1
|
Remba-Shapiro I, Nachtigall LB. Treatment of acromegaly with oral octreotide. Best Pract Res Clin Endocrinol Metab 2024; 38:101888. [PMID: 38443224 DOI: 10.1016/j.beem.2024.101888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/07/2024]
Abstract
Acromegaly is a rare disease caused by a growth hormone excess, usually due to a secreting pituitary adenoma. Somatostatin receptor ligands (SRL) are the mainstay of medical therapy for patients with acromegaly who fail to achieve biochemical control post-operatively or are not eligible for surgical treatment. SRLs are typically administered as monthly injections and have shown to be effective in maintaining biochemical and radiological control of acromegaly. However, these injections may cause local adverse events and are associated with increased psychological burden in some patients. Oral octreotide provides a new alternative for patients responding to injectable SRLs. This new formulation has shown to have similar safety and efficacy profiles compared to injectable SRLs and may be a preferable option for some patients with acromegaly. The aim of this review is to provide an overview of the role of oral octreotide in the management of acromegaly.
Collapse
Affiliation(s)
- Ilan Remba-Shapiro
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Lisa B Nachtigall
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
2
|
Sosa LDV, Picech F, Perez P, Gutierrez S, Leal RB, De Paul A, Torres A, Petiti JP. Regulation of FGF2-induced proliferation by inhibitory GPCR in normal pituitary cells. Front Endocrinol (Lausanne) 2023; 14:1183151. [PMID: 37576961 PMCID: PMC10414184 DOI: 10.3389/fendo.2023.1183151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 07/13/2023] [Indexed: 08/15/2023] Open
Abstract
Introduction Intracellular communication is essential for the maintenance of the anterior pituitary gland plasticity. The aim of this study was to evaluate whether GPCR-Gαi modulates basic fibroblast growth factor (FGF2)-induced proliferative activity in normal pituitary cell populations. Methods Anterior pituitary primary cell cultures from Wistar female rats were treated with FGF2 (10ng/mL) or somatostatin analog (SSTa, 100nM) alone or co-incubated with or without the inhibitors of GPCR-Gαi, pertussis toxin (PTX, 500nM), MEK inhibitor (U0126, 100µM) or PI3K inhibitor (LY 294002, 10 μM). Results FGF2 increased and SSTa decreased the lactotroph and somatotroph BrdU uptak2e (p<0.05) whereas the FGF2-induced S-phase entry was prevented by SSTa co-incubation in both cell types, with these effects being reverted by PTX, U0126 or LY294002 pre-incubation. The inhibition of lactotroph and somatotroph mitosis was associated with a downregulation of c-Jun expression, a decrease of phosphorylated (p) ERK and pAKT. Furthermore, SSTa was observed to inhibit the S-phase entry induced by FGF2, resulting in a further increase in the number of cells in the G1 phase and a concomitant reduction in the number of cells in the S phases (p< 0.05), effects related to a decrease of cyclin D1 expression and an increase in the expression of the cell cycle inhibitors p27 and p21. Discussion In summary, the GPCR-Gαi activated by SSTa blocked the pro-proliferative effect of FGF2 in normal pituitary cells via a MEK-dependent mechanism, which acts as a mediator of both anti and pro-mitogenic signals, that may regulate the principal effectors of the G1 to S-phase transition.
Collapse
Affiliation(s)
- Liliana del V. Sosa
- Universidad Nacional de Córdoba, Facultad de Ciencias Médicas, Centro de Microscopía Electrónica, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas Técnicas (CONICET), Instituto de Investigaciones en Ciencias de la Salud (INICSA), Córdoba, Argentina
| | - Florencia Picech
- Universidad Nacional de Córdoba, Facultad de Ciencias Médicas, Centro de Microscopía Electrónica, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas Técnicas (CONICET), Instituto de Investigaciones en Ciencias de la Salud (INICSA), Córdoba, Argentina
| | - Pablo Perez
- Universidad Nacional de Córdoba, Facultad de Ciencias Médicas, Centro de Microscopía Electrónica, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas Técnicas (CONICET), Instituto de Investigaciones en Ciencias de la Salud (INICSA), Córdoba, Argentina
| | - Silvina Gutierrez
- Universidad Nacional de Córdoba, Facultad de Ciencias Médicas, Centro de Microscopía Electrónica, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas Técnicas (CONICET), Instituto de Investigaciones en Ciencias de la Salud (INICSA), Córdoba, Argentina
| | - Rodrigo Bainy Leal
- Universidade Federal de Santa Catarina, Florianópolis, Departamento de Bioquímica e Programa de Pós-graduação em Bioquímica, Centro de Ciências Biológicas, Santa Catarina, Brazil
| | - Ana De Paul
