTR2, are key drug targets for neuroendocrine neoplasms, with synthetic peptide agonists currently in clinical use. Here, we show the cryogenic-electron microscopy structures of active-state SSTR2 in complex with heterotrimeric Gi3 and either the endogenous ligand SST14 or the FDA-approved drug octreotide. Complemented by biochemical assays and molecular dynamics simulations, these structures reveal key details of ligand recognition and receptor activation at SSTRs. We find that SSTR ligand recognition is highly diverse, as demonstrated by ligand-induced conformational changes in ECL2 and substantial sequence divergence across subtypes in extracellular regions. Despite this complexity, we rationalize several known sources of SSTR subtype selectivity and identify an additional interaction for specific binding. These results provide valuable insights for structure-based drug discovery at SSTRs.
Collapse
Affiliation(s)
- Michael J Robertson
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Justin G Meyerowitz
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Ouliana Panova
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Georgios Skiniotis
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
17
|
Gadelha MR, Wildemberg LE, Kasuki L. The Future of Somatostatin Receptor Ligands in Acromegaly. J Clin Endocrinol Metab 2022; 107:297-308. [PMID: 34618894 PMCID: PMC8764337 DOI: 10.1210/clinem/dgab726] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Indexed: 12/13/2022]
Abstract
Currently, the first-generation somatostatin receptor ligands (fg-SRLs), octreotide LAR and lanreotide autogel, are the mainstays of acromegaly treatment and achieve biochemical control in approximately 40% of patients and tumor shrinkage in over 60% of patients. Pasireotide, a second-generation SRL, shows higher efficacy with respect to both biochemical control and tumor shrinkage but has a worse safety profile. In this review, we discuss the future perspectives of currently available SRLs, focusing on the use of biomarkers of response and precision medicine, new formulations of these SRLs and new drugs, which are under development. Precision medicine, which is based on biomarkers of response to treatment, will help guide the decision-making process by allowing physicians to choose the appropriate drug for each patient and improving response rates. New formulations of available SRLs, such as oral, subcutaneous depot, and nasal octreotide, may improve patients' adherence to treatment and quality of life since there will be more options available that better suit each patient. Finally, new drugs, such as paltusotine, somatropin, ONO-5788, and ONO-ST-468, may improve treatment adherence and present higher efficacy than currently available drugs.
Collapse
Affiliation(s)
- Monica R Gadelha
- Endocrine Unit and Neuroendocrinology Research Center, Medical School and Hospital Universitário Clementino Fraga Filho - Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-913, Brazil
- Neuroendocrine Unit - Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria Estadual de Saúde, Rio de Janeiro, 20231-092, Brazil
- Neuropathology and Molecular Genetics Laboratory, Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria Estadual de Saúde, Rio de Janeiro, 20231-092, Brazil
- Correspondence: Mônica R. Gadelha, Rua Prof. Rodolpho Paulo Rocco, 255, 9th floor, Ilha do Fundão, Rio de Janeiro 21941-913, Brazil.
| | - Luiz Eduardo Wildemberg
- Endocrine Unit and Neuroendocrinology Research Center, Medical School and Hospital Universitário Clementino Fraga Filho - Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-913, Brazil
- Neuroendocrine Unit - Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria Estadual de Saúde, Rio de Janeiro, 20231-092, Brazil
| | - Leandro Kasuki
- Endocrine Unit and Neuroendocrinology Research Center, Medical School and Hospital Universitário Clementino Fraga Filho - Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-913, Brazil
- Neuroendocrine Unit - Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria Estadual de Saúde, Rio de Janeiro, 20231-092, Brazil
| |
Collapse
|
18
|
Peverelli E, Treppiedi D, Mangili F, Catalano R, Spada A, Mantovani G. Drug resistance in pituitary tumours: from cell membrane to intracellular signalling. Nat Rev Endocrinol 2021; 17:560-571. [PMID: 34194011 DOI: 10.1038/s41574-021-00514-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/19/2021] [Indexed: 02/06/2023]
Abstract
The pharmacological treatment of pituitary tumours is based on the use of stable analogues of somatostatin and dopamine. The analogues bind to somatostatin receptor types 2 and 5 (SST2 and SST5) and dopamine receptor type 2 (DRD2), respectively, and generate signal transduction cascades in cancerous pituitary cells that culminate in the inhibition of hormone secretion, cell growth and invasion. Drug resistance occurs in a subset of patients and can involve different steps at different stages, such as following receptor activation by the agonist or during the final biological responses. Although the expression of somatostatin and dopamine receptors in cancer cells is a prerequisite for these drugs to reach a biological effect, their presence does not guarantee the success of the therapy. Successful therapy also requires the proper functioning of the machinery of signal transduction and the finely tuned regulation of receptor desensitization, internalization and intracellular trafficking. The present Review provides an updated overview of the molecular factors underlying the pharmacological resistance of pituitary tumours. The Review discusses the experimental evidence that supports a role for receptors and intracellular proteins in the function of SSTs and DRD2 and their clinical importance.
Collapse
Affiliation(s)
- Erika Peverelli
- University of Milan, Department of Clinical Sciences and Community Health, Milan, Italy.
| | - Donatella Treppiedi
- University of Milan, Department of Clinical Sciences and Community Health, Milan, Italy
| | - Federica Mangili
- University of Milan, Department of Clinical Sciences and Community Health, Milan, Italy
| | - Rosa Catalano
- University of Milan, Department of Clinical Sciences and Community Health, Milan, Italy
- PhD Program in Endocrinological Sciences, Sapienza University of Rome, Rome, Italy
| | - Anna Spada
- University of Milan, Department of Clinical Sciences and Community Health, Milan, Italy
| | - Giovanna Mantovani
- University of Milan, Department of Clinical Sciences and Community Health, Milan, Italy
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Endocrinology Unit, Milan, Italy
| |
Collapse
|
19
|
Wildemberg LE, da Silva Camacho AH, Miranda RL, Elias PCL, de Castro Musolino NR, Nazato D, Jallad R, Huayllas MKP, Mota JIS, Almeida T, Portes E, Ribeiro-Oliveira A, Vilar L, Boguszewski CL, Winter Tavares AB, Nunes-Nogueira VS, Mazzuco TL, Rech CGSL, Marques NV, Chimelli L, Czepielewski M, Bronstein MD, Abucham J, de Castro M, Kasuki L, Gadelha M. Machine Learning-based Prediction Model for Treatment of Acromegaly With First-generation Somatostatin Receptor Ligands. J Clin Endocrinol Metab 2021; 106:2047-2056. [PMID: 33686418 DOI: 10.1210/clinem/dgab125] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Indexed: 01/12/2023]
Abstract
CONTEXT Artificial intelligence (AI), in particular machine learning (ML), may be used to deeply analyze biomarkers of response to first-generation somatostatin receptor ligands (fg-SRLs) in the treatment of acromegaly. OBJECTIVE To develop a prediction model of therapeutic response of acromegaly to fg-SRL. METHODS Patients with acromegaly not cured by primary surgical treatment and who had adjuvant therapy with fg-SRL for at least 6 months after surgery were included. Patients were considered controlled if they presented growth hormone (GH) <1.0 ng/mL and normal age-adjusted insulin-like growth factor (IGF)-I levels. Six AI models were evaluated: logistic regression, k-nearest neighbor classifier, support vector machine, gradient-boosted classifier, random forest, and multilayer perceptron. The features included in the analysis were age at diagnosis, sex, GH, and IGF-I levels at diagnosis and at pretreatment, somatostatin receptor subtype 2 and 5 (SST2 and SST5) protein expression and cytokeratin granulation pattern (GP). RESULTS A total of 153 patients were analyzed. Controlled patients were older (P = .002), had lower GH at diagnosis (P = .01), had lower pretreatment GH and IGF-I (P < .001), and more frequently harbored tumors that were densely granulated (P = .014) or highly expressed SST2 (P < .001). The model that performed best was the support vector machine with the features SST2, SST5, GP, sex, age, and pretreatment GH and IGF-I levels. It had an accuracy of 86.3%, positive predictive value of 83.3% and negative predictive value of 87.5%. CONCLUSION We developed a ML-based prediction model with high accuracy that has the potential to improve medical management of acromegaly, optimize biochemical control, decrease long-term morbidities and mortality, and reduce health services costs.
Collapse
Affiliation(s)
- Luiz Eduardo Wildemberg
- Endocrine Unit and Neuroendocrinology Research Center, Medical School and Hospital Universitário Clementino Fraga Filho-Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
- Neuroendocrine Unit-Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria Estadual de Saúde , Rio de Janeiro, RJ, Brazil
| | - Aline Helen da Silva Camacho
- Neuropathology and Molecular Genetics Laboratory, Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria Estadual de Saúde, Rio de Janeiro, RJ, Brazil
| | - Renan Lyra Miranda
- Neuropathology and Molecular Genetics Laboratory, Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria Estadual de Saúde, Rio de Janeiro, RJ, Brazil
| | - Paula C L Elias
- Division of Endocrinology-Department of Internal Medicine, Ribeirao Preto Medical School-University of Sao Paulo, São Paulo, SP, Brazil
| | - Nina R de Castro Musolino
- Neuroendocrine Unit, Division of Functional Neurosurgery, Hospital das Clinicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Debora Nazato
- Neuroendocrine Unit-Division of Endocrinology and Metabolism-Escola Paulista de Medicina-Universidade Federal de São Paulo (Unifesp), São Paulo, SP, Brazil
| | - Raquel Jallad
- Neuroendocrine Unit, Division of Endocrinology and Metabolism, Hospital das Clínicas, University of São Paulo Medical School, São Paulo, SP, Brazil
- Cellular and Molecular Endocrinology Laboratory/LIM25, Discipline of Endocrinology, Hospital das Clinicas HCFMUSP, Faculty of Medicine, University of Sao Paulo, São Paulo, SP, Brazil
| | - Martha K P Huayllas
- Neuroendocrinology and Neurosurgery unit Hospital Brigadeiro, São Paulo, SP, Brazil
| | - Jose Italo S Mota
- Endocrinology and Metabolism Unit, Hospital Geral de Fortaleza, Secretaria Estadual de Saúde, Fortaleza, CE, Brazil
| | - Tobias Almeida
- Division of Endocrinology, Hospital de Clinicas de Porto Alegre (UFRGS), Porto Alegre, RS, Brazil
| | - Evandro Portes
- Institute of Medical Assistance to the State Public Hospital, São Paulo, SP, Brazil
| | | | - Lucio Vilar
- Neuroendocrine Unit, Division of Endocrinology and Metabolism, Hospital das Clínicas, Federal University of Pernambuco Medical School, Recife, PE, Brazil
| | - Cesar Luiz Boguszewski
- Endocrine Division (SEMPR), Department of Internal Medicine, Universidade Federal do Parana, Curitiba, PR, Brazil
| | - Ana Beatriz Winter Tavares
- Endocrine Unit-Department of Internal Medicine, Faculty of Medical Sciences, Universidade do Estado do Rio de Janeiro, RJ, Brazil
| | - Vania S Nunes-Nogueira
- Department of Internal Medicine, São Paulo State University/UNESP, Medical School, Botucatu, SP, Brazil
| | - Tânia Longo Mazzuco
- Division of Endocrinology of Medical Clinical Department, Universidade Estadual de Londrina (UEL), Londrina, PR, Brazil
| | | | - Nelma Veronica Marques
- Endocrine Unit and Neuroendocrinology Research Center, Medical School and Hospital Universitário Clementino Fraga Filho-Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Leila Chimelli
- Neuropathology and Molecular Genetics Laboratory, Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria Estadual de Saúde, Rio de Janeiro, RJ, Brazil
| | - Mauro Czepielewski
- Division of Endocrinology, Hospital de Clinicas de Porto Alegre (UFRGS), Porto Alegre, RS, Brazil
| | - Marcello D Bronstein
- Neuroendocrine Unit, Division of Endocrinology and Metabolism, Hospital das Clínicas, University of São Paulo Medical School, São Paulo, SP, Brazil
- Cellular and Molecular Endocrinology Laboratory/LIM25, Discipline of Endocrinology, Hospital das Clinicas HCFMUSP, Faculty of Medicine, University of Sao Paulo, São Paulo, SP, Brazil
| | - Julio Abucham
- Neuroendocrine Unit-Division of Endocrinology and Metabolism-Escola Paulista de Medicina-Universidade Federal de São Paulo (Unifesp), São Paulo, SP, Brazil
| | - Margaret de Castro
- Division of Endocrinology-Department of Internal Medicine, Ribeirao Preto Medical School-University of Sao Paulo, São Paulo, SP, Brazil
| | - Leandro Kasuki
- Endocrine Unit and Neuroendocrinology Research Center, Medical School and Hospital Universitário Clementino Fraga Filho-Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
- Neuroendocrine Unit-Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria Estadual de Saúde , Rio de Janeiro, RJ, Brazil
| | - Mônica Gadelha
- Endocrine Unit and Neuroendocrinology Research Center, Medical School and Hospital Universitário Clementino Fraga Filho-Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
- Neuroendocrine Unit-Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria Estadual de Saúde , Rio de Janeiro, RJ, Brazil
- Neuropathology and Molecular Genetics Laboratory, Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria Estadual de Saúde, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
20
|
Swanson AA, Erickson D, Donegan DM, Jenkins SM, Van Gompel JJ, Atkinson JLD, Erickson BJ, Giannini C. Clinical, biological, radiological, and pathological comparison of sparsely and densely granulated somatotroph adenomas: a single center experience from a cohort of 131 patients with acromegaly. Pituitary 2021; 24:192-206. [PMID: 33074402 DOI: 10.1007/s11102-020-01096-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/13/2020] [Indexed: 01/01/2023]
Abstract
PURPOSE Growth hormone-producing pituitary adenomas are divided into two clinically relevant histologic subtypes, densely (DG-A) and sparsely (SG-A) granulated. Histologic subtype was evaluated in a large cohort of patients with acromegaly, separating DG-A and SG-A, and correlated with clinicopathological characteristics. METHODS Patients with acromegaly undergoing surgery as initial therapy between 1995 and 2015 were identified. Histologic subtype was determined by keratin expression pattern with CAM5.2 and correlated with clinical and imaging parameters, somatostatin receptor subtype 2 (SST2) expression, post-surgical remission rate, and application of a prognostic scoring system incorporating proliferation and invasiveness. RESULTS One hundred thirty-one patients were included. Tumors were classified as DG-A (75, 57.3%), SG-A (29, 22.1%), intermediate (I-A) (9, 6.9%), and unclassified (18, 13.7%) when CAM5.2 was negative. DG-A and I-A were combined for analysis (DG/I-A) and compared to SG-A. Age, gender, proliferation, and post-surgical remission did not differ. SG-A were larger [2 vs. 1.5 cm (median), p = 0.03], more frequently invasive [65.5% vs. 32.9%, p = 0.004], associated with higher MRI T2-weighted signal ratio [1.01 vs. 0.82 (median), p = 0.01], showed lower SST2 expression (p < 0.0001), and scored higher in the prognostic classification (p = 0.004). Surgical remission occurred in 41.7% DG/I-A and 41.4% SG-A (p = 1.0). On multivariate analysis, absence of invasion (p = 0.009) and lower pre-operative IGF-1 index (p = 0.0002) were associated with post-surgical remission. CONCLUSION CAM5.2 allowed distinction between DG/I-A and SG-A in most but not all cases. Histologic subtype did not predict surgical outcome. Absence of invasion and lower pre-operative IGF-1 index were the only significant predictors of post-surgical remission in this cohort.
Collapse
Affiliation(s)
- Amy A Swanson
- Division of Anatomic Pathology, Mayo Clinic, Rochester, MN, USA
| | - Dana Erickson
- Division of Endocrinology, Diabetes and Metabolism, Mayo Clinic, Rochester, MN, USA
| | - Diane Mary Donegan
- Division of Endocrinology, Diabetes and Metabolism, Mayo Clinic, Rochester, MN, USA
- Division of Endocrinology, Diabetes and Metabolism, Indiana University, Indianapolis, IN, USA
| | - Sarah M Jenkins
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | | | | | | | - Caterina Giannini
- Division of Anatomic Pathology, Mayo Clinic, Rochester, MN, USA.