- Universidad Nacional de Córdoba, Facultad de Ciencias Médicas, Centro de Microscopía Electrónica, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas Técnicas (CONICET), Instituto de Investigaciones en Ciencias de la Salud (INICSA), Córdoba, Argentina
| | - Alicia Torres
- Universidad Nacional de Córdoba, Facultad de Ciencias Médicas, Centro de Microscopía Electrónica, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas Técnicas (CONICET), Instituto de Investigaciones en Ciencias de la Salud (INICSA), Córdoba, Argentina
| | - Juan Pablo Petiti
- Universidad Nacional de Córdoba, Facultad de Ciencias Médicas, Centro de Microscopía Electrónica, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas Técnicas (CONICET), Instituto de Investigaciones en Ciencias de la Salud (INICSA), Córdoba, Argentina
| |
Collapse
|
3
|
Jensen LR, Maier AD, Lomstein A, Graillon T, Hrachova M, Bota D, Ruiz-Patiño A, Arrieta O, Cardona AF, Rudà R, Furtner J, Roeckle U, Clement P, Preusser M, Scheie D, Broholm H, Kristensen BW, Skjøth-Rasmussen J, Ziebell M, Munch TN, Fugleholm K, Walter MA, Mathiesen T, Mirian C. Somatostatin analogues in treatment-refractory meningioma: a systematic review with meta-analysis of individual patient data. Neurosurg Rev 2022; 45:3067-3081. [PMID: 35984552 DOI: 10.1007/s10143-022-01849-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 08/01/2022] [Accepted: 08/10/2022] [Indexed: 11/30/2022]
Abstract
Treatment-refractory meningiomas have a dismal prognosis and limited treatment options. Meningiomas express high-densities of somatostatin receptors (SSTR), thus potentially susceptible to antitumorigenic effects of somatostatin analogues (SSA). Evidence for SSA in meningiomas is scarce, and it is unclear if published literature would either (1) support wider use of SSA, if (2) more evidence is desirable, or if (3) available evidence is sufficient to discard SSA. We addressed the need for more evidence with a systematic review and meta-analysis. We performed an individual patient data (IPD) meta-analysis. Main outcomes were toxicity, best radiological response, progression-free survival, and overall survival. We applied multivariable logistic regression models to estimate the effect of SSA on the probability of obtaining radiological disease control. The predictive performance was evaluated using area under the curve and Brier scores. We included 16 studies and compiled IPD from 8/9 of all previous cohorts. Quality of evidence was overall ranked "very low." Stable disease was reported in 58% of patients as best radiological response. Per 100 mg increase in total SSA dosage, the odds ratios for obtaining radiological disease control was 1.42 (1.11 to 1.81, P = 0.005) and 1.44 (1.00 to 2.08, P = 0.05) for patients treated with SSA as monodrug therapy vs SSA in combination with everolimus, respectively. Low quality of evidence impeded exact quantification of treatment efficacy, and the association between response and treatment may represent reverse causality. Yet, the SSA treatment was well tolerated, and beneficial effect cannot be disqualified. A prospective trial without bias from inconsistent study designs is warranted to assess SSA therapy for well-defined meningioma subgroups.
Collapse
Affiliation(s)
- Lasse Rehné Jensen
- Department of Neurosurgery, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Andrea Daniela Maier
- Department of Neurosurgery, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Department of Pathology, Center of Diagnostic Investigation, Copenhagen University Hospital, Copenhagen, Denmark
| | - Atle Lomstein
- Department of Neurology, Aarhus University Hospital, Aarhus, Denmark
| | - Thomas Graillon
- Department of Neurosurgery, Hospital La Timone, Aix Marseille University, APHM, INSERM, MMG, Marseille, France
| | - Maya Hrachova
- Department of Neurosurgery, University of Oklahoma Health Science Center, Oklahoma, OK, USA
| | - Daniela Bota
- Department of Neurology, UC Irvine Medical Center, Orange, CA, USA
- Department of Neurological Surgery, UC Irvine Medical Center, Orange, CA, USA
| | | | - Oscar Arrieta
- Thoracic Oncology Unit, Instituto Nacional de Cancerología (INCaN), Mexico City, México
| | | | - Roberta Rudà
- Department of Neurology, Castelfranco Veneto/Treviso, Treviso, Italy
- Department of Neuro-Oncology, City of Health and Science Hospital and University of Turin, Turin, Italy
| | - Julia Furtner
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Ulrich Roeckle
- Department of Neurology and Brain Tumor Center, Cantonal Hospital Aarau, Aarau, Switzerland
| | - Paul Clement
- Department of Oncology, Leuven Cancer Institute, KU Leuven, Louvain, Belgium
| | - Matthias Preusser
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - David Scheie
- Department of Pathology, Center of Diagnostic Investigation, Copenhagen University Hospital, Copenhagen, Denmark
| | - Helle Broholm
- Department of Pathology, Center of Diagnostic Investigation, Copenhagen University Hospital, Copenhagen, Denmark
| | - Bjarne Winther Kristensen
- Department of Clinical Neuroscience, Section for Neurosurgery, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Medicine and Biotech Research and Innovation Center (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Jane Skjøth-Rasmussen
- Department of Neurosurgery, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Morten Ziebell
- Department of Neurosurgery, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Tina Nørgaard Munch
- Department of Neurosurgery, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - Kåre Fugleholm
- Department of Neurosurgery, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Martin A Walter
- Department of Nuclear Medicine, University Hospital of Geneva, Geneva, Switzerland
- Gesundheitswissenschaften Und Medizin EN, University of Lucerne, Lucerne, Switzerland
| | - Tiit Mathiesen
- Department of Neurosurgery, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Institute of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Christian Mirian
- Department of Neurosurgery, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
4
|
Somatostatin and Its Receptor System in Colorectal Cancer. Biomedicines 2021; 9:biomedicines9111743. [PMID: 34829972 PMCID: PMC8615525 DOI: 10.3390/biomedicines9111743] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/19/2021] [Accepted: 11/20/2021] [Indexed: 12/14/2022] Open
Abstract
Somatostatin (SST)/somatotropin release-inhibiting factor (SRIF) is a well-known neuropeptide, widely distributed in the central and peripheral nervous systems, that regulates the endocrine system and affects neurotransmission via interaction with five SST receptors (SST1-5). In the gastrointestinal tract, the main SST-producing cells include intestinal enteroendocrine cells (EECs) restricted to the mucosa, and neurons of the submucosal and myenteric plexuses. The action of the SRIF system is based on the inhibition of endocrine and exocrine secretion, as well as the proliferative responses of target cells. The SST1–5 share common signaling pathways, and are not only widely expressed on normal tissues, but also frequently overexpressed by several tumors, particularly neuroendocrine neoplasms (NENs). Furthermore, the SRIF system represents the only peptide/G protein-coupled receptor (GPCR) system with multiple approved clinical applications for the diagnosis and treatment of several NENs. The role of the SRIF system in the histogenesis of colorectal cancer (CRC) subtypes (e.g., adenocarcinoma and signet ring-cell carcinoma), as well as diagnosis and prognosis of mixed adenoneuroendocrine carcinoma (MANEC) and pure adenocarcinoma, is poorly understood. Moreover, the impact of the SRIF system signaling on CRC cell proliferation and its potential role in the progression of this cancer remains unknown. Therefore, this review summarizes the recent collective knowledge and understanding of the clinical significance of the SRIF system signaling in CRC, aiming to evaluate the potential role of its components in CRC histogenesis, diagnosis, and potential therapy.
Collapse
|
5
|
Zheng Y, Karnoub AE. Endocrine regulation of cancer stem cell compartments in breast tumors. Mol Cell Endocrinol 2021; 535:111374. [PMID: 34242715 DOI: 10.1016/j.mce.2021.111374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 05/28/2021] [Accepted: 06/01/2021] [Indexed: 10/20/2022]
Abstract
Cancer cells within breast tumors exist within a hierarchy in which only a small and rare subset of cells is able to regenerate growths with the heterogeneity of the original tumor. These highly malignant cancer cells, which behave like stem cells for new cancers and are called "cancer stem cells" or CSCs, have also been shown to possess increased resistance to therapeutics, and represent the root cause underlying therapy failures, persistence of residual disease, and relapse. As >90% of cancer deaths are due to refractory tumors, identification of critical molecular drivers of the CSC-state would reveal vulnerabilities that can be leveraged in designing therapeutics that eradicate advanced disease and improve patient survival outcomes. An expanding and complex body of work has now described the exquisite susceptibility of CSC pools to the regulatory influences of local and systemic hormones. Indeed, breast CSCs express a plethora of hormonal receptors, which funnel hormonal influences over every aspect of breast neoplasia - be it tumor onset, growth, survival, invasion, metastasis, or therapy resistance - via directly impacting CSC behavior. This article is intended to shed light on this active area of investigation by attempting to provide a systematic and comprehensive overview of the available evidence directly linking hormones to breast CSC biology.