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
21
|
Störmann S, Schopohl J, Bullmann C, Terkamp C, Christ-Crain M, Finke R, Flitsch J, Kreitschmann-Andermahr I, Luger A, Stalla G, Houchard A, Helbig D, Petersenn S. Multicenter, Observational Study of Lanreotide Autogel for the Treatment of Patients with Acromegaly in Routine Clinical Practice in Germany, Austria and Switzerland. Exp Clin Endocrinol Diabetes 2020; 129:224-233. [PMID: 33285601 DOI: 10.1055/a-1247-4713] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND Evidence from controlled trials has shown that lanreotide autogel is effective in achieving biochemical and symptom control in patients with acromegaly. However, it is important to better understand the real-world patient population receiving lanreotide autogel treatment. METHODS In this non-interventional study the long-term treatment response to lanreotide autogel in adult patients with acromegaly from office-based centers or clinics in Germany, Austria and Switzerland was studied. Assessments included growth hormone and insulin-like growth factor-I levels, symptoms, quality of life, lanreotide plasma levels and tumor somatostatin receptor subtype expression. The primary endpoint was achievement of full biochemical control, defined as growth hormone ≤2.5 µg/L and insulin-like growth factor I normalization at month 12. RESULTS 76 patients were enrolled from 21 sites. 7/51 (13.7%) patients of the efficacy population had full biochemical control at baseline, 15/33 (45.5%) at month 12 and 10/26 (38.5%) at month 24 of treatment. At 12 months of treatment higher rates of biochemical control were observed in the following subgroups: older patients (>53 years [median]), females, treatment-naïve patients, and patients with a time since diagnosis of longer than 1.4 years (median). No clinically relevant differences in acromegaly symptoms or quality of life scores were observed. Median fasting blood glucose and glycated hemoglobin levels remained unchanged throughout the study. No new safety signals were observed. Overall tolerability of treatment with lanreotide autogel was judged by 80.8% of the enrolled patients at month 12 as 'very good' or 'good'. CONCLUSION Treatment with lanreotide autogel in a real-world setting showed long-term effectiveness and good tolerability in patients with acromegaly.
Collapse
Affiliation(s)
- Sylvère Störmann
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, München, Germany
| | - Jochen Schopohl
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, München, Germany
| | | | - Christoph Terkamp
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover
| | - Mirjam Christ-Crain
- Clinic of Endocrinology, Diabetology and Metabolism, University Hospital Basel, Basel
| | | | - Jörg Flitsch
- Department of Neurosurgery, University Hospital Hamburg-Eppendorf, Hamburg
| | - Ilonka Kreitschmann-Andermahr
- Department of Neurosurgery and Spine Surgery, University Medicine Essen, University of Duisburg-Essen, Essen, Germany
| | - Anton Luger
- Division of Endocrinology and Metabolism, Medical University of Vienna, Vienna
| | - Günter Stalla
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, München, Germany.,MEDICOVER Neuroendocrinology MVZ, Munich
| | | | | | | |
Collapse
|
22
|
Abstract
The development of peptide-based drugs, which are usually synthetic analogues of endogenous peptides, is currently one of the most topical directions in drug development. Among them, antitumor peptide-based drugs are of great interest. Anticancer peptides can be classified into three main groups based on their mechanism of action: inhibitory, necrosis-inducing and pro-apoptotic peptides. As an antitumor therapy, peptides are considered to have at least the same efficacy as chemotherapy or surgical treatment, but offer advantages in terms of safety and tolerability, given that chemotherapy is usually characterized by severe adverse effects, and surgery carries additional risks for patients. Short peptides have a number of benefits over other molecules. First, compared with full-length proteins and antibodies, short peptides are less immunogenic, more stable ex-vivo (prolonged storage at room temperature), and have better tumor or organ permeability. Moreover, the production of such short peptide-based drugs is more cost effective. Second, in comparison with small organic molecules, peptides have higher efficacy and specificity. Finally, due to the fact that the main products of peptide metabolism are amino acids, these drugs are usually characterized by lower toxicity. Short peptides have a highly selective mechanism of action, thereby demonstrating low toxicity. Furthermore, with the addition of different stabilizing structural modifications, as well as novel drug delivery systems, the peptide-based drugs are proving to be promising therapeutics for cancer mono- or polytherapy. However, challenges remain including that endogenous and synthetic peptide molecules can be oncogenic. Therefore, it is important to investigate whether peptides contribute to tumor growth. In order to answer such questions, numerous preclinical and clinical studies of peptide-based therapeutics are currently being conducted.
Collapse
|
23
|
Cozzi R, Ambrosio MR, Attanasio R, Bozzao A, De Marinis L, De Menis E, Guastamacchia E, Lania A, Lasio G, Logoluso F, Maffei P, Poggi M, Toscano V, Zini M, Chanson P, Katznelson L. Italian Association of Clinical Endocrinologists (AME) and Italian AACE Chapter Position Statement for Clinical Practice: Acromegaly - Part 2: Therapeutic Issues. Endocr Metab Immune Disord Drug Targets 2020; 20:1144-1155. [PMID: 31995025 PMCID: PMC7579256 DOI: 10.2174/1871530320666200129113328] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 12/02/2019] [Accepted: 12/02/2019] [Indexed: 12/03/2022]
Abstract
Any newly diagnosed patient should be referred to a multidisciplinary team experienced in the treatment of pituitary adenomas. The therapeutic management of acromegaly always requires a personalized strategy. Normal age-matched IGF-I values are the treatment goal. Transsphenoidal surgery by an expert neurosurgeon is the primary treatment modality for most patients, especially if there are neurological complications. In patients with poor clinical conditions or who refuse surgery, primary medical treatment should be offered, firstly with somatostatin analogs (SSAs). In patients who do not reach hormonal targets with first-generation depot SSAs, a second pharmacological option with pasireotide LAR or pegvisomant (alone or combined with SSA) should be offered. Irradiation could be proposed to patients with surgical remnants who would like to be free from long-term medical therapies or those with persistent disease activity or tumor growth despite surgery or medical therapy. Since the therapeutic tools available enable therapeutic targets to be achieved in most cases, the challenge is to focus more on the quality of life.
Collapse
Affiliation(s)
- Renato Cozzi
- Address correspondence to this author at the Endocrinologia, ASST Grande Ospedale Metropolitano Niguarda, Piazza Ospedale Maggiore 3, 20162 Milano, Italy; Tel: +39.347.5225490; E-mail:
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Fuentes-Fayos AC, García-Martínez A, Herrera-Martínez AD, Jiménez-Vacas JM, Vázquez-Borrego MC, Castaño JP, Picó A, Gahete MD, Luque RM. Molecular determinants of the response to medical treatment of growth hormone secreting pituitary neuroendocrine tumors. MINERVA ENDOCRINOL 2019; 44:109-128. [PMID: 30650942 DOI: 10.23736/s0391-1977.19.02970-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Acromegaly is a chronic systemic disease mainly caused by a growth hormone (GH)-secreting pituitary neuroendocrine tumor (PitNETs), which is associated with many health complications and increased mortality when not adequately treated. Transsphenoidal surgery is considered the treatment of choice in GH-secreting PitNETs, but patients in whom surgery cannot be considered or with persistent disease after surgery require medical therapy. Treatment with available synthetic somatostatin analogues (SSAs) is considered the mainstay in the medical management of acromegaly which exert their beneficial effects through the binding to a family of G-protein coupled receptors encoded by 5 genes (SSTR1-5). However, although it has been demonstrated that the SST1-5 receptors are physically present in tumor cells, SSAs are in many cases ineffective (i.e. approximately 10-30% of patients with GH-secreting PitNET are unresponsive to SSAs), suggesting that other cellular/molecular determinants could be essential for the response to the pharmacological treatment in patients with GH-secreting PitNETs. Therefore, the scrutiny of these determinants might be used for the identification of subgroups of patients in whom an appropriate pharmacological treatment can be successfully employed (responders vs. non-responders). In this review, we will describe some of the existing, classical and novel, genetic and molecular determinants involved in the response of patients with GH-secreting PitNETs to the available therapeutic treatments, as well as new molecular/therapeutic approaches that could be potentially useful for the treatment of GH-secreting PitNETs.
Collapse
Affiliation(s)
- Antonio C Fuentes-Fayos
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Cordoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain.,Reina Sofia University Hospital (HURS), Cordoba, Spain.,CIBER Physiopathology of Obesity and Nutrition (CIBERobn), Cordoba, Spain
| | - Araceli García-Martínez
- Research Laboratory, Hospital General Universitario de Alicante-Institute for Health and Biomedical Research (ISABIAL), Alicante, Spain
| | - Aura D Herrera-Martínez
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Cordoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain.,Reina Sofia University Hospital (HURS), Cordoba, Spain.,CIBER Physiopathology of Obesity and Nutrition (CIBERobn), Cordoba, Spain
| | - Juan M Jiménez-Vacas
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Cordoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain.,Reina Sofia University Hospital (HURS), Cordoba, Spain.,CIBER Physiopathology of Obesity and Nutrition (CIBERobn), Cordoba, Spain
| | - Mari C Vázquez-Borrego
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Cordoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain.,Reina Sofia University Hospital (HURS), Cordoba, Spain.,CIBER Physiopathology of Obesity and Nutrition (CIBERobn), Cordoba, Spain
| | - Justo P Castaño
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Cordoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain.,Reina Sofia University Hospital (HURS), Cordoba, Spain.,CIBER Physiopathology of Obesity and Nutrition (CIBERobn), Cordoba, Spain
| | - Antonio Picó
- Department of Endocrinology and Nutrition, Hospital General Universitario de Alicante-ISABIAL, Miguel Hernández University, CIBERER, Alicante, Spain
| | - Manuel D Gahete
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Cordoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain.,Reina Sofia University Hospital (HURS), Cordoba, Spain.,CIBER Physiopathology of Obesity and Nutrition (CIBERobn), Cordoba, Spain
| | - Raúl M Luque
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Cordoba, Spain - .,Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain.,Reina Sofia University Hospital (HURS), Cordoba, Spain.,CIBER Physiopathology of Obesity and Nutrition (CIBERobn), Cordoba, Spain
| |
Collapse
|
25
|
Ezzat S, Caspar-Bell GM, Chik CL, Denis MC, Domingue MÈ, Imran SA, Johnson MD, Lochnan HA, Grégoire Nyomba BL, Prebtani A, Ridout R, Ramirez JAR, Van Uum S. PREDICTIVE MARKERS FOR POSTSURGICAL MEDICAL MANAGEMENT OF ACROMEGALY: A SYSTEMATIC REVIEW AND CONSENSUS TREATMENT GUIDELINE. Endocr Pract 2019; 25:379-393. [PMID: 30657362 DOI: 10.4158/ep-2018-0500] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Objective: To clarify the selection of medical therapy following transsphenoidal surgery in patients with acromegaly, based on growth hormone (GH)/insulin-like growth factor 1 (IGF-1) response and glucometabolic control. Methods: We carried out a systematic literature review on three of the best studied and most practical predictive markers of the response to somatostatin analogues (SSAs): somatostatin receptor (SSTR) expression, tumor morphologic classification, and T2-weighted magnetic resonance imaging (MRI) signal intensity. Additional analyses focused on glucose metabolism in treated patients. Results: The literature survey confirmed significant associations of all three factors with SSA responsiveness. SSTR expression appears necessary for the SSA response; however, it is not sufficient, as approximately half of SSTR2-positive tumors failed to respond clinically to first-generation SSAs. MRI findings (T2-hypo-intensity) and a densely granulated phenotype also correlate with SSA efficacy, and are advantageous as predictive markers relative to SSTR expression alone. Glucometabolic control declines with SSA monotherapy, whereas GH receptor antagonist (GHRA) monotherapy may restore normoglycemia. Conclusion: We propose a decision tree to guide selection among SSAs, dopamine agonists (DAs), and GHRA for medical treatment of acromegaly in the postsurgical setting. This decision tree employs three validated predictive markers and other clinical considerations, to determine whether SSAs are appropriate first-line medical therapy in the postsurgical setting. DA treatment is favored in patients with modest IGF-1 elevation. GHRA treatment should be considered for patients with T2-hyperintense tumors with a sparsely granulated phenotype and/or low SSTR2 staining, and may also be favored for individuals with diabetes. Prospective analyses are required to test the utility of this therapeutic paradigm. Abbreviations: DA = dopamine agonist; DG = densely granulated; GH = growth hormone; GHRA = growth hormone receptor antagonist; HbA1c = glycated hemoglobin; IGF-1 = insulin-like growth factor-1; MRI = magnetic resonance imaging; SG = sparsely granulated; SSA = somatostatin analogue; SSTR = somatostatin receptor.
Collapse
|
26
|
Scudder CJ, Mirczuk SM, Richardson KM, Crossley VJ, Regan JTC, Gostelow R, Forcada Y, Hazuchova K, Harrington N, McGonnell IM, Church DB, Kenny PJ, Korbonits M, Fowkes RC, Niessen SJM. Pituitary Pathology and Gene Expression in Acromegalic Cats. J Endocr Soc 2019; 3:181-200. [PMID: 30620005 PMCID: PMC6316999 DOI: 10.1210/js.2018-00226] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 10/10/2018] [Indexed: 12/11/2022] Open
Abstract
The prevalence of GH-secreting pituitary tumors in domestic cats (Felis catus) is 10-fold greater than in humans. The predominant inhibitory receptors of GH-secreting pituitary tumors are somatostatin receptors (SSTRs) and D2 dopamine receptor (DRD2). The expression of these receptors is associated with the response to somatostatin analog and dopamine agonist treatment in human patients with acromegaly. The aim of this study was to describe pathological features of pituitaries from domestic cats with acromegaly, pituitary receptor expression, and investigate correlates with clinical data, including pituitary volume, time since diagnosis of diabetes, insulin requirement, and serum IGF1 concentration. Loss of reticulin structure was identified in 15 of 21 pituitaries, of which 10 of 15 exhibited acinar hyperplasia. SSTR1, SSTR2, SSTR5, and DRD2 mRNA were identified in the feline pituitary whereas SSTR3 and SSTR4 were not. Expression of SSTR1, SSTR2, and SSTR5 was greater in acromegalic cats compared with controls. A negative correlation was identified between DRD2 mRNA expression and pituitary volume. The loss of DRD2 expression should be investigated as a mechanism allowing the development of larger pituitary tumors.
Collapse
Affiliation(s)
- Christopher J Scudder
- Diabetic Remission Clinic, Department of Clinical Science and Services, Royal Veterinary College, North Mymms, United Kingdom
- Endocrine Signaling Group, Department of Comparative Biomedical Sciences, Royal Veterinary College, London, United Kingdom
| | - Samantha M Mirczuk
- Endocrine Signaling Group, Department of Comparative Biomedical Sciences, Royal Veterinary College, London, United Kingdom
| | - Karen M Richardson
- Endocrine Signaling Group, Department of Comparative Biomedical Sciences, Royal Veterinary College, London, United Kingdom
- Pathobiology and Population Sciences, Royal Veterinary College, London, United Kingdom
| | - Victoria J Crossley
- Endocrine Signaling Group, Department of Comparative Biomedical Sciences, Royal Veterinary College, London, United Kingdom
| | - Jacob T C Regan
- Endocrine Signaling Group, Department of Comparative Biomedical Sciences, Royal Veterinary College, London, United Kingdom
| | - Ruth Gostelow
- Diabetic Remission Clinic, Department of Clinical Science and Services, Royal Veterinary College, North Mymms, United Kingdom
| | - Yaiza Forcada
- Diabetic Remission Clinic, Department of Clinical Science and Services, Royal Veterinary College, North Mymms, United Kingdom
| | - Katarina Hazuchova
- Diabetic Remission Clinic, Department of Clinical Science and Services, Royal Veterinary College, North Mymms, United Kingdom
| | - Norelene Harrington
- Comparative Biomedical Sciences, Royal Veterinary College, London, United Kingdom
| | - Imelda M McGonnell
- Comparative Biomedical Sciences, Royal Veterinary College, London, United Kingdom
| | - David B Church
- Diabetic Remission Clinic, Department of Clinical Science and Services, Royal Veterinary College, North Mymms, United Kingdom
| | - Patrick J Kenny
- Diabetic Remission Clinic, Department of Clinical Science and Services, Royal Veterinary College, North Mymms, United Kingdom
- SASH Vets, Neurology and Neurosurgery, Sydney, New South Wales, Australia
| | - Márta Korbonits
- Department of Endocrinology, William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, United Kingdom
| | - Robert C Fowkes
- Endocrine Signaling Group, Department of Comparative Biomedical Sciences, Royal Veterinary College, London, United Kingdom
- Comparative Biomedical Sciences, Royal Veterinary College, London, United Kingdom
| | - Stijn J M Niessen
- Diabetic Remission Clinic, Department of Clinical Science and Services, Royal Veterinary College, North Mymms, United Kingdom
- The Diabetes Research Group, Institute of Cellular Medicine, University of Newcastle, Newcastle, Tyne and Wear, United Kingdom
| |
Collapse
|
27
|
Marazuela M, Ramos-Leví AM, Borges de Souza P, Zatelli MC. Is receptor profiling useful for predicting pituitary therapy? Eur J Endocrinol 2018; 179:D15-D25. [PMID: 30139823 DOI: 10.1530/eje-18-0549] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 08/13/2018] [Accepted: 08/22/2018] [Indexed: 12/31/2022]
Abstract
Medical treatment of pituitary tumours may present important challenges in the presence of resistance to first line therapy. In this setting, the availability of specific markers of responsiveness/resistance could be helpful to provide tailored patients' treatment. Pituitary receptor profiling has emerged as a potentially useful tool for predicting the response to specific pituitary-directed medical therapy, mainly somatostatin analogues and dopamine agonists. However, its utility is not always straightforward. In fact, agonist-receptor coupling to the consequent biological response is complex and sometimes jeopardizes the understanding of the molecular basis of pharmacological resistance. Defective expression of pituitary receptors, genetic alterations, truncated variants, impaired signal transduction or involvement of other proteins, such as cytoskeleton proteins or the Aryl hydrocarbon receptor interacting protein amongst others, have been linked to differential tumour phenotype or treatment responsiveness with conflicting results, keeping the debate on the utility of pituitary receptor profiling open. Why does this occur? How can we overcome the difficulties? Is there a true role for pituitary receptor profiling in the near future? All authors of this debate article agree on the need of prospective studies using standardized methods in order to assess the efficacy of receptor profiling as a reliable clinical predictive factor.