Collapse
Affiliation(s)
- Yurong Zheng
- Department of Pathology and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Antoine E Karnoub
- Department of Pathology and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA; Harvard Stem Cell Institute, Cambridge, MA, 02138, USA; Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA.
| |
Collapse
|
6
|
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: 5] [Impact Index Per Article: 1.7] [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
|
7
|
Costanzi E, Simioni C, Conti I, Laface I, Varano G, Brenna C, Neri LM. Two neuroendocrine G protein-coupled receptor molecules, somatostatin and melatonin: Physiology of signal transduction and therapeutic perspectives. J Cell Physiol 2020; 236:2505-2518. [PMID: 32989768 DOI: 10.1002/jcp.30062] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/04/2020] [Accepted: 09/10/2020] [Indexed: 12/15/2022]
Abstract
Recent studies have shown that G protein-coupled receptors (GPCRs), the largest signal-conveying receptor family, are targets for mutations occurring frequently in different cancer types. GPCR alterations associated with cancer development represent significant challenges for the discovery and the advancement of targeted therapeutics. Among the different molecules that can activate GPCRs, we focused on two molecules that exert their biological actions regulating many typical features of tumorigenesis such as cellular proliferation, survival, and invasion: somatostatin and melatonin. The modulation of signaling pathways, that involves these two molecules, opens an interesting scenario for cancer therapy, with the opportunity to act at different molecular levels. Therefore, the aim of this review is the analysis of the biological activity and the therapeutic potential of somatostatin and melatonin, displaying a high affinity for GPCRs, that interfere with cancer development and maintenance.
Collapse
Affiliation(s)
- Eva Costanzi
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Carolina Simioni
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy.,Laboratory for Technologies of Advanced Therapies (LTTA)-Electron Microscopy Center, University of Ferrara, Ferrara, Italy
| | - Ilaria Conti
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Ilaria Laface
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Gabriele Varano
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Cinzia Brenna
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Luca M Neri
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy.,Laboratory for Technologies of Advanced Therapies (LTTA)-Electron Microscopy Center, University of Ferrara, Ferrara, Italy
| |
Collapse
|
8
|
Shimoyama R, Hijioka S, Mizuno N, Ogawa G, Kataoka T, Katayama H, Machida N, Honma Y, Boku N, Hamaguchi T, Fukuda H, Terashima M, Kanemitsu Y, Furuse J. Study protocol for a multi-institutional randomized phase III study comparing combined everolimus plus lanreotide therapy and everolimus monotherapy in patients with unresectable or recurrent gastroenteropancreatic neuroendocrine tumors; Japan Clinical Oncology Group Study JCOG1901 (STARTER-NET study). Pancreatology 2020; 20:1183-1188. [PMID: 32798144 DOI: 10.1016/j.pan.2020.07.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/24/2020] [Accepted: 07/17/2020] [Indexed: 12/11/2022]
Abstract
Everolimus is recognized as one of the standard drugs for the treatment of unresectable or recurrent gastroenteropancreatic neuroendocrine tumors (NET). However, recent evidence has suggested that addition of somatostatin analogs to everolimus may yield better survival outcomes as compared to everolimus alone. In April 2020, we have initiated a randomized phase III trial in Japan, to confirm the superiority of combined everolimus plus lanreotide therapy over everolimus monotherapy in patients with unresectable or recurrent gastroenteropancreatic NETs with poor prognostic factors (Ki-67 labeling index: LI 5%-20% or Ki-67 LI < 5% with diffuse liver metastases). We plan to enroll a total of 250 patients from 76 institutions over an accrual period of 5 years. The primary endpoint is progression-free survival. The key secondary endpoint is overall survival, with response rate, disease control rate, and proportion of patients with adverse events as the other secondary endpoints. This trial is registered with the Japan Registry of Clinical Trials as jRCT1031200023 [https://jrct.niph.go.jp/en-latest-detail/jRCT1031200023].
Collapse
Affiliation(s)
- Ryo Shimoyama
- JCOG Data Center/Operations Office, National Cancer Center Hospital, Tokyo, Japan
| | - Susumu Hijioka
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo, Japan.