Collapse
Affiliation(s)
- Monica Marazuela
- Department of Endocrinology, Hospital Universitario La Princesa, Instituto de Investigación Princesa, Universidad Autónoma de Madrid, Madrid, Spain
| | - Ana M Ramos-Leví
- Department of Endocrinology, Hospital Universitario La Princesa, Instituto de Investigación Princesa, Universidad Autónoma de Madrid, Madrid, Spain
| | - Patricia Borges de Souza
- Section of Endocrinology and Internal Medicine, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Maria Chiara Zatelli
- Section of Endocrinology and Internal Medicine, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| |
Collapse
|
28
|
Bernabeu I, Cámara R, Marazuela M, Puig Domingo M. Documento de expertos sobre el manejo de la acromegalia. ENDOCRINOL DIAB NUTR 2018; 65:428-437. [DOI: 10.1016/j.endinu.2018.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 05/03/2018] [Accepted: 05/04/2018] [Indexed: 11/26/2022]
|
29
|
Günther T, Tulipano G, Dournaud P, Bousquet C, Csaba Z, Kreienkamp HJ, Lupp A, Korbonits M, Castaño JP, Wester HJ, Culler M, Melmed S, Schulz S. International Union of Basic and Clinical Pharmacology. CV. Somatostatin Receptors: Structure, Function, Ligands, and New Nomenclature. Pharmacol Rev 2018; 70:763-835. [PMID: 30232095 PMCID: PMC6148080 DOI: 10.1124/pr.117.015388] [Citation(s) in RCA: 165] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Somatostatin, also known as somatotropin-release inhibitory factor, is a cyclopeptide that exerts potent inhibitory actions on hormone secretion and neuronal excitability. Its physiologic functions are mediated by five G protein-coupled receptors (GPCRs) called somatostatin receptor (SST)1-5. These five receptors share common structural features and signaling mechanisms but differ in their cellular and subcellular localization and mode of regulation. SST2 and SST5 receptors have evolved as primary targets for pharmacological treatment of pituitary adenomas and neuroendocrine tumors. In addition, SST2 is a prototypical GPCR for the development of peptide-based radiopharmaceuticals for diagnostic and therapeutic interventions. This review article summarizes findings published in the last 25 years on the physiology, pharmacology, and clinical applications related to SSTs. We also discuss potential future developments and propose a new nomenclature.
Collapse
Affiliation(s)
- Thomas Günther
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Giovanni Tulipano
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Pascal Dournaud
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Corinne Bousquet
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Zsolt Csaba
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Hans-Jürgen Kreienkamp
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Amelie Lupp
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Márta Korbonits
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Justo P Castaño
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Hans-Jürgen Wester
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Michael Culler
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Shlomo Melmed
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Stefan Schulz
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| |
Collapse
|
30
|
Ozkaya HM, Comunoglu N, Sayitoglu M, Keskin FE, Firtina S, Khodzhaev K, Apaydin T, Gazioglu N, Tanriover N, Oz B, Kadioglu P. Germline mutations of aryl hydrocarbon receptor-interacting protein (AIP) gene and somatostatin receptor 1-5 and AIP immunostaining in patients with sporadic acromegaly with poor versus good response to somatostatin analogues. Pituitary 2018; 21:335-346. [PMID: 29455389 DOI: 10.1007/s11102-018-0876-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVE To determine aryl hydrocarbon interacting protein (AIP) gene variations and AIP and somatostatin receptor (SSTR) 1-5 immunostaining in patients with apparently sporadic acromegaly with poor versus good response to somatostatin analogues (SRLs). METHODS A total of 94 patients (66 with poor and 28 with good response to SRLs) were screened for the AIP gene variations using Sanger sequencing. Immunostaining was performed in 60 tumors. RESULTS Several variations, albeit some with undetermined significance, were detected, especially in poor responder patients. The prevalence of AIP mutation was 2.1% in the whole group and 1.5% in patients with poor response to SRLs. AIP, SSTR2A, and SSTR2B immunostainings were decreased in patients with poor response (p < 0.05 for all), and other SSTRs did not differ between the groups (p > 0.05 for all). Patients with low AIP had decreased levels of SSTR2A and SSTR3 (p < 0.05 for all). AIP and SSTR2A immunostainings were positively correlated to the treatment response and age at diagnosis was negatively correlated (p < 0.05 for all). In poor responder patients with high SSTR2A immunostaining, SSTR2B immunostaining and preoperative tumor size were positively and negatively correlated, respectively, to SRL response (p < 0.05 for all). CONCLUSIONS Lack of response to SRLs does not necessarily increase the risk of harboring AIP mutations. The finding of decreased AIP, SSTR2A, and SSTR2B immunostaining in patients with poor response to SRLs and decreased SSTR2A and SSTR3 level in those with low AIP immunostaining suggests a possible interaction between AIP and some SSTR subtypes that might alter SRL sensitivity.
Collapse
Affiliation(s)
- Hande Mefkure Ozkaya
- Department of Endocrinology and Metabolism, Cerrahpasa Medical School, Istanbul University, Cerrahpasa, 34303, Istanbul, Turkey
| | - Nil Comunoglu
- Department of Pathology, Cerrahpasa Medical School, Istanbul University, Istanbul, Turkey
| | - Muge Sayitoglu
- Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Fatma Ela Keskin
- Department of Endocrinology and Metabolism, Cerrahpasa Medical School, Istanbul University, Cerrahpasa, 34303, Istanbul, Turkey
| | - Sinem Firtina
- Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Khusan Khodzhaev
- Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Tugce Apaydin
- Department of Endocrinology and Metabolism, Cerrahpasa Medical School, Istanbul University, Cerrahpasa, 34303, Istanbul, Turkey
| | - Nurperi Gazioglu
- Department of Neurosurgery, Cerrahpasa Medical School, Istanbul University, Istanbul, Turkey
- Pituitary Center, Istanbul University, Istanbul, Turkey
| | - Necmettin Tanriover
- Department of Neurosurgery, Cerrahpasa Medical School, Istanbul University, Istanbul, Turkey
- Pituitary Center, Istanbul University, Istanbul, Turkey
| | - Buge Oz
- Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Pinar Kadioglu
- Department of Endocrinology and Metabolism, Cerrahpasa Medical School, Istanbul University, Cerrahpasa, 34303, Istanbul, Turkey.
- Pituitary Center, Istanbul University, Istanbul, Turkey.
| |
Collapse
|
31
|
Alhambra-Expósito MR, Ibáñez-Costa A, Moreno-Moreno P, Rivero-Cortés E, Vázquez-Borrego MC, Blanco-Acevedo C, Toledano-Delgado Á, Lombardo-Galera MS, Vallejo-Casas JA, Gahete MD, Castaño JP, Gálvez MA, Luque RM. Association between radiological parameters and clinical and molecular characteristics in human somatotropinomas. Sci Rep 2018; 8:6173. [PMID: 29670116 PMCID: PMC5906631 DOI: 10.1038/s41598-018-24260-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 03/09/2018] [Indexed: 02/08/2023] Open
Abstract
Acromegaly is a rare but severe disease, originated in 95% of cases by a growth hormone-secreting adenoma (somatotropinoma) in the pituitary. Magnetic resonance imaging (MRI) is a non-invasive technique used for the diagnosis and prognosis of pituitary tumours. The aim of this study was to determine whether the use of T2-weighted signal intensity at MRI could help to improve the characterisation of somatotropinomas, by analysing its relationship with clinical/molecular features. An observational study was implemented in a cohort of 22 patients (mean age = 42.1 ± 17.2 years; 59% women; 95% size>10 mm). Suprasellar-extended somatotropinomas presented larger diameters vs. non-extended tumours. T2-imaging revealed that 59% of tumours were hyperintense and 41% isointense adenomas, wherein hyperintense were more invasive (according to Knosp-score) than isointense adenomas. A higher proportion of hyperintense somatotropinomas presented extrasellar-growth, suprasellar-growth and invasion of the cavernous sinus compared to isointense adenomas. Interestingly, somatostatin receptor-3 and dopamine receptor-5 (DRD5) expression levels were associated with extrasellar and/or suprasellar extension. Additionally, DRD5 was also higher in hyperintense adenomas and its expression was directly correlated with Knosp-score and with tumour diameter. Hence, T2-weighted MRI on somatotropinomas represents a potential tool to refine their diagnosis and prognosis, and could support the election of preoperative treatment, when required.
Collapse
Affiliation(s)
- María R Alhambra-Expósito
- Maimonides Institute of Biomedical Research of Cordoba, Córdoba, 14004, Spain.,Reina Sofia University Hospital (HURS), Córdoba, 14004, Spain.,Service of Endocrinology and Nutrition, HURS, Córdoba, 14004, Spain
| | - Alejandro Ibáñez-Costa
- Maimonides Institute of Biomedical Research of Cordoba, Córdoba, 14004, Spain.,Reina Sofia University Hospital (HURS), Córdoba, 14004, Spain.,Department of Cell Biology, Physiology and Immunology, Universidad de Córdoba, Córdoba, 14004, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, 14004, Spain.,Campus de Excelencia Internacional Agroalimentario (ceiA3), Córdoba, 14004, Spain
| | - Paloma Moreno-Moreno
- Maimonides Institute of Biomedical Research of Cordoba, Córdoba, 14004, Spain.,Reina Sofia University Hospital (HURS), Córdoba, 14004, Spain.,Service of Endocrinology and Nutrition, HURS, Córdoba, 14004, Spain
| | - Esther Rivero-Cortés
- Maimonides Institute of Biomedical Research of Cordoba, Córdoba, 14004, Spain.,Reina Sofia University Hospital (HURS), Córdoba, 14004, Spain.,Department of Cell Biology, Physiology and Immunology, Universidad de Córdoba, Córdoba, 14004, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, 14004, Spain.,Campus de Excelencia Internacional Agroalimentario (ceiA3), Córdoba, 14004, Spain
| | - Mari C Vázquez-Borrego
- Maimonides Institute of Biomedical Research of Cordoba, Córdoba, 14004, Spain.,Reina Sofia University Hospital (HURS), Córdoba, 14004, Spain.,Department of Cell Biology, Physiology and Immunology, Universidad de Córdoba, Córdoba, 14004, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, 14004, Spain.,Campus de Excelencia Internacional Agroalimentario (ceiA3), Córdoba, 14004, Spain
| | - Cristóbal Blanco-Acevedo
- Maimonides Institute of Biomedical Research of Cordoba, Córdoba, 14004, Spain.,Reina Sofia University Hospital (HURS), Córdoba, 14004, Spain.,Service of Neurosurgery, HURS, Córdoba, 14004, Spain
| | - Álvaro Toledano-Delgado
- Maimonides Institute of Biomedical Research of Cordoba, Córdoba, 14004, Spain.,Reina Sofia University Hospital (HURS), Córdoba, 14004, Spain.,Service of Neurosurgery, HURS, Córdoba, 14004, Spain
| | | | | | - Manuel D Gahete
- Maimonides Institute of Biomedical Research of Cordoba, Córdoba, 14004, Spain.,Reina Sofia University Hospital (HURS), Córdoba, 14004, Spain.,Department of Cell Biology, Physiology and Immunology, Universidad de Córdoba, Córdoba, 14004, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, 14004, Spain.,Campus de Excelencia Internacional Agroalimentario (ceiA3), Córdoba, 14004, Spain
| | - Justo P Castaño
- Maimonides Institute of Biomedical Research of Cordoba, Córdoba, 14004, Spain. .,Reina Sofia University Hospital (HURS), Córdoba, 14004, Spain. .,Department of Cell Biology, Physiology and Immunology, Universidad de Córdoba, Córdoba, 14004, Spain. .,CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, 14004, Spain. .,Campus de Excelencia Internacional Agroalimentario (ceiA3), Córdoba, 14004, Spain.
| | - María A Gálvez
- Maimonides Institute of Biomedical Research of Cordoba, Córdoba, 14004, Spain. .,Reina Sofia University Hospital (HURS), Córdoba, 14004, Spain. .,Service of Endocrinology and Nutrition, HURS, Córdoba, 14004, Spain.
| | - Raúl M Luque
- Maimonides Institute of Biomedical Research of Cordoba, Córdoba, 14004, Spain. .,Reina Sofia University Hospital (HURS), Córdoba, 14004, Spain. .,Department of Cell Biology, Physiology and Immunology, Universidad de Córdoba, Córdoba, 14004, Spain. .,CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, 14004, Spain. .,Campus de Excelencia Internacional Agroalimentario (ceiA3), Córdoba, 14004, Spain.
| |
Collapse
|
32
|
Venegas-Moreno E, Vazquez-Borrego MC, Dios E, Gros-Herguido N, Flores-Martinez A, Rivero-Cortés E, Madrazo-Atutxa A, Japón MA, Luque RM, Castaño JP, Cano DA, Soto-Moreno A. Association between dopamine and somatostatin receptor expression and pharmacological response to somatostatin analogues in acromegaly. J Cell Mol Med 2017; 22:1640-1649. [PMID: 29266696 PMCID: PMC5824369 DOI: 10.1111/jcmm.13440] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 09/27/2017] [Indexed: 12/14/2022] Open
Abstract
Acromegaly is a hormonal disorder resulting from excessive growth hormone (GH) secretion frequently produced by pituitary adenomas and consequent increase in insulin‐like growth factor 1 (IGF‐I). Elevated GH and IGF‐I levels result in a wide range of somatic, cardiovascular, endocrine, metabolic and gastrointestinal morbidities. Somatostatin analogues (SSAs) form the basis of medical therapy for acromegaly and are currently used as first‐line treatment or as second‐line therapy in patients undergoing unsuccessful surgery. However, a considerable percentage of patients do not respond to SSAs treatment. Somatostatin receptors (SSTR1‐5) and dopamine receptors (DRD1‐5) subtypes play critical roles in the regulation of hormone secretion. These receptors are considered important pharmacological targets to inhibit hormone oversecretion. It has been proposed that decreased expression of SSTRs may be associated with poor response to SSAs. Here, we systematically examine SSTRs and DRDs expression in human somatotroph adenomas by quantitative PCR. We observed an association between the response to SSAs treatment and DRD4, DRD5, SSTR1 and SSTR2 expression. We also examined SSTR expression by immunohistochemistry and found that the immunohistochemical detection of SSTR2 in particular might be a good predictor of response to SSAs.