| | - Nobumasa Mizuno
- Department of Gastroenterology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Gakuto Ogawa
- JCOG Data Center/Operations Office, National Cancer Center Hospital, Tokyo, Japan
| | - Tomoko Kataoka
- JCOG Data Center/Operations Office, National Cancer Center Hospital, Tokyo, Japan
| | - Hiroshi Katayama
- JCOG Data Center/Operations Office, National Cancer Center Hospital, Tokyo, Japan
| | - Nozomu Machida
- Division of Gastrointestinal Oncology, Shizuoka Cancer Center, Shizuoka, Japan
| | - Yoshitaka Honma
- Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, Tokyo, Japan
| | - Narikazu Boku
- Department of Gastrointestinal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Tetsuya Hamaguchi
- Department of Gastroenterological Oncology, Saitama Medical University International Medical Center, Saitama, Japan
| | - Haruhiko Fukuda
- JCOG Data Center/Operations Office, National Cancer Center Hospital, Tokyo, Japan
| | | | - Yukihide Kanemitsu
- Department of Colorectal Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Junji Furuse
- Department of Medical Oncology, Kyorin University, Tokyo, Japan
| | | |
Collapse
|
9
|
Loos JA, Negro P, Cumino AC. In vitro anti-echinococcal activity of octreotide: Additive effect of metformin linked to autophagy. Acta Trop 2020; 203:105312. [PMID: 31870710 DOI: 10.1016/j.actatropica.2019.105312] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 12/07/2019] [Accepted: 12/12/2019] [Indexed: 01/28/2023]
Abstract
Cystic echinococcosis (CE) is a worldwide zoonosis caused by the Echinococcus granulosus larval stage. The currently available therapy for this disease is based on benzimidazoles, which are rarely curative and cause several adverse effects. Therefore, new treatment options are needed. Octreotide (Oct) is a somatostatin analogue which exhibits anti-proliferative and anti-secretory effects over several cancer cell lines expressing somatostatin receptors. Here, we assessed the in vitro pharmacological effect of Oct against the E. granulosus larval stage. The drug caused a significant dose-dependent decrease in the viability of both protoscoleces and metacestodes. SEM and TEM analysis showed ultrastructural damage in both larval forms under drug treatment. Based on this, we investigated the possible presence of an Oct binding receptor in the parasite. The putative somatostatin/allatostatin-like receptor (Eg-s/ast) conserves the characteristic topology and signature sequences of the prototype somatostatin receptor common to vertebrates and is expressed in both metacestodes and protoscoleces. Moreover, Oct treated-parasites showed the presence of autophagic structures and a significant increase in transcriptional expression of autophagy key genes such as Eg-atg6, Eg-atg8, Eg-atg12 and Eg-atg16. In addition, by in toto immunolocalization assays, an increase in the punctate pattern and Eg-Atg8 protein expression was detected in Oct-treated metacestodes. Subsequently, the combination of Oct and Met had an additive effect on the viability of both larval forms. Our results provide additional evidence for the participation of PI3K/AKT/TOR/autophagy pathway in the Echinococcus survival and suggest the concomitant use of these drugs as potential therapeutic agents in treating of CE.
Collapse
|
10
|
Biological and Biochemical Basis of the Differential Efficacy of First and Second Generation Somatostatin Receptor Ligands in Neuroendocrine Neoplasms. Int J Mol Sci 2019; 20:ijms20163940. [PMID: 31412614 PMCID: PMC6720449 DOI: 10.3390/ijms20163940] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/1970] [Revised: 08/05/2019] [Accepted: 08/08/2019] [Indexed: 02/07/2023] Open
Abstract
Endogenous somatostatin shows anti-secretory effects in both physiological and pathological settings, as well as inhibitory activity on cell growth. Since somatostatin is not suitable for clinical practice, researchers developed synthetic somatostatin receptor ligands (SRLs) to overcome this limitation. Currently, SRLs represent pivotal tools in the treatment algorithm of neuroendocrine tumors (NETs). Octreotide and lanreotide are the first-generation SRLs developed and show a preferential binding affinity to somatostatin receptor (SST) subtype 2, while pasireotide, which is a second-generation SRL, has high affinity for multiple SSTs (SST5 > SST2 > SST3 > SST1). A number of studies demonstrated that first-generation and second-generation SRLs show distinct functional properties, besides the mere receptor affinity. Therefore, the aim of the present review is to critically review the current evidence on the biological effects of SRLs in pituitary adenomas and neuroendocrine tumors, by mainly focusing on the differences between first-generation and second-generation ligands.
Collapse
|
11
|
Stueven AK, Kayser A, Wetz C, Amthauer H, Wree A, Tacke F, Wiedenmann B, Roderburg C, Jann H. Somatostatin Analogues in the Treatment of Neuroendocrine Tumors: Past, Present and Future. Int J Mol Sci 2019; 20:ijms20123049. [PMID: 31234481 PMCID: PMC6627451 DOI: 10.3390/ijms20123049] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 06/06/2019] [Accepted: 06/19/2019] [Indexed: 12/14/2022] Open
Abstract
In recent decades, the incidence of neuroendocrine tumors (NETs) has steadily increased. Due to the slow-growing nature of these tumors and the lack of early symptoms, most cases are diagnosed at advanced stages, when curative treatment options are no longer available. Prognosis and survival of patients with NETs are determined by the location of the primary lesion, biochemical functional status, differentiation, initial staging, and response to treatment. Somatostatin analogue (SSA) therapy has been a mainstay of antisecretory therapy in functioning neuroendocrine tumors, which cause various clinical symptoms depending on hormonal hypersecretion. Beyond symptomatic management, recent research demonstrates that SSAs exert antiproliferative effects and inhibit tumor growth via the somatostatin receptor 2 (SSTR2). Both the PROMID (placebo-controlled, prospective, randomized study in patients with metastatic neuroendocrine midgut tumors) and the CLARINET (controlled study of lanreotide antiproliferative response in neuroendocrine tumors) trial showed a statistically significant prolongation of time to progression/progression-free survival (TTP/PFS) upon SSA treatment, compared to placebo. Moreover, the combination of SSA with peptide receptor radionuclide therapy (PRRT) in small intestinal NETs has proven efficacy in the phase 3 neuroendocrine tumours therapy (NETTER 1) trial. PRRT is currently being tested for enteropancreatic NETs versus everolimus in the COMPETE trial, and the potential of SSTR-antagonists in PRRT is now being evaluated in early phase I/II clinical trials. This review provides a synopsis on the pharmacological development of SSAs and their use as antisecretory drugs. Moreover, this review highlights the clinical evidence of SSAs in monotherapy, and in combination with other treatment modalities, as applied to the antiproliferative management of neuroendocrine tumors with special attention to recent high-quality phase III trials.