Collapse
Affiliation(s)
- Eva Venegas-Moreno
- Unidad de Gestión de Endocrinología y Nutrición, Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Mari C Vazquez-Borrego
- Department of Cell Biology, Physiology and Immunology, Maimonides Institute For Biomedical Research of Cordoba (IMIBIC), Reina Sofia University Hospital (HURS), CIBER Physiopathology of Obesity and Nutrition (CIBERobn), Agrifood Campus of International Excellence (ceiA3), University of Cordoba, Cordoba, Spain
| | - Elena Dios
- Unidad de Gestión de Endocrinología y Nutrición, Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Noelia Gros-Herguido
- Unidad de Gestión de Endocrinología y Nutrición, Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Alvaro Flores-Martinez
- Unidad de Gestión de Endocrinología y Nutrición, Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Esther Rivero-Cortés
- Department of Cell Biology, Physiology and Immunology, Maimonides Institute For Biomedical Research of Cordoba (IMIBIC), Reina Sofia University Hospital (HURS), CIBER Physiopathology of Obesity and Nutrition (CIBERobn), Agrifood Campus of International Excellence (ceiA3), University of Cordoba, Cordoba, Spain
| | - Ainara Madrazo-Atutxa
- Unidad de Gestión de Endocrinología y Nutrición, Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Miguel A Japón
- Department of Pathology, Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Raúl M Luque
- Department of Cell Biology, Physiology and Immunology, Maimonides Institute For Biomedical Research of Cordoba (IMIBIC), Reina Sofia University Hospital (HURS), CIBER Physiopathology of Obesity and Nutrition (CIBERobn), Agrifood Campus of International Excellence (ceiA3), University of Cordoba, Cordoba, Spain
| | - Justo P Castaño
- Department of Cell Biology, Physiology and Immunology, Maimonides Institute For Biomedical Research of Cordoba (IMIBIC), Reina Sofia University Hospital (HURS), CIBER Physiopathology of Obesity and Nutrition (CIBERobn), Agrifood Campus of International Excellence (ceiA3), University of Cordoba, Cordoba, Spain
| | - David A Cano
- Unidad de Gestión de Endocrinología y Nutrición, Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Alfonso Soto-Moreno
- Unidad de Gestión de Endocrinología y Nutrición, Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| |
Collapse
|
33
|
Sizdahkhani S, Feldman MJ, Piazza MG, Ksendzovsky A, Edwards NA, Ray-Chaudhury A, Maric D, Merrill MJ, Pacak K, Zhuang Z, Chittiboina P. Somatostatin receptor expression on von Hippel-Lindau-associated hemangioblastomas offers novel therapeutic target. Sci Rep 2017; 7:40822. [PMID: 28094316 PMCID: PMC5240113 DOI: 10.1038/srep40822] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 12/12/2016] [Indexed: 01/03/2023] Open
Abstract
Von Hippel-Lindau (VHL)-associated hemangioblastomas (VHL-HB) arise in the central nervous system (CNS), and are a leading cause of morbidity and mortality in VHL disease. Currently, surgical resection is the most effective way to manage symptomatic VHL-HBs. Surgically unresectable VHL-HBs or those in frail patients are challenging problems. Therapies targeting oncologic and vascular endothelial growth factor (VEGF) pathways have failed to demonstrate tumor control. Our experience and previous reports on VHL-HB avidity to somatostatin analogues suggested somatostatin receptor (SSTR) expression in VHL-HBs, offering an alternative therapeutic strategy. We explored this possibility by demonstrating consistent histologic expression of SSTR1, 2a, 4, and 5 in VHL-HBs. We found that somatostatin analogue octreotide induces apoptosis in VHL-HB stromal cells in a dose-dependent fashion by BAX – caspase-3 pathway unrelated to canonical VHL pathway. When administered to a patient with unresectable symptomatic suprasellar hemangioblastoma, octreotide resulted in tumor volume reduction, symptom stabilization, and tumor cytopenia on repeat 68Ga-DOTA-TATE positron emission tomography (PET) within 6 months, suggesting tumor infarction. We conclude that VHL-HBs harbor multiple SSTR subtypes that offer actionable chemo-therapeutic strategy for management of symptomatic, unresectable tumors by somatostatin analogue therapy.
Collapse
Affiliation(s)
- Saman Sizdahkhani
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Michael J Feldman
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Martin G Piazza
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Alexander Ksendzovsky
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA.,Department of Neurosurgery, University of Virginia Health System, Charlottesville, Virginia USA
| | - Nancy A Edwards
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Abhik Ray-Chaudhury
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Dragan Maric
- Flow Cytometry Core, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Marsha J Merrill
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Karel Pacak
- Section on Medical Neuroendocrinology, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Zhengping Zhuang
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Prashant Chittiboina
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
34
|
Franck SE, Gatto F, van der Lely AJ, Janssen JA, Dallenga AH, Nagtegaal AP, Hofland LJ, Neggers SJ. Somatostatin Receptor Expression in GH-Secreting Pituitary Adenomas Treated with Long-Acting Somatostatin Analogues in Combination with Pegvisomant. Neuroendocrinology 2017; 105:44-53. [PMID: 27455094 PMCID: PMC5475231 DOI: 10.1159/000448429] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 07/17/2016] [Indexed: 11/19/2022]
Abstract
BACKGROUND Growth hormone-secreting pituitary adenomas (somatotroph adenoma) predominantly express somatostatin receptors (SSTRs) subtypes 2 and 5. Higher SSTR2 expression on somatotroph adenomas results in a better response to somatostatin analogues (SSAs), which preferentially bind, but also downregulate, SSTR2. The effect of the combined treatment with SSAs and the GH receptor antagonist pegvisomant (PEGV) on SSTR expression in somatotroph adenomas is currently unknown. AIM OF THE STUDY To assess SSTR2 and SSTR5 expression in three groups of somatotroph adenomas: drug-naive, treated with long-acting (LA) SSA monotherapy, or LA-SSA/PEGV combination therapy before surgery. Additionally, we evaluated the required PEGV dose to achieve insulin-like growth factor I (IGF-I) normalization in relation to the SSTR expression. MATERIALS AND METHODS At our Pituitary Center Rotterdam, we selected acromegalic patients who underwent transsphenoidal neurosurgery. All patients were eventually treated with LA-SSA/PEGV combination therapy during their medical history. SSTR2 and SSTR5 expression in somatotroph adenoma tissues was determined using immunohistochemistry. RESULTS Out of 39 somatotroph adenoma tissue samples, 23 were drug-naive, 9 received pretreatment with LA-SSA and 7 LA-SSA/PEGV combined treatment. SSTR2 expression was significantly higher in treatment-naive compared to combined treatment somatotroph adenomas (p = 0.048), while SSTR5 expression did not differ. Noteworthy, SSTR2 expression in naive somatotroph adenoma tissues was inversely correlated with the required PEGV dose to achieve IGF-I normalization during postsurgical medical treatment (ρ = -0.538, p = 0.024). CONCLUSION In our specific cohort, the SSTR2 expression was lower in patients pretreated with LA-SSA/PEGV compared to the drug-naive acromegalic patients. Additionally, the SSTR2 expression in treatment-naive somatotroph adenoma tissues was inversely correlated with the required PEGV dose to achieve IGF-I normalization.
Collapse
Affiliation(s)
- Sanne E. Franck
- Endocrinology Section, Department of Internal Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
- *S.E. Franck, MD, Endocrinology Section, Department of Internal Medicine, Erasmus University Medical Center, PO Box 2040, NL-3000 CA Rotterdam (The Netherlands), E-Mail
| | - Federico Gatto
- Endocrinology Section, Department of Internal Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Aart Jan van der Lely
- Endocrinology Section, Department of Internal Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Joseph A.M.J.L. Janssen
- Endocrinology Section, Department of Internal Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Alof H.G. Dallenga
- Department of Neurosurgery, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - A. Paul Nagtegaal
- Department of Otorhinolaryngology, Pituitary Center Rotterdam, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Leo J. Hofland
- Endocrinology Section, Department of Internal Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Sebastian J.C.M.M. Neggers
- Endocrinology Section, Department of Internal Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
35
|
Mooney MA, Simon ED, Little AS. Advancing Treatment of Pituitary Adenomas through Targeted Molecular Therapies: The Acromegaly and Cushing Disease Paradigms. Front Surg 2016; 3:45. [PMID: 27517036 PMCID: PMC4963385 DOI: 10.3389/fsurg.2016.00045] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 07/12/2016] [Indexed: 11/14/2022] Open
Abstract
The current treatment of pituitary adenomas requires a balance of conservative management, surgical resection, and in select tumor types, molecular therapy. Acromegaly treatment is an evolving field where our understanding of molecular targets and drug therapies has improved treatment options for patients with excess growth hormone levels. We highlight the use of molecular therapies in this disease process and advances in this field, which may represent a paradigm shift for the future of pituitary adenoma treatment.
Collapse
Affiliation(s)
- Michael A Mooney
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center , Phoenix, AZ , USA
| | - Elias D Simon
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center , Phoenix, AZ , USA
| | - Andrew S Little
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center , Phoenix, AZ , USA
| |
Collapse
|
36
|
Bronstein MD, Jallad RS. Pasireotide for treating acromegaly. Expert Opin Orphan Drugs 2016. [DOI: 10.1517/21678707.2016.1167593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
37
|
Abstract
INTRODUCTION In nearly all cases, acromegaly is caused by excess GH from a pituitary adenoma, resulting in elevated circulating levels of GH and, subsequently, IGF-1. Treatment goals are to eliminate morbidity and restore the increased mortality to normal rates. Therapeutic strategies aim to minimize tumor mass and normalize GH and IGF-1 levels. Somatostatin analogues are the medical treatment of choice in acromegaly, as first-line or post-surgical therapy, and have proven efficacy in pituitary tumor volume reduction (TVR). METHODS Here we review the effects of somatostatin analogue therapy on pituitary tumor volume in patients with acromegaly. RESULTS TVR with somatostatin analogues may be mediated by direct anti-proliferative effects via activation of somatostatin receptors, or by indirect effects, such as angiogenesis inhibition, and is more pronounced when they are administered as first-line therapy. Various studies of first-line treatment with octreotide LAR have shown significant TVR in ≥73% of patients. First-line treatment with lanreotide Autogel has shown evidence of TVR, although more studies are needed. In a recent randomized, double-blind, 12-month trial in 358 medical-treatment-naïve acromegaly patients, significant TVR was achieved by 81% of patients administered pasireotide LAR and 77% administered octreotide LAR. Pre-operative somatostatin analogue therapy may also induce TVR and improve post-operative disease control compared with surgery alone. TVR is progressive with prolonged treatment, and decreased IGF-1 levels may be its best predictor, followed by age and degree of GH decrease. However, TVR does not always correlate with degree of biochemical control. CONCLUSION Somatostatin analogues (first- or second-line treatment) are the mainstay of medical therapy and, as first-line medical therapy, are associated with significant pituitary TVR in most patients.
Collapse
Affiliation(s)
- Annamaria Colao
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, via S Pansini 5, 80131, Naples, Italy.
| | - Renata S Auriemma
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, via S Pansini 5, 80131, Naples, Italy
| | - Rosario Pivonello
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, via S Pansini 5, 80131, Naples, Italy
| |
Collapse
|
38
|
Ramos-Leví AM, Bernabeu I, Sampedro-Núñez M, Marazuela M. Genetic Predictors of Response to Different Medical Therapies in Acromegaly. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 138:85-114. [PMID: 26940388 DOI: 10.1016/bs.pmbts.2015.10.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
In the era of predictive medicine, management of diseases is evolving into a more personal and individualized approach, as more data are available regarding clinical, biochemical, radiological, molecular, histopathological, and genetic aspects. In the particular setting of acromegaly, which is a rare, chronic, debilitating, and disfiguring disease, an optimized approach deems even more necessary, especially because of an associated increased morbidity and mortality, the impact on patients' quality of life, and the increased cost of frequently necessary life-long treatments. In this paper, we review the available studies that address potential genetic influences on acromegaly, their role in the outcome, and response to treatments, as well as their contribution to the risk of developing side effects. We focus mainly on pharmacogenetic factors involved during treatment with dopamine agonists, somatostatin analogs, and pegvisomant. Specifically, mutations in dopamine receptors, somatostatin receptors, growth hormone receptors, and metabolic pathways involved in growth hormone action; polymorphisms in the insulin-like growth factor and the insulin-like growth factor binding proteins; and polymorphisms in other genes that may determine differences in the frequency of developing adverse events.
Collapse
Affiliation(s)
- Ana M Ramos-Leví
- Department of Endocrinology and Nutrition, Hospital Universitario la Princesa, Instituto de Investigación Princesa, Universidad Autónoma de Madrid, Madrid, Spain
| | - Ignacio Bernabeu
- Department of Endocrinology and Nutrition, Complejo Hospitalario Universitario de Santiago de Compostela, Servicio Gallego de Salud (SERGAS); Universidad de Santiago de Compostela, La Coruña, Spain
| | - Miguel Sampedro-Núñez
- Department of Endocrinology and Nutrition, Hospital Universitario la Princesa, Instituto de Investigación Princesa, Universidad Autónoma de Madrid, Madrid, Spain
| | - Mónica Marazuela
- Department of Endocrinology and Nutrition, Hospital Universitario la Princesa, Instituto de Investigación Princesa, Universidad Autónoma de Madrid, Madrid, Spain.
| |
Collapse
|
39
|
Iacovazzo D, Carlsen E, Lugli F, Chiloiro S, Piacentini S, Bianchi A, Giampietro A, Mormando M, Clear AJ, Doglietto F, Anile C, Maira G, Lauriola L, Rindi G, Roncaroli F, Pontecorvi A, Korbonits M, De Marinis L. Factors predicting pasireotide responsiveness in somatotroph pituitary adenomas resistant to first-generation somatostatin analogues: an immunohistochemical study. Eur J Endocrinol 2016; 174:241-50. [PMID: 26586796 DOI: 10.1530/eje-15-0832] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Accepted: 11/19/2015] [Indexed: 01/02/2023]
Abstract
AIM To gather data regarding factors predicting responsiveness to pasireotide in acromegaly. PATIENTS AND METHODS SSTR2a, SSTR3, SSTR5, AIP, Ki-67 and the adenoma subtype were evaluated in somatotroph adenomas from 39 patients treated post-operatively with somatostatin analogues (SSAs). A standardized SSTR scoring system was applied (scores 0-3). All patients received first-generation SSAs, and 11 resistant patients were subsequently treated with pasireotide LAR. RESULTS None of the patients with negative or cytoplasmic-only SSTR2a expression (scores 0-1) were responsive to first-generation SSAs, as opposed to 20% (score 2) and 50% of patients with a score of 3 (P=0.04). None of the patients with an SSTR5 score of 0-1 were responsive to pasireotide, as opposed to 5/7 cases with a score of 2 or 3 (P=0.02). SSTR3 expression did not influence first-generation SSAs or pasireotide responsiveness. Tumours with low AIP were resistant to first-generation SSAs (100 vs 60%; P=0.02), while they had similar responsiveness to pasireotide compared to tumours with conserved AIP expression (50 vs 40%; P=0.74). Tumours with low AIP displayed reduced SSTR2 (SSTR2a scores 0-1 44.4 vs 6.7%; P=0.006) while no difference was seen in SSTR5 (SSTR5 scores 0-1 33.3 vs 23.3%; P=0.55). Sparsely granulated adenomas responded better to pasireotide compared to densely granulated ones (80 vs 16.7%; P=0.04). CONCLUSION The expression of SSTR5 might predict responsiveness to pasireotide in acromegaly. AIP deficient and sparsely granulated adenomas may benefit from pasireotide treatment. These results need to be confirmed in larger series of pasireotide-treated patients.