Collapse
Affiliation(s)
- Anna Kathrin Stueven
- Charité, Campus Virchow Klinikum and Charité, Campus Mitte, Department of Hepatology and Gastroenterology, Universitätsmedizin Berlin, 10117 Berlin, Germany.
| | - Antonin Kayser
- Charité, Campus Virchow Klinikum and Charité, Campus Mitte, Department of Hepatology and Gastroenterology, Universitätsmedizin Berlin, 10117 Berlin, Germany.
| | - Christoph Wetz
- Charité, Campus Virchow Klinikum and Charité, Campus Mitte, Department of Nuclear Medicine, Universitätsmedizin Berlin, 10117 Berlin, Germany.
| | - Holger Amthauer
- Charité, Campus Virchow Klinikum and Charité, Campus Mitte, Department of Nuclear Medicine, Universitätsmedizin Berlin, 10117 Berlin, Germany.
| | - Alexander Wree
- Charité, Campus Virchow Klinikum and Charité, Campus Mitte, Department of Hepatology and Gastroenterology, Universitätsmedizin Berlin, 10117 Berlin, Germany.
| | - Frank Tacke
- Charité, Campus Virchow Klinikum and Charité, Campus Mitte, Department of Hepatology and Gastroenterology, Universitätsmedizin Berlin, 10117 Berlin, Germany.
| | - Bertram Wiedenmann
- Charité, Campus Virchow Klinikum and Charité, Campus Mitte, Department of Hepatology and Gastroenterology, Universitätsmedizin Berlin, 10117 Berlin, Germany.
| | - Christoph Roderburg
- Charité, Campus Virchow Klinikum and Charité, Campus Mitte, Department of Hepatology and Gastroenterology, Universitätsmedizin Berlin, 10117 Berlin, Germany.
| | - Henning Jann
- Charité, Campus Virchow Klinikum and Charité, Campus Mitte, Department of Hepatology and Gastroenterology, Universitätsmedizin Berlin, 10117 Berlin, Germany.
| |
Collapse
|
12
|
Pasireotide protects mammalian cochlear hair cells from gentamicin ototoxicity by activating the PI3K-Akt pathway. Cell Death Dis 2019; 10:110. [PMID: 30728348 PMCID: PMC6365508 DOI: 10.1038/s41419-019-1386-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 12/18/2018] [Accepted: 01/15/2019] [Indexed: 01/07/2023]
Abstract
Gentamicin is a widely used antibiotic for the treatment of gram-negative bacterial infections; however, its use often results in significant and permanent hearing loss. Hearing loss resulting from hair cell (HC) degeneration affects millions of people worldwide, and one major cause is the loss of sensory HCs in the inner ear due to aminoglycoside exposure. Strategies to overcome the apparently irreversible loss of HCs in mammals are crucial for hearing protection. Here, we report that the somatostatin analog pasireotide protects mouse cochlear HCs from gentamicin damage using a well-established in vitro gentamicin-induced HC loss model and that the otoprotective effects of pasireotide are due to Akt up-regulation via the PI3K–Akt signal pathway activation. We demonstrate active caspase signal in organ of Corti (OC) explants exposed to gentamicin and show that pasireotide treatment activates survival genes, reduces caspase signal, and increases HC survival. The neuropeptide somatostatin and its selective analogs have provided neuroprotection by activating five somatostatin receptor (SSTR1–SSTR5) subtypes. Pasireotide has a high affinity for SSTR2 and SSTR5, and the addition of SSTR2- and SSTR5-specific antagonists leads to a loss of protection. The otoprotective effects of pasireotide were also observed in a gentamicin-injured animal model. In vivo studies have shown that 13 days of subcutaneous pasireotide application prevents gentamicin-induced HC death and permanent hearing loss in mice. Auditory brainstem response analysis confirmed the protective effect of pasireotide, and we found a significant threshold shift at all measured frequencies (4, 8, 16, 24, and 32 kHz). Together, these findings indicate that pasireotide is a novel otoprotective peptide acting via the PI3K–Akt pathway and may be of therapeutic value for HC protection from ototoxic insults.