Collapse
Affiliation(s)
- Donato Iacovazzo
- EndocrinologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKPathologySTHF, N-3710 Skien, NorwayEndocrinologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyHaemato-OncologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKNeurosurgeryUniversità di Brescia, 25121 Brescia, ItalyNeurosurgeryUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyNeurosurgeryHumanitas, 20089 Milan, ItalyPathologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyBrainBehaviour and Mental Health, University of Manchester, M13 9PT Manchester, UK EndocrinologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKPathologySTHF, N-3710 Skien, NorwayEndocrinologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyHaemato-OncologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKNeurosurgeryUniversità di Brescia, 25121 Brescia, ItalyNeurosurgeryUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyNeurosurgeryHumanitas, 20089 Milan, ItalyPathologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyBrainBehaviour and Mental Health, University of Manchester, M13 9PT Manchester, UK
| | - Eivind Carlsen
- EndocrinologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKPathologySTHF, N-3710 Skien, NorwayEndocrinologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyHaemato-OncologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKNeurosurgeryUniversità di Brescia, 25121 Brescia, ItalyNeurosurgeryUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyNeurosurgeryHumanitas, 20089 Milan, ItalyPathologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyBrainBehaviour and Mental Health, University of Manchester, M13 9PT Manchester, UK
| | - Francesca Lugli
- EndocrinologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKPathologySTHF, N-3710 Skien, NorwayEndocrinologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyHaemato-OncologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKNeurosurgeryUniversità di Brescia, 25121 Brescia, ItalyNeurosurgeryUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyNeurosurgeryHumanitas, 20089 Milan, ItalyPathologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyBrainBehaviour and Mental Health, University of Manchester, M13 9PT Manchester, UK
| | - Sabrina Chiloiro
- EndocrinologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKPathologySTHF, N-3710 Skien, NorwayEndocrinologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyHaemato-OncologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKNeurosurgeryUniversità di Brescia, 25121 Brescia, ItalyNeurosurgeryUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyNeurosurgeryHumanitas, 20089 Milan, ItalyPathologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyBrainBehaviour and Mental Health, University of Manchester, M13 9PT Manchester, UK
| | - Serena Piacentini
- EndocrinologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKPathologySTHF, N-3710 Skien, NorwayEndocrinologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyHaemato-OncologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKNeurosurgeryUniversità di Brescia, 25121 Brescia, ItalyNeurosurgeryUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyNeurosurgeryHumanitas, 20089 Milan, ItalyPathologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyBrainBehaviour and Mental Health, University of Manchester, M13 9PT Manchester, UK
| | - Antonio Bianchi
- EndocrinologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKPathologySTHF, N-3710 Skien, NorwayEndocrinologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyHaemato-OncologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKNeurosurgeryUniversità di Brescia, 25121 Brescia, ItalyNeurosurgeryUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyNeurosurgeryHumanitas, 20089 Milan, ItalyPathologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyBrainBehaviour and Mental Health, University of Manchester, M13 9PT Manchester, UK
| | - Antonella Giampietro
- EndocrinologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKPathologySTHF, N-3710 Skien, NorwayEndocrinologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyHaemato-OncologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKNeurosurgeryUniversità di Brescia, 25121 Brescia, ItalyNeurosurgeryUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyNeurosurgeryHumanitas, 20089 Milan, ItalyPathologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyBrainBehaviour and Mental Health, University of Manchester, M13 9PT Manchester, UK
| | - Marilda Mormando
- EndocrinologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKPathologySTHF, N-3710 Skien, NorwayEndocrinologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyHaemato-OncologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKNeurosurgeryUniversità di Brescia, 25121 Brescia, ItalyNeurosurgeryUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyNeurosurgeryHumanitas, 20089 Milan, ItalyPathologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyBrainBehaviour and Mental Health, University of Manchester, M13 9PT Manchester, UK
| | - Andrew J Clear
- EndocrinologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKPathologySTHF, N-3710 Skien, NorwayEndocrinologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyHaemato-OncologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKNeurosurgeryUniversità di Brescia, 25121 Brescia, ItalyNeurosurgeryUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyNeurosurgeryHumanitas, 20089 Milan, ItalyPathologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyBrainBehaviour and Mental Health, University of Manchester, M13 9PT Manchester, UK
| | - Francesco Doglietto
- EndocrinologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKPathologySTHF, N-3710 Skien, NorwayEndocrinologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyHaemato-OncologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKNeurosurgeryUniversità di Brescia, 25121 Brescia, ItalyNeurosurgeryUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyNeurosurgeryHumanitas, 20089 Milan, ItalyPathologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyBrainBehaviour and Mental Health, University of Manchester, M13 9PT Manchester, UK
| | - Carmelo Anile
- EndocrinologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKPathologySTHF, N-3710 Skien, NorwayEndocrinologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyHaemato-OncologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKNeurosurgeryUniversità di Brescia, 25121 Brescia, ItalyNeurosurgeryUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyNeurosurgeryHumanitas, 20089 Milan, ItalyPathologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyBrainBehaviour and Mental Health, University of Manchester, M13 9PT Manchester, UK
| | - Giulio Maira
- EndocrinologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKPathologySTHF, N-3710 Skien, NorwayEndocrinologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyHaemato-OncologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKNeurosurgeryUniversità di Brescia, 25121 Brescia, ItalyNeurosurgeryUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyNeurosurgeryHumanitas, 20089 Milan, ItalyPathologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyBrainBehaviour and Mental Health, University of Manchester, M13 9PT Manchester, UK
| | - Libero Lauriola
- EndocrinologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKPathologySTHF, N-3710 Skien, NorwayEndocrinologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyHaemato-OncologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKNeurosurgeryUniversità di Brescia, 25121 Brescia, ItalyNeurosurgeryUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyNeurosurgeryHumanitas, 20089 Milan, ItalyPathologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyBrainBehaviour and Mental Health, University of Manchester, M13 9PT Manchester, UK
| | - Guido Rindi
- EndocrinologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKPathologySTHF, N-3710 Skien, NorwayEndocrinologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyHaemato-OncologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKNeurosurgeryUniversità di Brescia, 25121 Brescia, ItalyNeurosurgeryUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyNeurosurgeryHumanitas, 20089 Milan, ItalyPathologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyBrainBehaviour and Mental Health, University of Manchester, M13 9PT Manchester, UK
| | - Federico Roncaroli
- EndocrinologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKPathologySTHF, N-3710 Skien, NorwayEndocrinologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyHaemato-OncologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKNeurosurgeryUniversità di Brescia, 25121 Brescia, ItalyNeurosurgeryUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyNeurosurgeryHumanitas, 20089 Milan, ItalyPathologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyBrainBehaviour and Mental Health, University of Manchester, M13 9PT Manchester, UK
| | - Alfredo Pontecorvi
- EndocrinologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKPathologySTHF, N-3710 Skien, NorwayEndocrinologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyHaemato-OncologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKNeurosurgeryUniversità di Brescia, 25121 Brescia, ItalyNeurosurgeryUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyNeurosurgeryHumanitas, 20089 Milan, ItalyPathologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyBrainBehaviour and Mental Health, University of Manchester, M13 9PT Manchester, UK
| | - Márta Korbonits
- EndocrinologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKPathologySTHF, N-3710 Skien, NorwayEndocrinologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyHaemato-OncologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKNeurosurgeryUniversità di Brescia, 25121 Brescia, ItalyNeurosurgeryUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyNeurosurgeryHumanitas, 20089 Milan, ItalyPathologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyBrainBehaviour and Mental Health, University of Manchester, M13 9PT Manchester, UK
| | - Laura De Marinis
- EndocrinologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKPathologySTHF, N-3710 Skien, NorwayEndocrinologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyHaemato-OncologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKNeurosurgeryUniversità di Brescia, 25121 Brescia, ItalyNeurosurgeryUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyNeurosurgeryHumanitas, 20089 Milan, ItalyPathologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyBrainBehaviour and Mental Health, University of Manchester, M13 9PT Manchester, UK
| |
Collapse
|
40
|
Kiseljak-Vassiliades K, Xu M, Mills TS, Smith EE, Silveira LJ, Lillehei KO, Kerr JM, Kleinschmidt-DeMasters BK, Wierman ME. Differential somatostatin receptor (SSTR) 1-5 expression and downstream effectors in histologic subtypes of growth hormone pituitary tumors. Mol Cell Endocrinol 2015; 417:73-83. [PMID: 26391562 PMCID: PMC4641524 DOI: 10.1016/j.mce.2015.09.016] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 09/11/2015] [Accepted: 09/17/2015] [Indexed: 12/27/2022]
Abstract
PURPOSE The aim of this study was to examine whether differential expression of somatostatin receptors (SSTR) 1-5 and downstream effectors are different in densely (DG) and sparsely (SG) granulated histological growth hormone (GH) pituitary tumor subtypes. METHODS The study included 33 acromegalic patients with 23 DG and 10 SG tumors. SSTR1-5 were measured by qPCR and immunoblotting. Signaling candidates downstream of SSTR2 were also assessed. RESULTS SSTR2 mRNA and protein levels were significantly higher in DG compared to SG tumors. Downstream of SSTR2, p27(kip1) was decreased (2.6-fold) in SG compared to DG tumors, suggesting a potential mechanism of SSA resistance in SG tumors with intact SSTR2 expression. Re-expression of E-cadherin in GH pituitary cell increased p27(kip1) levels. CONCLUSIONS Histological subtyping correlated with SSTR2, E cadherin and p27(kip) protein levels and these may serve as useful biomarkers in GH tumors to predict behavior and response to therapy with SSA.
Collapse
Affiliation(s)
- Katja Kiseljak-Vassiliades
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Research Service Veterans Affairs Medical Center, Denver, CO 80220, USA.
| | - Mei Xu
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Taylor S Mills
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Elizabeth E Smith
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Lori J Silveira
- Division of Biostatistics and Bioinformatics, National Jewish Health, Denver, CO 80206, USA
| | - Kevin O Lillehei
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Janice M Kerr
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - B K Kleinschmidt-DeMasters
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Margaret E Wierman
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Research Service Veterans Affairs Medical Center, Denver, CO 80220, USA
| |
Collapse
|
41
|
Fan X, Mao Z, He D, Liao C, Jiang X, Lei N, Hu B, Wang X, Li Z, Lin Y, Gou X, Zhu Y, Wang H. Expression of somatostatin receptor subtype 2 in growth hormone-secreting pituitary adenoma and the regulation of miR-185. J Endocrinol Invest 2015; 38:1117-28. [PMID: 26036598 DOI: 10.1007/s40618-015-0306-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2014] [Accepted: 04/30/2015] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Long-acting somatostatin analogs (SSAs) are most widely used to treat growth hormone (GH)-secreting pituitary adenoma. However, approximately 30 % of treated patients show resistance to SSAs, which may be associated with the reduction of somatostatin receptor subtype 2 (SSTR2) mRNA and protein expression. MATERIALS AND METHODS The present study used immunohistochemistry to detect the expression of SSTR2 and SSTR5 in twenty human GH-secreting adenoma samples treated with SSAs and seven normal pituitary samples. RESULTS The staining intensities of SSTR2 and SSTR5 were stronger in most adenoma samples than in normal pituitary. The expression of SSTR2 tended to be lower in the SSA non-responder group than in responders. A search of the Bioinformatics data bank and the miRCURY™ LNA array confirmed miR-185 as the putative mircoRNA (miRNA) regulating the expression of SSTR2. An in vitro study using Dual Luciferase reporter assay demonstrated that miR-185 likely targets the 3'-UTR of SSTR2 mRNA in the rat pituitary adenoma GH3 cell line. MiR-185 also downregulated or upregulated the expression of SSTR2 mRNA and SSTR2 protein, following transfection with miR-185 mimics or inhibitors, respectively. CONCLUSION MiR-185 enhanced the cell proliferation and inhibited the apoptosis of GH3 cells.
Collapse
Affiliation(s)
- X Fan
- Department of Neurosurgery and Pituitary Tumor Center, Zhongshan School of Medicine, The First Affiliated Hospital of Sun Yat-sen University, 52# Zhongshan Er Road, Guangzhou, 510080, China
- Department of Neurosurgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, China
| | - Z Mao
- Department of Neurosurgery and Pituitary Tumor Center, Zhongshan School of Medicine, The First Affiliated Hospital of Sun Yat-sen University, 52# Zhongshan Er Road, Guangzhou, 510080, China
| | - D He
- Department of Neurosurgery and Pituitary Tumor Center, Zhongshan School of Medicine, The First Affiliated Hospital of Sun Yat-sen University, 52# Zhongshan Er Road, Guangzhou, 510080, China
| | - C Liao
- Department of Neurosurgery and Pituitary Tumor Center, Zhongshan School of Medicine, The First Affiliated Hospital of Sun Yat-sen University, 52# Zhongshan Er Road, Guangzhou, 510080, China
| | - X Jiang
- Department of Neurosurgery and Pituitary Tumor Center, Zhongshan School of Medicine, The First Affiliated Hospital of Sun Yat-sen University, 52# Zhongshan Er Road, Guangzhou, 510080, China
| | - N Lei
- Department of Histology and Embryology, Medical school of Sun Yat-sen University, 74# Zhongshan Er Road, Guangzhou, 510080, China
| | - B Hu
- Department of Neurosurgery and Pituitary Tumor Center, Zhongshan School of Medicine, The First Affiliated Hospital of Sun Yat-sen University, 52# Zhongshan Er Road, Guangzhou, 510080, China
| | - X Wang
- Department of Histology and Embryology, Medical school of Sun Yat-sen University, 74# Zhongshan Er Road, Guangzhou, 510080, China
| | - Z Li
- Department of Pathology, The First Affiliated hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Y Lin
- Department of Pathology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, China
| | - X Gou
- Department of Pathology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, China
| | - Y Zhu
- Department of Histology and Embryology, Medical school of Sun Yat-sen University, 74# Zhongshan Er Road, Guangzhou, 510080, China.
| | - H Wang
- Department of Neurosurgery and Pituitary Tumor Center, Zhongshan School of Medicine, The First Affiliated Hospital of Sun Yat-sen University, 52# Zhongshan Er Road, Guangzhou, 510080, China.
| |
Collapse
|
42
|
Fernandez-Rodriguez E, Casanueva FF, Bernabeu I. Update on prognostic factors in acromegaly: Is a risk score possible? Pituitary 2015; 18:431-40. [PMID: 24858722 DOI: 10.1007/s11102-014-0574-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Certain clinical conditions and markers have recently been demonstrated to modify the natural history of acromegaly in affected patients. Thus, some clinical, histological, radiological and molecular factors are associated with more aggressive pituitary tumors that have higher biochemical activity, higher tumor volumes and decreased tumoral and biochemical responses to current therapies. However, these factors do not seem to have an equal influence on the prognosis of patients with acromegaly. We present a review of the factors that influence the clinical course of patients with acromegaly and propose a risk value for each factor that will allow prognostic scoring for affected patients by considering a combination of these factors.
Collapse
Affiliation(s)
- E Fernandez-Rodriguez
- Endocrinology Division, Servicio Gallego de Salud (SERGAS), Complejo Hospitalario Universitario de Santiago de Compostela, 15706, Santiago de Compostela, La Coruña, Spain
| | | | | |
Collapse
|
43
|
Peverelli E, Treppiedi D, Giardino E, Vitali E, Lania AG, Mantovani G. Dopamine and Somatostatin Analogues Resistance of Pituitary Tumors: Focus on Cytoskeleton Involvement. Front Endocrinol (Lausanne) 2015; 6:187. [PMID: 26733942 PMCID: PMC4686608 DOI: 10.3389/fendo.2015.00187] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 12/07/2015] [Indexed: 12/15/2022] Open
Abstract
Pituitary tumors, that origin from excessive proliferation of a specific subtype of pituitary cell, are mostly benign tumors, but may cause significant morbidity in affected patients, including visual and neurologic manifestations from mass-effect, or endocrine syndromes caused by hormone hypersecretion. Dopamine (DA) receptor DRD2 and somatostatin (SS) receptors (SSTRs) represent the main targets of pharmacological treatment of pituitary tumors since they mediate inhibitory effects on both hormone secretion and cell proliferation, and their expression is retained by most of these tumors. Although long-acting DA and SS analogs are currently used in the treatment of prolactin (PRL)- and growth hormone (GH)-secreting pituitary tumors, respectively, clinical practice indicates a great variability in the frequency and entity of favorable responses. The molecular basis of the pharmacological resistance are still poorly understood, and several potential molecular mechanisms have been proposed, including defective expression or genetic alterations of DRD2 and SSTRs, or an impaired signal transduction. Recently, a role for cytoskeleton protein filamin A (FLNA) in DRD2 and SSTRs receptors expression and signaling in PRL- and GH-secreting tumors, respectively, has been demonstrated, first revealing a link between FLNA expression and responsiveness of pituitary tumors to pharmacological therapy. This review provides an overview of the known molecular events involved in SS and DA resistance, focusing on the role played by FLNA.
Collapse
Affiliation(s)
- Erika Peverelli
- Endocrinology and Diabetology Unit, Department of Clinical Sciences and Community Health, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Donatella Treppiedi
- Endocrinology and Diabetology Unit, Department of Clinical Sciences and Community Health, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Elena Giardino
- Endocrinology and Diabetology Unit, Department of Clinical Sciences and Community Health, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Eleonora Vitali
- Laboratory of Cellular and Molecular Endocrinology, IRCCS Clinical and Research Institute Humanitas, Milan, Italy
| | - Andrea G. Lania
- Endocrine Unit, IRCCS Humanitas Clinical Institute, University of Milan, Milan, Italy
| | - Giovanna Mantovani
- Endocrinology and Diabetology Unit, Department of Clinical Sciences and Community Health, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
- *Correspondence: Giovanna Mantovani,
| |
Collapse
|
44
|
Norden AD, Ligon KL, Hammond SN, Muzikansky A, Reardon DA, Kaley TJ, Batchelor TT, Plotkin SR, Raizer JJ, Wong ET, Drappatz J, Lesser GJ, Haidar S, Beroukhim R, Lee EQ, Doherty L, Lafrankie D, Gaffey SC, Gerard M, Smith KH, McCluskey C, Phuphanich S, Wen PY. Phase II study of monthly pasireotide LAR (SOM230C) for recurrent or progressive meningioma. Neurology 2014; 84:280-6. [PMID: 25527270 DOI: 10.1212/wnl.0000000000001153] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE A subset of meningiomas recur after surgery and radiation therapy, but no medical therapy for recurrent meningioma has proven effective. METHODS Pasireotide LAR is a long-acting somatostatin analog that may inhibit meningioma growth. This was a phase II trial in patients with histologically confirmed recurrent or progressive meningioma designed to evaluate whether pasireotide LAR prolongs progression-free survival at 6 months (PFS6). Patients were stratified by histology (atypical [World Health Organization grade 2] and malignant [grade 3] meningiomas in cohort A and benign [grade 3] in cohort B). RESULTS Eighteen patients were accrued in cohort A and 16 in cohort B. Cohort A had median age 59 years, median Karnofsky performance status 80, 17 (94%) had previous radiation therapy, and 11 (61%) showed high octreotide uptake. Cohort B had median age 52 years, median Karnofsky performance status 90, 11 (69%) had previous radiation therapy, and 12 (75%) showed high octreotide uptake. There were no radiographic responses to pasireotide LAR therapy in either cohort. Twelve patients (67%) in cohort A and 13 (81%) in cohort B achieved stable disease. In cohort A, PFS6 was 17% and median PFS 15 weeks (95% confidence interval: 8-20). In cohort B, PFS6 was 50% and median PFS 26 weeks (12-43). Treatment was well tolerated. Octreotide uptake and insulin-like growth factor-1 levels did not predict outcome. Expression of somatostatin receptor 3 predicted favorable PFS and overall survival. CONCLUSIONS Pasireotide LAR has limited activity in recurrent meningiomas. The finding that somatostatin receptor 3 is associated with favorable outcomes warrants further investigation. CLASSIFICATION OF EVIDENCE This study provides Class IV evidence that in patients with recurrent or progressive meningioma, pasireotide LAR does not significantly increase the proportion of patients with PFS at 6 months.
Collapse
Affiliation(s)
- Andrew D Norden
- From the Division of Cancer Neurology, Department of Neurology (A.D.N., E.Q.L., P.Y.W.), and Departments of Pathology (K.L.L.) and Medicine (R.B.), Brigham and Women's Hospital, Boston, MA; Center for Neuro-Oncology (A.D.N., S.N.H., D.A.R., R.B., E.Q.L., L.D., D.L., M.G., K.H.S., C.M., P.Y.W.), Dana-Farber/Brigham and Women's Cancer Center; Department of Medicine (R.B.), Harvard Medical School (A.D.N., K.L.L., D.A.R., T.T.B., S.R.P., E.T.W., R.B., E.Q.L., P.Y.W.), Boston; Department of Medical Oncology, Center for Molecular Oncologic Pathology (K.L.L., S.H.), and Departments of Medical Oncology and Cancer Biology (R.B.), Dana-Farber Cancer Institute, Boston; Massachusetts General Hospital Biostatistics Center (A.M.); Brain Tumor Center (T.J.K.), Memorial Sloan-Kettering Cancer Center, New York, NY; Pappas Center for Neuro-Oncology (T.T.B., S.R.P.), Massachusetts General Hospital, Boston; Department of Neurology (J.J.R.), Northwestern University Feinberg School of Medicine, Chicago, IL; Brain Tumor Center (E.T.W.), Beth-Israel Deaconess Medical Center, Boston; Adult Neuro-Oncology Program (J.D.), University of Pittsburgh Medical Center, Pittsburgh, PA; Comprehensive Cancer Center (G.J.L.), Wake Forest University Baptist Medical Center, Winston-Salem, NC; and Departments of Neurosurgery and Neurology (S.P.), Cedars-Sinai Medical Center, Los Angeles, CA
| | - Keith L Ligon
- From the Division of Cancer Neurology, Department of Neurology (A.D.N., E.Q.L., P.Y.W.), and Departments of Pathology (K.L.L.) and Medicine (R.B.), Brigham and Women's Hospital, Boston, MA; Center for Neuro-Oncology (A.D.N., S.N.H., D.A.R., R.B., E.Q.L., L.D., D.L., M.G., K.H.S., C.M., P.Y.W.), Dana-Farber/Brigham and Women's Cancer Center; Department of Medicine (R.B.), Harvard Medical School (A.D.N., K.L.L., D.A.R., T.T.B., S.R.P., E.T.W., R.B., E.Q.L., P.Y.W.), Boston; Department of Medical Oncology, Center for Molecular Oncologic Pathology (K.L.L., S.H.), and Departments of Medical Oncology and Cancer Biology (R.B.), Dana-Farber Cancer Institute, Boston; Massachusetts General Hospital Biostatistics Center (A.M.); Brain Tumor Center (T.J.K.), Memorial Sloan-Kettering Cancer Center, New York, NY; Pappas Center for Neuro-Oncology (T.T.B., S.R.P.), Massachusetts General Hospital, Boston; Department of Neurology (J.J.R.), Northwestern University Feinberg School of Medicine, Chicago, IL; Brain Tumor Center (E.T.W.), Beth-Israel Deaconess Medical Center, Boston; Adult Neuro-Oncology Program (J.D.), University of Pittsburgh Medical Center, Pittsburgh, PA; Comprehensive Cancer Center (G.J.L.), Wake Forest University Baptist Medical Center, Winston-Salem, NC; and Departments of Neurosurgery and Neurology (S.P.), Cedars-Sinai Medical Center, Los Angeles, CA
| | - Samantha N Hammond
- From the Division of Cancer Neurology, Department of Neurology (A.D.N., E.Q.L., P.Y.W.), and Departments of Pathology (K.L.L.) and Medicine (R.B.), Brigham and Women's Hospital, Boston, MA; Center for Neuro-Oncology (A.D.N., S.N.H., D.A.R., R.B., E.Q.L., L.D., D.L., M.G., K.H.S., C.M., P.Y.W.), Dana-Farber/Brigham and Women's Cancer Center; Department of Medicine (R.B.), Harvard Medical School (A.D.N., K.L.L., D.A.R., T.T.B., S.R.P., E.T.W., R.B., E.Q.L., P.Y.W.), Boston; Department of Medical Oncology, Center for Molecular Oncologic Pathology (K.L.L., S.H.), and Departments of Medical Oncology and Cancer Biology (R.B.), Dana-Farber Cancer Institute, Boston; Massachusetts General Hospital Biostatistics Center (A.M.); Brain Tumor Center (T.J.K.), Memorial Sloan-Kettering Cancer Center, New York, NY; Pappas Center for Neuro-Oncology (T.T.B., S.R.P.), Massachusetts General Hospital, Boston; Department of Neurology (J.J.R.), Northwestern University Feinberg School of Medicine, Chicago, IL; Brain Tumor Center (E.T.W.), Beth-Israel Deaconess Medical Center, Boston; Adult Neuro-Oncology Program (J.D.), University of Pittsburgh Medical Center, Pittsburgh, PA; Comprehensive Cancer Center (G.J.L.), Wake Forest University Baptist Medical Center, Winston-Salem, NC; and Departments of Neurosurgery and Neurology (S.P.), Cedars-Sinai Medical Center, Los Angeles, CA
| | - Alona Muzikansky
- From the Division of Cancer Neurology, Department of Neurology (A.D.N., E.Q.L., P.Y.W.), and Departments of Pathology (K.L.L.) and Medicine (R.B.), Brigham and Women's Hospital, Boston, MA; Center for Neuro-Oncology (A.D.N., S.N.H., D.A.R., R.B., E.Q.L., L.D., D.L., M.G., K.H.S., C.M., P.Y.W.), Dana-Farber/Brigham and Women's Cancer Center; Department of Medicine (R.B.), Harvard Medical School (A.D.N., K.L.L., D.A.R., T.T.B., S.R.P., E.T.W., R.B., E.Q.L., P.Y.W.), Boston; Department of Medical Oncology, Center for Molecular Oncologic Pathology (K.L.L., S.H.), and Departments of Medical Oncology and Cancer Biology (R.B.), Dana-Farber Cancer Institute, Boston; Massachusetts General Hospital Biostatistics Center (A.M.); Brain Tumor Center (T.J.K.), Memorial Sloan-Kettering Cancer Center, New York, NY; Pappas Center for Neuro-Oncology (T.T.B., S.R.P.), Massachusetts General Hospital, Boston; Department of Neurology (J.J.R.), Northwestern University Feinberg School of Medicine, Chicago, IL; Brain Tumor Center (E.T.W.), Beth-Israel Deaconess Medical Center, Boston; Adult Neuro-Oncology Program (J.D.), University of Pittsburgh Medical Center, Pittsburgh, PA; Comprehensive Cancer Center (G.J.L.), Wake Forest University Baptist Medical Center, Winston-Salem, NC; and Departments of Neurosurgery and Neurology (S.P.), Cedars-Sinai Medical Center, Los Angeles, CA
| | - David A Reardon
- From the Division of Cancer Neurology, Department of Neurology (A.D.N., E.Q.L., P.Y.W.), and Departments of Pathology (K.L.L.) and Medicine (R.B.), Brigham and Women's Hospital, Boston, MA; Center for Neuro-Oncology (A.D.N., S.N.H., D.A.R., R.B., E.Q.L., L.D., D.L., M.G., K.H.S., C.M., P.Y.W.), Dana-Farber/Brigham and Women's Cancer Center; Department of Medicine (R.B.), Harvard Medical School (A.D.N., K.L.L., D.A.R., T.T.B., S.R.P., E.T.W., R.B., E.Q.L., P.Y.W.), Boston; Department of Medical Oncology, Center for Molecular Oncologic Pathology (K.L.L., S.H.), and Departments of Medical Oncology and Cancer Biology (R.B.), Dana-Farber Cancer Institute, Boston; Massachusetts General Hospital Biostatistics Center (A.M.); Brain Tumor Center (T.J.K.), Memorial Sloan-Kettering Cancer Center, New York, NY; Pappas Center for Neuro-Oncology (T.T.B., S.R.P.), Massachusetts General Hospital, Boston; Department of Neurology (J.J.R.), Northwestern University Feinberg School of Medicine, Chicago, IL; Brain Tumor Center (E.T.W.), Beth-Israel Deaconess Medical Center, Boston; Adult Neuro-Oncology Program (J.D.), University of Pittsburgh Medical Center, Pittsburgh, PA; Comprehensive Cancer Center (G.J.L.), Wake Forest University Baptist Medical Center, Winston-Salem, NC; and Departments of Neurosurgery and Neurology (S.P.), Cedars-Sinai Medical Center, Los Angeles, CA
| | - Thomas J Kaley
- From the Division of Cancer Neurology, Department of Neurology (A.D.N., E.Q.L., P.Y.W.), and Departments of Pathology (K.L.L.) and Medicine (R.B.), Brigham and Women's Hospital, Boston, MA; Center for Neuro-Oncology (A.D.N., S.N.H., D.A.R., R.B., E.Q.L., L.D., D.L., M.G., K.H.S., C.M., P.Y.W.), Dana-Farber/Brigham and Women's Cancer Center; Department of Medicine (R.B.), Harvard Medical School (A.D.N., K.L.L., D.A.R., T.T.B., S.R.P., E.T.W., R.B., E.Q.L., P.Y.W.), Boston; Department of Medical Oncology, Center for Molecular Oncologic Pathology (K.L.L., S.H.), and Departments of Medical Oncology and Cancer Biology (R.B.), Dana-Farber Cancer Institute, Boston; Massachusetts General Hospital Biostatistics Center (A.M.); Brain Tumor Center (T.J.K.), Memorial Sloan-Kettering Cancer Center, New York, NY; Pappas Center for Neuro-Oncology (T.T.B., S.R.P.), Massachusetts General Hospital, Boston; Department of Neurology (J.J.R.), Northwestern University Feinberg School of Medicine, Chicago, IL; Brain Tumor Center (E.T.W.), Beth-Israel Deaconess Medical Center, Boston; Adult Neuro-Oncology Program (J.D.), University of Pittsburgh Medical Center, Pittsburgh, PA; Comprehensive Cancer Center (G.J.L.), Wake Forest University Baptist Medical Center, Winston-Salem, NC; and Departments of Neurosurgery and Neurology (S.P.), Cedars-Sinai Medical Center, Los Angeles, CA
| | - Tracy T Batchelor
- From the Division of Cancer Neurology, Department of Neurology (A.D.N., E.Q.L., P.Y.W.), and Departments of Pathology (K.L.L.) and Medicine (R.B.), Brigham and Women's Hospital, Boston, MA; Center for Neuro-Oncology (A.D.N., S.N.H., D.A.R., R.B., E.Q.L., L.D., D.L., M.G., K.H.S., C.M., P.Y.W.), Dana-Farber/Brigham and Women's Cancer Center; Department of Medicine (R.B.), Harvard Medical School (A.D.N., K.L.L., D.A.R., T.T.B., S.R.P., E.T.W., R.B., E.Q.L., P.Y.W.), Boston; Department of Medical Oncology, Center for Molecular Oncologic Pathology (K.L.L., S.H.), and Departments of Medical Oncology and Cancer Biology (R.B.), Dana-Farber Cancer Institute, Boston; Massachusetts General Hospital Biostatistics Center (A.M.); Brain Tumor Center (T.J.K.), Memorial Sloan-Kettering Cancer Center, New York, NY; Pappas Center for Neuro-Oncology (T.T.B., S.R.P.), Massachusetts General Hospital, Boston; Department of Neurology (J.J.R.), Northwestern University Feinberg School of Medicine, Chicago, IL; Brain Tumor Center (E.T.W.), Beth-Israel Deaconess Medical Center, Boston; Adult Neuro-Oncology Program (J.D.), University of Pittsburgh Medical Center, Pittsburgh, PA; Comprehensive Cancer Center (G.J.L.), Wake Forest University Baptist Medical Center, Winston-Salem, NC; and Departments of Neurosurgery and Neurology (S.P.), Cedars-Sinai Medical Center, Los Angeles, CA
| | - Scott R Plotkin
- From the Division of Cancer Neurology, Department of Neurology (A.D.N., E.Q.L., P.Y.W.), and Departments of Pathology (K.L.L.) and Medicine (R.B.), Brigham and Women's Hospital, Boston, MA; Center for Neuro-Oncology (A.D.N., S.N.H., D.A.R., R.B., E.Q.L., L.D., D.L., M.G., K.H.S., C.M., P.Y.W.), Dana-Farber/Brigham and Women's Cancer Center; Department of Medicine (R.B.), Harvard Medical School (A.D.N., K.L.L., D.A.R., T.T.B., S.R.P., E.T.W., R.B., E.Q.L., P.Y.W.), Boston; Department of Medical Oncology, Center for Molecular Oncologic Pathology (K.L.L., S.H.), and Departments of Medical Oncology and Cancer Biology (R.B.), Dana-Farber Cancer Institute, Boston; Massachusetts General Hospital Biostatistics Center (A.M.); Brain Tumor Center (T.J.K.), Memorial Sloan-Kettering Cancer Center, New York, NY; Pappas Center for Neuro-Oncology (T.T.B., S.R.P.), Massachusetts General Hospital, Boston; Department of Neurology (J.J.R.), Northwestern University Feinberg School of Medicine, Chicago, IL; Brain Tumor Center (E.T.W.), Beth-Israel Deaconess Medical Center, Boston; Adult Neuro-Oncology Program (J.D.), University of Pittsburgh Medical Center, Pittsburgh, PA; Comprehensive Cancer Center (G.J.L.), Wake Forest University Baptist Medical Center, Winston-Salem, NC; and Departments of Neurosurgery and Neurology (S.P.), Cedars-Sinai Medical Center, Los Angeles, CA
| | - Jeffrey J Raizer
- From the Division of Cancer Neurology, Department of Neurology (A.D.N., E.Q.L., P.Y.W.), and Departments of Pathology (K.L.L.) and Medicine (R.B.), Brigham and Women's Hospital, Boston, MA; Center for Neuro-Oncology (A.D.N., S.N.H., D.A.R., R.B., E.Q.L., L.D., D.L., M.G., K.H.S., C.M., P.Y.W.), Dana-Farber/Brigham and Women's Cancer Center; Department of Medicine (R.B.), Harvard Medical School (A.D.N., K.L.L., D.A.R., T.T.B., S.R.P., E.T.W., R.B., E.Q.L., P.Y.W.), Boston; Department of Medical Oncology, Center for Molecular Oncologic Pathology (K.L.L., S.H.), and Departments of Medical Oncology and Cancer Biology (R.B.), Dana-Farber Cancer Institute, Boston; Massachusetts General Hospital Biostatistics Center (A.M.); Brain Tumor Center (T.J.K.), Memorial Sloan-Kettering Cancer Center, New York, NY; Pappas Center for Neuro-Oncology (T.T.B., S.R.P.), Massachusetts General Hospital, Boston; Department of Neurology (J.J.R.), Northwestern University Feinberg School of Medicine, Chicago, IL; Brain Tumor Center (E.T.W.), Beth-Israel Deaconess Medical Center, Boston; Adult Neuro-Oncology Program (J.D.), University of Pittsburgh Medical Center, Pittsburgh, PA; Comprehensive Cancer Center (G.J.L.), Wake Forest University Baptist Medical Center, Winston-Salem, NC; and Departments of Neurosurgery and Neurology (S.P.), Cedars-Sinai Medical Center, Los Angeles, CA
| | - Eric T Wong
- From the Division of Cancer Neurology, Department of Neurology (A.D.N., E.Q.L., P.Y.W.), and Departments of Pathology (K.L.L.) and Medicine (R.B.), Brigham and Women's Hospital, Boston, MA; Center for Neuro-Oncology (A.D.N., S.N.H., D.A.R., R.B., E.Q.L., L.D., D.L., M.G., K.H.S., C.M., P.Y.W.), Dana-Farber/Brigham and Women's Cancer Center; Department of Medicine (R.B.), Harvard Medical School (A.D.N., K.L.L., D.A.R., T.T.B., S.R.P., E.T.W., R.B., E.Q.L., P.Y.W.), Boston; Department of Medical Oncology, Center for Molecular Oncologic Pathology (K.L.L., S.H.), and Departments of Medical Oncology and Cancer Biology (R.B.), Dana-Farber Cancer Institute, Boston; Massachusetts General Hospital Biostatistics Center (A.M.); Brain Tumor Center (T.J.K.), Memorial Sloan-Kettering Cancer Center, New York, NY; Pappas Center for Neuro-Oncology (T.T.B., S.R.P.), Massachusetts General Hospital, Boston; Department of Neurology (J.J.R.), Northwestern University Feinberg School of Medicine, Chicago, IL; Brain Tumor Center (E.T.W.), Beth-Israel Deaconess Medical Center, Boston; Adult Neuro-Oncology Program (J.D.), University of Pittsburgh Medical Center, Pittsburgh, PA; Comprehensive Cancer Center (G.J.L.), Wake Forest University Baptist Medical Center, Winston-Salem, NC; and Departments of Neurosurgery and Neurology (S.P.), Cedars-Sinai Medical Center, Los Angeles, CA
| | - Jan Drappatz