Collapse
|
13
|
Expression and selective activation of somatostatin receptor subtypes induces cell cycle arrest in cancer cells. Oncol Lett 2018; 17:1723-1731. [PMID: 30675231 PMCID: PMC6341781 DOI: 10.3892/ol.2018.9773] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 10/18/2018] [Indexed: 12/12/2022] Open
Abstract
Somatostatin receptors (SSTRs) are G-protein-coupled plasma membrane receptors that have been determined to be expressed in normal and cancer tissues. Activation of SSTRs frequently results in inhibition of cell proliferation and therefore somatostatin analogues (SSAs) have been used in cancer treatment. However, the variable outcomes of SSA treatment were considered to be the consequences of loss-of-expression of SSTRs and/or subtype-specific effects. In the present study, the patterns of SSTR expression in 160 breast cancer tissues were investigated, and the mechanisms of SSTR activation and the influence on cell proliferation were further characterized. The expression levels of SSTR1-5 were determined using immunohistology. Hemagglutinin-SSTR1 and MYC-SSTR4 were transiently overexpressed in MDA-MB-435S cells, and the potential receptor dimerization was determined using immunofluorescence and co-immunoprecipitation. The influence of SSTR1 and SSTR4 expression/activation on cell proliferation was monitored using flow cytometry. The results demonstrated that all five SSTR subtypes were expressed at variable levels in tumor tissues, with the highest positive expression instance being determined for SSTR1 and SSTR4, with positive expression levels in 90.0 and 71.3% of tumor tissues, respectively. Immunofluorescence and co-immunoprecipitation revealed SSTR1/SSTR4 heterodimerization, which was increased in response to receptor activation using the subtype-specific SSA L-803087. The translocation of SSTR1/SSTR4 dimers into the cytoplasm upon receptor activation was also observed. Additionally, it was identified using flow cytometry that co-expression and activation of SSTR1 and SSTR4 in MDA-MB-435S cells resulted in a decreased proportion of S-phase cells. The results of the present study revealed that SSTR1 and SSTR4 are the most frequently expressed SSTR subtypes in breast cancer, and that the cell cycle arrest was mediated by SSTR1/SSTR4 dimerization/activation.
Collapse
|
14
|
Lehman JM, Hoeksema MD, Staub J, Qian J, Harris B, Callison JC, Miao J, Shi C, Eisenberg R, Chen H, Chen SC, Massion PP. Somatostatin receptor 2 signaling promotes growth and tumor survival in small-cell lung cancer. Int J Cancer 2018; 144:1104-1114. [PMID: 30152518 DOI: 10.1002/ijc.31771] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 06/18/2018] [Accepted: 06/25/2018] [Indexed: 12/12/2022]
Abstract
Somatostatin receptor 2 (SSTR2) is overexpressed in a majority of neuroendocrine neoplasms, including small-cell lung carcinomas (SCLCs). SSTR2 was previously considered an inhibitory receptor on cell growth, but its agonists had poor clinical responses in multiple clinical trials. The role of this receptor as a potential therapeutic target in lung cancer merits further investigation. We evaluated the expression of SSTR2 in a cohort of 96 primary tumors from patients with SCLC and found 48% expressed SSTR2. Correlation analysis in both CCLE and an SCLC RNAseq cohort confirmed high-level expression and identified an association between NEUROD1 and SSTR2. There was a significant association with SSTR2 expression profile and poor clinical outcome. We tested whether SSTR2 expression might contribute to tumor progression through activation of downstream signaling pathways, using in vitro and in vivo systems and downregulated SSTR2 expression in lung cancer cells by shRNA. SSTR2 downregulation led to increased apoptosis and dramatically decreased tumor growth in vitro and in vivo in multiple cell lines with decreased AMPKα phosphorylation and increased oxidative metabolism. These results demonstrate a role for SSTR2 signaling in SCLC and suggest that SSTR2 is a poor prognostic biomarker in SCLC and potential future therapeutic signaling target.