- From the Division of Cancer Neurology, Department of Neurology (A.D.N., E.Q.L., P.Y.W.), and Departments of Pathology (K.L.L.) and Medicine (R.B.), Brigham and Women's Hospital, Boston, MA; Center for Neuro-Oncology (A.D.N., S.N.H., D.A.R., R.B., E.Q.L., L.D., D.L., M.G., K.H.S., C.M., P.Y.W.), Dana-Farber/Brigham and Women's Cancer Center; Department of Medicine (R.B.), Harvard Medical School (A.D.N., K.L.L., D.A.R., T.T.B., S.R.P., E.T.W., R.B., E.Q.L., P.Y.W.), Boston; Department of Medical Oncology, Center for Molecular Oncologic Pathology (K.L.L., S.H.), and Departments of Medical Oncology and Cancer Biology (R.B.), Dana-Farber Cancer Institute, Boston; Massachusetts General Hospital Biostatistics Center (A.M.); Brain Tumor Center (T.J.K.), Memorial Sloan-Kettering Cancer Center, New York, NY; Pappas Center for Neuro-Oncology (T.T.B., S.R.P.), Massachusetts General Hospital, Boston; Department of Neurology (J.J.R.), Northwestern University Feinberg School of Medicine, Chicago, IL; Brain Tumor Center (E.T.W.), Beth-Israel Deaconess Medical Center, Boston; Adult Neuro-Oncology Program (J.D.), University of Pittsburgh Medical Center, Pittsburgh, PA; Comprehensive Cancer Center (G.J.L.), Wake Forest University Baptist Medical Center, Winston-Salem, NC; and Departments of Neurosurgery and Neurology (S.P.), Cedars-Sinai Medical Center, Los Angeles, CA
| | - Glenn J Lesser
- From the Division of Cancer Neurology, Department of Neurology (A.D.N., E.Q.L., P.Y.W.), and Departments of Pathology (K.L.L.) and Medicine (R.B.), Brigham and Women's Hospital, Boston, MA; Center for Neuro-Oncology (A.D.N., S.N.H., D.A.R., R.B., E.Q.L., L.D., D.L., M.G., K.H.S., C.M., P.Y.W.), Dana-Farber/Brigham and Women's Cancer Center; Department of Medicine (R.B.), Harvard Medical School (A.D.N., K.L.L., D.A.R., T.T.B., S.R.P., E.T.W., R.B., E.Q.L., P.Y.W.), Boston; Department of Medical Oncology, Center for Molecular Oncologic Pathology (K.L.L., S.H.), and Departments of Medical Oncology and Cancer Biology (R.B.), Dana-Farber Cancer Institute, Boston; Massachusetts General Hospital Biostatistics Center (A.M.); Brain Tumor Center (T.J.K.), Memorial Sloan-Kettering Cancer Center, New York, NY; Pappas Center for Neuro-Oncology (T.T.B., S.R.P.), Massachusetts General Hospital, Boston; Department of Neurology (J.J.R.), Northwestern University Feinberg School of Medicine, Chicago, IL; Brain Tumor Center (E.T.W.), Beth-Israel Deaconess Medical Center, Boston; Adult Neuro-Oncology Program (J.D.), University of Pittsburgh Medical Center, Pittsburgh, PA; Comprehensive Cancer Center (G.J.L.), Wake Forest University Baptist Medical Center, Winston-Salem, NC; and Departments of Neurosurgery and Neurology (S.P.), Cedars-Sinai Medical Center, Los Angeles, CA
| | - Sam Haidar
- From the Division of Cancer Neurology, Department of Neurology (A.D.N., E.Q.L., P.Y.W.), and Departments of Pathology (K.L.L.) and Medicine (R.B.), Brigham and Women's Hospital, Boston, MA; Center for Neuro-Oncology (A.D.N., S.N.H., D.A.R., R.B., E.Q.L., L.D., D.L., M.G., K.H.S., C.M., P.Y.W.), Dana-Farber/Brigham and Women's Cancer Center; Department of Medicine (R.B.), Harvard Medical School (A.D.N., K.L.L., D.A.R., T.T.B., S.R.P., E.T.W., R.B., E.Q.L., P.Y.W.), Boston; Department of Medical Oncology, Center for Molecular Oncologic Pathology (K.L.L., S.H.), and Departments of Medical Oncology and Cancer Biology (R.B.), Dana-Farber Cancer Institute, Boston; Massachusetts General Hospital Biostatistics Center (A.M.); Brain Tumor Center (T.J.K.), Memorial Sloan-Kettering Cancer Center, New York, NY; Pappas Center for Neuro-Oncology (T.T.B., S.R.P.), Massachusetts General Hospital, Boston; Department of Neurology (J.J.R.), Northwestern University Feinberg School of Medicine, Chicago, IL; Brain Tumor Center (E.T.W.), Beth-Israel Deaconess Medical Center, Boston; Adult Neuro-Oncology Program (J.D.), University of Pittsburgh Medical Center, Pittsburgh, PA; Comprehensive Cancer Center (G.J.L.), Wake Forest University Baptist Medical Center, Winston-Salem, NC; and Departments of Neurosurgery and Neurology (S.P.), Cedars-Sinai Medical Center, Los Angeles, CA
| | - Rameen Beroukhim
- From the Division of Cancer Neurology, Department of Neurology (A.D.N., E.Q.L., P.Y.W.), and Departments of Pathology (K.L.L.) and Medicine (R.B.), Brigham and Women's Hospital, Boston, MA; Center for Neuro-Oncology (A.D.N., S.N.H., D.A.R., R.B., E.Q.L., L.D., D.L., M.G., K.H.S., C.M., P.Y.W.), Dana-Farber/Brigham and Women's Cancer Center; Department of Medicine (R.B.), Harvard Medical School (A.D.N., K.L.L., D.A.R., T.T.B., S.R.P., E.T.W., R.B., E.Q.L., P.Y.W.), Boston; Department of Medical Oncology, Center for Molecular Oncologic Pathology (K.L.L., S.H.), and Departments of Medical Oncology and Cancer Biology (R.B.), Dana-Farber Cancer Institute, Boston; Massachusetts General Hospital Biostatistics Center (A.M.); Brain Tumor Center (T.J.K.), Memorial Sloan-Kettering Cancer Center, New York, NY; Pappas Center for Neuro-Oncology (T.T.B., S.R.P.), Massachusetts General Hospital, Boston; Department of Neurology (J.J.R.), Northwestern University Feinberg School of Medicine, Chicago, IL; Brain Tumor Center (E.T.W.), Beth-Israel Deaconess Medical Center, Boston; Adult Neuro-Oncology Program (J.D.), University of Pittsburgh Medical Center, Pittsburgh, PA; Comprehensive Cancer Center (G.J.L.), Wake Forest University Baptist Medical Center, Winston-Salem, NC; and Departments of Neurosurgery and Neurology (S.P.), Cedars-Sinai Medical Center, Los Angeles, CA
| | - Eudocia Q Lee
- From the Division of Cancer Neurology, Department of Neurology (A.D.N., E.Q.L., P.Y.W.), and Departments of Pathology (K.L.L.) and Medicine (R.B.), Brigham and Women's Hospital, Boston, MA; Center for Neuro-Oncology (A.D.N., S.N.H., D.A.R., R.B., E.Q.L., L.D., D.L., M.G., K.H.S., C.M., P.Y.W.), Dana-Farber/Brigham and Women's Cancer Center; Department of Medicine (R.B.), Harvard Medical School (A.D.N., K.L.L., D.A.R., T.T.B., S.R.P., E.T.W., R.B., E.Q.L., P.Y.W.), Boston; Department of Medical Oncology, Center for Molecular Oncologic Pathology (K.L.L., S.H.), and Departments of Medical Oncology and Cancer Biology (R.B.), Dana-Farber Cancer Institute, Boston; Massachusetts General Hospital Biostatistics Center (A.M.); Brain Tumor Center (T.J.K.), Memorial Sloan-Kettering Cancer Center, New York, NY; Pappas Center for Neuro-Oncology (T.T.B., S.R.P.), Massachusetts General Hospital, Boston; Department of Neurology (J.J.R.), Northwestern University Feinberg School of Medicine, Chicago, IL; Brain Tumor Center (E.T.W.), Beth-Israel Deaconess Medical Center, Boston; Adult Neuro-Oncology Program (J.D.), University of Pittsburgh Medical Center, Pittsburgh, PA; Comprehensive Cancer Center (G.J.L.), Wake Forest University Baptist Medical Center, Winston-Salem, NC; and Departments of Neurosurgery and Neurology (S.P.), Cedars-Sinai Medical Center, Los Angeles, CA
| | - Lisa Doherty
- From the Division of Cancer Neurology, Department of Neurology (A.D.N., E.Q.L., P.Y.W.), and Departments of Pathology (K.L.L.) and Medicine (R.B.), Brigham and Women's Hospital, Boston, MA; Center for Neuro-Oncology (A.D.N., S.N.H., D.A.R., R.B., E.Q.L., L.D., D.L., M.G., K.H.S., C.M., P.Y.W.), Dana-Farber/Brigham and Women's Cancer Center; Department of Medicine (R.B.), Harvard Medical School (A.D.N., K.L.L., D.A.R., T.T.B., S.R.P., E.T.W., R.B., E.Q.L., P.Y.W.), Boston; Department of Medical Oncology, Center for Molecular Oncologic Pathology (K.L.L., S.H.), and Departments of Medical Oncology and Cancer Biology (R.B.), Dana-Farber Cancer Institute, Boston; Massachusetts General Hospital Biostatistics Center (A.M.); Brain Tumor Center (T.J.K.), Memorial Sloan-Kettering Cancer Center, New York, NY; Pappas Center for Neuro-Oncology (T.T.B., S.R.P.), Massachusetts General Hospital, Boston; Department of Neurology (J.J.R.), Northwestern University Feinberg School of Medicine, Chicago, IL; Brain Tumor Center (E.T.W.), Beth-Israel Deaconess Medical Center, Boston; Adult Neuro-Oncology Program (J.D.), University of Pittsburgh Medical Center, Pittsburgh, PA; Comprehensive Cancer Center (G.J.L.), Wake Forest University Baptist Medical Center, Winston-Salem, NC; and Departments of Neurosurgery and Neurology (S.P.), Cedars-Sinai Medical Center, Los Angeles, CA
| | - Debra Lafrankie
- From the Division of Cancer Neurology, Department of Neurology (A.D.N., E.Q.L., P.Y.W.), and Departments of Pathology (K.L.L.) and Medicine (R.B.), Brigham and Women's Hospital, Boston, MA; Center for Neuro-Oncology (A.D.N., S.N.H., D.A.R., R.B., E.Q.L., L.D., D.L., M.G., K.H.S., C.M., P.Y.W.), Dana-Farber/Brigham and Women's Cancer Center; Department of Medicine (R.B.), Harvard Medical School (A.D.N., K.L.L., D.A.R., T.T.B., S.R.P., E.T.W., R.B., E.Q.L., P.Y.W.), Boston; Department of Medical Oncology, Center for Molecular Oncologic Pathology (K.L.L., S.H.), and Departments of Medical Oncology and Cancer Biology (R.B.), Dana-Farber Cancer Institute, Boston; Massachusetts General Hospital Biostatistics Center (A.M.); Brain Tumor Center (T.J.K.), Memorial Sloan-Kettering Cancer Center, New York, NY; Pappas Center for Neuro-Oncology (T.T.B., S.R.P.), Massachusetts General Hospital, Boston; Department of Neurology (J.J.R.), Northwestern University Feinberg School of Medicine, Chicago, IL; Brain Tumor Center (E.T.W.), Beth-Israel Deaconess Medical Center, Boston; Adult Neuro-Oncology Program (J.D.), University of Pittsburgh Medical Center, Pittsburgh, PA; Comprehensive Cancer Center (G.J.L.), Wake Forest University Baptist Medical Center, Winston-Salem, NC; and Departments of Neurosurgery and Neurology (S.P.), Cedars-Sinai Medical Center, Los Angeles, CA
| | - Sarah C Gaffey
- From the Division of Cancer Neurology, Department of Neurology (A.D.N., E.Q.L., P.Y.W.), and Departments of Pathology (K.L.L.) and Medicine (R.B.), Brigham and Women's Hospital, Boston, MA; Center for Neuro-Oncology (A.D.N., S.N.H., D.A.R., R.B., E.Q.L., L.D., D.L., M.G., K.H.S., C.M., P.Y.W.), Dana-Farber/Brigham and Women's Cancer Center; Department of Medicine (R.B.), Harvard Medical School (A.D.N., K.L.L., D.A.R., T.T.B., S.R.P., E.T.W., R.B., E.Q.L., P.Y.W.), Boston; Department of Medical Oncology, Center for Molecular Oncologic Pathology (K.L.L., S.H.), and Departments of Medical Oncology and Cancer Biology (R.B.), Dana-Farber Cancer Institute, Boston; Massachusetts General Hospital Biostatistics Center (A.M.); Brain Tumor Center (T.J.K.), Memorial Sloan-Kettering Cancer Center, New York, NY; Pappas Center for Neuro-Oncology (T.T.B., S.R.P.), Massachusetts General Hospital, Boston; Department of Neurology (J.J.R.), Northwestern University Feinberg School of Medicine, Chicago, IL; Brain Tumor Center (E.T.W.), Beth-Israel Deaconess Medical Center, Boston; Adult Neuro-Oncology Program (J.D.), University of Pittsburgh Medical Center, Pittsburgh, PA; Comprehensive Cancer Center (G.J.L.), Wake Forest University Baptist Medical Center, Winston-Salem, NC; and Departments of Neurosurgery and Neurology (S.P.), Cedars-Sinai Medical Center, Los Angeles, CA
| | - Mary Gerard
- From the Division of Cancer Neurology, Department of Neurology (A.D.N., E.Q.L., P.Y.W.), and Departments of Pathology (K.L.L.) and Medicine (R.B.), Brigham and Women's Hospital, Boston, MA; Center for Neuro-Oncology (A.D.N., S.N.H., D.A.R., R.B., E.Q.L., L.D., D.L., M.G., K.H.S., C.M., P.Y.W.), Dana-Farber/Brigham and Women's Cancer Center; Department of Medicine (R.B.), Harvard Medical School (A.D.N., K.L.L., D.A.R., T.T.B., S.R.P., E.T.W., R.B., E.Q.L., P.Y.W.), Boston; Department of Medical Oncology, Center for Molecular Oncologic Pathology (K.L.L., S.H.), and Departments of Medical Oncology and Cancer Biology (R.B.), Dana-Farber Cancer Institute, Boston; Massachusetts General Hospital Biostatistics Center (A.M.); Brain Tumor Center (T.J.K.), Memorial Sloan-Kettering Cancer Center, New York, NY; Pappas Center for Neuro-Oncology (T.T.B., S.R.P.), Massachusetts General Hospital, Boston; Department of Neurology (J.J.R.), Northwestern University Feinberg School of Medicine, Chicago, IL; Brain Tumor Center (E.T.W.), Beth-Israel Deaconess Medical Center, Boston; Adult Neuro-Oncology Program (J.D.), University of Pittsburgh Medical Center, Pittsburgh, PA; Comprehensive Cancer Center (G.J.L.), Wake Forest University Baptist Medical Center, Winston-Salem, NC; and Departments of Neurosurgery and Neurology (S.P.), Cedars-Sinai Medical Center, Los Angeles, CA
| | - Katrina H Smith
- From the Division of Cancer Neurology, Department of Neurology (A.D.N., E.Q.L., P.Y.W.), and Departments of Pathology (K.L.L.) and Medicine (R.B.), Brigham and Women's Hospital, Boston, MA; Center for Neuro-Oncology (A.D.N., S.N.H., D.A.R., R.B., E.Q.L., L.D., D.L., M.G., K.H.S., C.M., P.Y.W.), Dana-Farber/Brigham and Women's Cancer Center; Department of Medicine (R.B.), Harvard Medical School (A.D.N., K.L.L., D.A.R., T.T.B., S.R.P., E.T.W., R.B., E.Q.L., P.Y.W.), Boston; Department of Medical Oncology, Center for Molecular Oncologic Pathology (K.L.L., S.H.), and Departments of Medical Oncology and Cancer Biology (R.B.), Dana-Farber Cancer Institute, Boston; Massachusetts General Hospital Biostatistics Center (A.M.); Brain Tumor Center (T.J.K.), Memorial Sloan-Kettering Cancer Center, New York, NY; Pappas Center for Neuro-Oncology (T.T.B., S.R.P.), Massachusetts General Hospital, Boston; Department of Neurology (J.J.R.), Northwestern University Feinberg School of Medicine, Chicago, IL; Brain Tumor Center (E.T.W.), Beth-Israel Deaconess Medical Center, Boston; Adult Neuro-Oncology Program (J.D.), University of Pittsburgh Medical Center, Pittsburgh, PA; Comprehensive Cancer Center (G.J.L.), Wake Forest University Baptist Medical Center, Winston-Salem, NC; and Departments of Neurosurgery and Neurology (S.P.), Cedars-Sinai Medical Center, Los Angeles, CA
| | - Christine McCluskey
- From the Division of Cancer Neurology, Department of Neurology (A.D.N., E.Q.L., P.Y.W.), and Departments of Pathology (K.L.L.) and Medicine (R.B.), Brigham and Women's Hospital, Boston, MA; Center for Neuro-Oncology (A.D.N., S.N.H., D.A.R., R.B., E.Q.L., L.D., D.L., M.G., K.H.S., C.M., P.Y.W.), Dana-Farber/Brigham and Women's Cancer Center; Department of Medicine (R.B.), Harvard Medical School (A.D.N., K.L.L., D.A.R., T.T.B., S.R.P., E.T.W., R.B., E.Q.L., P.Y.W.), Boston; Department of Medical Oncology, Center for Molecular Oncologic Pathology (K.L.L., S.H.), and Departments of Medical Oncology and Cancer Biology (R.B.), Dana-Farber Cancer Institute, Boston; Massachusetts General Hospital Biostatistics Center (A.M.); Brain Tumor Center (T.J.K.), Memorial Sloan-Kettering Cancer Center, New York, NY; Pappas Center for Neuro-Oncology (T.T.B., S.R.P.), Massachusetts General Hospital, Boston; Department of Neurology (J.J.R.), Northwestern University Feinberg School of Medicine, Chicago, IL; Brain Tumor Center (E.T.W.), Beth-Israel Deaconess Medical Center, Boston; Adult Neuro-Oncology Program (J.D.), University of Pittsburgh Medical Center, Pittsburgh, PA; Comprehensive Cancer Center (G.J.L.), Wake Forest University Baptist Medical Center, Winston-Salem, NC; and Departments of Neurosurgery and Neurology (S.P.), Cedars-Sinai Medical Center, Los Angeles, CA
| | - Surasak Phuphanich
- From the Division of Cancer Neurology, Department of Neurology (A.D.N., E.Q.L., P.Y.W.), and Departments of Pathology (K.L.L.) and Medicine (R.B.), Brigham and Women's Hospital, Boston, MA; Center for Neuro-Oncology (A.D.N., S.N.H., D.A.R., R.B., E.Q.L., L.D., D.L., M.G., K.H.S., C.M., P.Y.W.), Dana-Farber/Brigham and Women's Cancer Center; Department of Medicine (R.B.), Harvard Medical School (A.D.N., K.L.L., D.A.R., T.T.B., S.R.P., E.T.W., R.B., E.Q.L., P.Y.W.), Boston; Department of Medical Oncology, Center for Molecular Oncologic Pathology (K.L.L., S.H.), and Departments of Medical Oncology and Cancer Biology (R.B.), Dana-Farber Cancer Institute, Boston; Massachusetts General Hospital Biostatistics Center (A.M.); Brain Tumor Center (T.J.K.), Memorial Sloan-Kettering Cancer Center, New York, NY; Pappas Center for Neuro-Oncology (T.T.B., S.R.P.), Massachusetts General Hospital, Boston; Department of Neurology (J.J.R.), Northwestern University Feinberg School of Medicine, Chicago, IL; Brain Tumor Center (E.T.W.), Beth-Israel Deaconess Medical Center, Boston; Adult Neuro-Oncology Program (J.D.), University of Pittsburgh Medical Center, Pittsburgh, PA; Comprehensive Cancer Center (G.J.L.), Wake Forest University Baptist Medical Center, Winston-Salem, NC; and Departments of Neurosurgery and Neurology (S.P.), Cedars-Sinai Medical Center, Los Angeles, CA
| | - Patrick Y Wen
- From the Division of Cancer Neurology, Department of Neurology (A.D.N., E.Q.L., P.Y.W.), and Departments of Pathology (K.L.L.) and Medicine (R.B.), Brigham and Women's Hospital, Boston, MA; Center for Neuro-Oncology (A.D.N., S.N.H., D.A.R., R.B., E.Q.L., L.D., D.L., M.G., K.H.S., C.M., P.Y.W.), Dana-Farber/Brigham and Women's Cancer Center; Department of Medicine (R.B.), Harvard Medical School (A.D.N., K.L.L., D.A.R., T.T.B., S.R.P., E.T.W., R.B., E.Q.L., P.Y.W.), Boston; Department of Medical Oncology, Center for Molecular Oncologic Pathology (K.L.L., S.H.), and Departments of Medical Oncology and Cancer Biology (R.B.), Dana-Farber Cancer Institute, Boston; Massachusetts General Hospital Biostatistics Center (A.M.); Brain Tumor Center (T.J.K.), Memorial Sloan-Kettering Cancer Center, New York, NY; Pappas Center for Neuro-Oncology (T.T.B., S.R.P.), Massachusetts General Hospital, Boston; Department of Neurology (J.J.R.), Northwestern University Feinberg School of Medicine, Chicago, IL; Brain Tumor Center (E.T.W.), Beth-Israel Deaconess Medical Center, Boston; Adult Neuro-Oncology Program (J.D.), University of Pittsburgh Medical Center, Pittsburgh, PA; Comprehensive Cancer Center (G.J.L.), Wake Forest University Baptist Medical Center, Winston-Salem, NC; and Departments of Neurosurgery and Neurology (S.P.), Cedars-Sinai Medical Center, Los Angeles, CA.
| |
Collapse
|
45
|
Syro LV, Builes CE, Di Ieva A, Sav A, Rotondo F, Kovacs K. Improving differential diagnosis of pituitary adenomas. Expert Rev Endocrinol Metab 2014; 9:377-386. [PMID: 30763997 DOI: 10.1586/17446651.2014.922412] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Pituitary adenomas are common tumors arising in adenohypophysial cells or their precursors. For improving control of the disease an early diagnosis is important. Initially considered sporadic tumors, some of them are associated with familial syndromes and their recognition and classification is also required. Morphologically, pituitary adenomas represent a heterogeneous group of tumors with several subtypes and different clinical behavior thus a precise pathological diagnosis is crucial. The simple diagnosis of pituitary adenoma is not satisfactory and the correct classification of histological subtypes may predict aggressiveness in the majority of cases. Although considered not malignant, some of them are clinically aggressive and their recognition remains a challenge. In this paper we present the recent advances in the event of improving early recognition and differential diagnosis of pituitary tumors.
Collapse
Affiliation(s)
- Luis V Syro
- a Department of Neurosurgery, Hospital Pablo Tobon Uribe and Clinica Medellin, Calle 54 # 46-27, Cons 501, Medellin, Colombia
| | - Carlos E Builes
- b Department of Endocrinology, Hospital Pablo Tobon Uribe, Medellin, Colombia
| | - Antonio Di Ieva
- c Department of Surgery, Division of Neurosurgery, St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Aydin Sav
- d Department of Pathology, Acibadem University, School of Medicine, Atasehir, Istanbul, Turkey
| | - Fabio Rotondo
- e Laboratory Medicine, St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Kalman Kovacs
- e Laboratory Medicine, St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
46
|
Cuevas-Ramos D, Fleseriu M. Somatostatin receptor ligands and resistance to treatment in pituitary adenomas. J Mol Endocrinol 2014; 52:R223-40. [PMID: 24647046 DOI: 10.1530/jme-14-0011] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Somatostatin (SST), an inhibitory polypeptide with two biologically active forms SST14 and SST28, inhibits GH, prolactin (PRL), TSH, and ACTH secretion in the anterior pituitary gland. SST also has an antiproliferative effect inducing cell cycle arrest and apoptosis. Such actions are mediated through five G-protein-coupled somatostatin receptors (SSTR): SSTR1-SSTR5. In GH-secreting adenomas, SSTR2 expression predominates, and somatostatin receptor ligands (SRLs; octreotide and lanreotide) directed to SSTR2 are presently the mainstays of medical therapy. However, about half of patients show incomplete biochemical remission, but the definition of resistance per se remains controversial. We summarize here the determinants of SRL resistance in acromegaly patients, including clinical, imaging features as well as molecular (mutations, SSTR variants, and polymorphisms), and histopathological (granulation pattern, and proteins and receptor expression) predictors. The role of SSTR5 may explain the partial responsiveness to SRLs in patients with adequate SSTR2 density in the cell membrane. In patients with ACTH-secreting pituitary adenomas, i.e. Cushing's disease (CD), SSTR5 is the most abundant receptor expressed and tumors show low SSTR2 density due to hypercortisolism-induced SSTR2 down-regulation. Clinical studies with pasireotide, a multireceptor-targeted SRL with increased SSTR5 activity, lead to approval of pasireotide for treatment of patients with CD. Other SRL delivery modes (oral octreotide), multireceptor-targeted SRL (somatoprim) or chimeric compounds targeting dopamine D2 receptors and SSTR2 (dopastatin), are briefly discussed.
Collapse
Affiliation(s)
- Daniel Cuevas-Ramos
- Department of MedicinePituitary Center, Cedars-Sinai Medical Center, Los Angeles, California, USANorthwest Pituitary Center and Departments of Medicine and Neurological SurgeryOregon Health and Science University, 3181 SW Sam Jackson Park Road (BTE 472), Portland, Oregon 97239, USA
| | - Maria Fleseriu
- Department of MedicinePituitary Center, Cedars-Sinai Medical Center, Los Angeles, California, USANorthwest Pituitary Center and Departments of Medicine and Neurological SurgeryOregon Health and Science University, 3181 SW Sam Jackson Park Road (BTE 472), Portland, Oregon 97239, USA
| |
Collapse
|
47
|
Schulz S, Lehmann A, Kliewer A, Nagel F. Fine-tuning somatostatin receptor signalling by agonist-selective phosphorylation and dephosphorylation: IUPHAR Review 5. Br J Pharmacol 2014; 171:1591-9. [PMID: 24328848 PMCID: PMC3966740 DOI: 10.1111/bph.12551] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Revised: 10/08/2013] [Accepted: 10/31/2013] [Indexed: 12/28/2022] Open
Abstract
The biological actions of somatostatin are mediated by a family of five GPCRs, named sst1 to sst5 . Somatostatin receptors exhibit equally high-binding affinities to their natural ligand somatostatin-14 and largely overlapping distributions. The overexpression of somatostatin receptors in human tumours is the molecular basis for diagnostic and therapeutic application of the stable somatostatin analogues octreotide, lanreotide and pasireotide. The efficiency of somatostatin receptor signalling is tightly regulated and ultimately limited by the coordinated phosphorylation and dephosphorylation of intracellular carboxyl-terminal serine and threonine residues. Here, we review and discuss recent progress in the generation and application of phosphosite-specific antibodies for human sst2 and sst5 receptors. These phosphosite-specific antibodies are unique tools to monitor the spatial and temporal dynamics of receptors phosphorylation and dephosphorylation. Using a combined approach of phosphosite-specific antibodies and siRNA knock-down screening, relevant kinases and phosphatases were identified. Emerging evidence suggests distinct mechanisms of agonist-selective fine-tuning for individual somatostatin receptors. The recently uncovered differences in phosphorylation and dephosphorylation of these receptors may hence be of physiological significance in mediating responses to acute, persistent or repeated stimuli in a variety of target tissues.
Collapse
Affiliation(s)
- Stefan Schulz
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-UniversityJena, Germany
| | - Andreas Lehmann
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-UniversityJena, Germany
| | - Andrea Kliewer
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-UniversityJena, Germany
| | - Falko Nagel
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-UniversityJena, Germany
| |
Collapse
|
48
|
Eigler T, Ben-Shlomo A, Zhou C, Khalafi R, Ren SG, Melmed S. Constitutive somatostatin receptor subtype-3 signaling suppresses growth hormone synthesis. Mol Endocrinol 2014; 28:554-64. [PMID: 24606125 PMCID: PMC3968402 DOI: 10.1210/me.2013-1327] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Somatostatin signals through somatostatin receptor subtypes (SSTR) 2 and 5 to attenuate GH secretion. Although expressed in normal pituitary glands and in GH-secreting pituitary tumors, SSTR3 function was unclear, and we have now determined the role of SSTR3 in somatotroph function. Stable rat pituitary tumor cell (GC) transfectants of human SSTR3 (GpSSTR3(WT)) showed suppression of rat (r) GH promoter activity, GH mRNA expression, and secreted GH concordant with suppressed cAMP/protein kinase A (PKA) signaling. In contrast, cAMP levels and GH expression were unchanged in cells expressing a mutant SSTR3 DRY motif (GpSSTR3(R141A)). GH expression was rescued by treatment of GpSSTR3(WT) with forskolin and 8-bromo-cAMP. GpSSTR3(WT) exhibited activation of glycogen synthase kinase3-β (GSK3-β), a PKA substrate, which was also reversed by 8-Bromo-cAMP treatment. Moreover, SSTR3-dependent GH transcriptional inhibition was rescued by inhibition of GSK3-β. GpSSTR3(WT) exhibited elevated Pit-1 serine phosphorylation and decreased Pit-1 occupancy of the rGH promoter with sustained Pit-1 expression. GSK3-β and Pit-1 physically interacted with each other, indicating that Pit-1 may be a GSK3-β phosphorylation substrate. In conclusion, constitutive SSTR3 activity mediates transcriptional repression of GH through cAMP/PKA, leading to subsequent activation of GSK3-β and increased Pit-1 phosphorylation and ultimately attenuating Pit-1 binding to the rGH promoter.
Collapse
Affiliation(s)
- Tamar Eigler
- Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California 90048
| | | | | | | | | | | |
Collapse
|
49
|
Korytnaya E, Barkan A. Pharmacological treatment of acromegaly: its place in the overall therapeutic approach. J Neurooncol 2014; 117:415-20. [PMID: 24442401 DOI: 10.1007/s11060-013-1353-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Accepted: 12/29/2013] [Indexed: 11/29/2022]
|
50
|
Brzana J, Yedinak CG, Gultekin SH, Delashaw JB, Fleseriu M. Growth hormone granulation pattern and somatostatin receptor subtype 2A correlate with postoperative somatostatin receptor ligand response in acromegaly: a large single center experience. Pituitary 2013. [PMID: 23184260 DOI: 10.1007/s11102-012-0445-1] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Acromegaly is associated with serious morbidity and mortality, if not well controlled. Approved somatostatin receptor ligands (SRLs) are a mainstay of medical therapy and exhibit preferential affinity for somatostatin receptor (SSTR) subtype 2. Our objective was to assess whether characteristic features of individual growth hormone (GH)-secreting adenomas at diagnosis, correlated with SRL sensitivity, using defined tumor markers. A retrospective review of 86 consecutive acromegaly surgeries (70 patients) performed between January 2006 and December 2011 was undertaken. Patients with any preoperative medical treatment were excluded. Response to SRL therapy was defined as normalization of insulin-like growth factor 1 (IGF1) and random GH < 1.0 ng/dl. Immunohistochemical staining pattern: sparsely granulated, densely granulated, mixed growth hormone-prolactin (GH/PRL) and SSRT2 positivity (+) were correlated with clinicopathologic features, adenoma recurrence, and SRL treatment response. Two-tailed t test, univariate ANOVA, Kruskal-Wallis and bivariate correlation were performed using PAWS 18. The cohort eligible for analysis comprised 59 patients (41 female and 18 male). Based on pre-surgery adenoma imaging dimensions, 81.3% (48) were macroadenomas and average maximum tumor diameter was 18.1 ± 9.9 mm. Patients on SRLs were followed for 13.4 ± 15.8 (mean ± SD) months. Sparsely granulated adenomas were significantly larger at diagnosis, exhibited lower SSTR2 positivity and had a lower rate of biochemical normalization to SRLs. Densely granulated adenomas were highly responsive to SRLs. Overall, patients with SSTR2A+ adenomas responded more favorably to SRL treatment than those with SSTR2A- adenomas. Eighty-one percent of patients with SSTR2A+ adenomas were biochemically controlled (both GH and IGF1) on SRL treatment, e.g. a much higher normalization rate than that reported in the unselected acromegaly population (20-30%). Detailed knowledge of adenoma GH granularity and the immunohistochemical SSTR2A+ status is a predictor of SRL response. These immunoreactive markers should be assessed routinely on surgical specimens to assess subsequent SRL responsiveness and potential need for adjunctive therapy after surgery.
Collapse
Affiliation(s)
- Jessica Brzana
- OHSU Northwest Pituitary Center, Oregon Health & Science University, 3181 SW Sam Jackson Park Road (BTE 472), Portland, OR, 97239, USA
| | | | | | | | | |
Collapse
|