Collapse
Affiliation(s)
- Jonathan M Lehman
- Division of Medical Oncology, Vanderbilt Ingram Cancer Center, Nashville, TN.,Cancer Early Detection and Prevention Initiative, Vanderbilt Ingram Cancer Center, Nashville, TN.,Veterans Affairs, Tennessee Valley Healthcare System, Nashville Campus, Nashville, TN
| | - Megan D Hoeksema
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt Ingram Cancer Center, Nashville, TN.,Cancer Early Detection and Prevention Initiative, Vanderbilt Ingram Cancer Center, Nashville, TN
| | - Jeremy Staub
- Division of Medical Oncology, Vanderbilt Ingram Cancer Center, Nashville, TN
| | - Jun Qian
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt Ingram Cancer Center, Nashville, TN.,Cancer Early Detection and Prevention Initiative, Vanderbilt Ingram Cancer Center, Nashville, TN
| | - Bradford Harris
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt Ingram Cancer Center, Nashville, TN
| | - J Clay Callison
- University of Tennessee Graduate School of Medicine, Tennessee Valley Healthcare System, Nashville Campus, Nashville, TN
| | - Jennifer Miao
- Vanderbilt University School of Medicine, Tennessee Valley Healthcare System, Nashville Campus, Nashville, TN
| | - Chanjuan Shi
- Department of Pathology, Microbiology and Immunology, Tennessee Valley Healthcare System, Nashville Campus, Nashville, TN
| | - Rosana Eisenberg
- Department of Pathology, Microbiology and Immunology, Tennessee Valley Healthcare System, Nashville Campus, Nashville, TN
| | - Heidi Chen
- Vanderbilt University Department of Biostatistics, Tennessee Valley Healthcare System, Nashville Campus, Nashville, TN
| | - Sheau-Chiann Chen
- Vanderbilt University Department of Biostatistics, Tennessee Valley Healthcare System, Nashville Campus, Nashville, TN
| | - Pierre P Massion
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt Ingram Cancer Center, Nashville, TN.,Cancer Early Detection and Prevention Initiative, Vanderbilt Ingram Cancer Center, Nashville, TN.,Veterans Affairs, Tennessee Valley Healthcare System, Nashville Campus, Nashville, TN
| |
Collapse
|
15
|
Roof AK, Gutierrez-Hartmann A. Consider the context: Ras/ERK and PI3K/AKT/mTOR signaling outcomes are pituitary cell type-specific. Mol Cell Endocrinol 2018; 463:87-96. [PMID: 28445712 DOI: 10.1016/j.mce.2017.04.019] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 04/20/2017] [Accepted: 04/21/2017] [Indexed: 12/11/2022]
Abstract
Conserved signaling pathways are critical regulators of pituitary homeostasis and, when dysregulated, contribute to adenoma formation. Pituitary adenomas are typically benign and rarely progress to malignant cancer. Pituitary and other neuroendocrine cell types often display non-proliferative responses to ERK and PI3K, in contrast to non-endocrine cell types which typically proliferate in response to ERK and PI3K activation. These differences likely contribute to the infrequent progression to malignancy in many endocrine tumors. In this review, we highlight the Ras/ERK and PI3K/AKT/mTOR signaling pathways in each pituitary cell type, as well as in other endocrine tissues. Furthermore, we provide evidence that a balance of ERK and PI3K signaling is required to maintain pituitary homeostasis. It is unlikely that one sole oncogene will be identified as being responsible for sporadic pituitary adenoma formation. This review emphasizes the necessity to consider endocrine cell-specific contexts and the interplay of signaling pathways to define the mechanisms underlying pituitary tumorigenesis.
Collapse
Affiliation(s)
- Allyson K Roof
- Program in Integrated Physiology and Reproductive Sciences, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, United States
| | - Arthur Gutierrez-Hartmann
- Program in Integrated Physiology and Reproductive Sciences, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, United States; Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, United States; Department of Biochemistry and Molecular Genetics, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, United States.
| |
Collapse
|
16
|
Graillon T, Romano D, Defilles C, Lisbonis C, Saveanu A, Figarella-Branger D, Roche PH, Fuentes S, Chinot O, Dufour H, Barlier A. Pasireotide is more effective than octreotide, alone or combined with everolimus on human meningioma in vitro. Oncotarget 2017; 8:55361-55373. [PMID: 28903425 PMCID: PMC5589664 DOI: 10.18632/oncotarget.19517] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 07/12/2017] [Indexed: 12/18/2022] Open
Abstract
Pasireotide is a somatostatin analog (SSA) that targets somatostatin receptor subtype 1 (SST1), SST2, SST3, and SST5 with a high affinity. Pasireotide has a better antisecretory effect in acromegaly, Cushing's disease, and neuroendocrine tumors than octreotide. In this study, we compared the effects of pasireotide to those of octreotide in vitro on meningioma primary cell cultures, both alone and in combination with the mTOR inhibitor everolimus. Significant mRNA expression levels of SST1, SST2, and SST5 were observed in 40.5%, 100%, and 35% of meningioma samples, respectively. Pasireotide had a significantly stronger inhibitory effect on cell proliferation than octreotide. The effect of pasireotide, but not of octreotide, was significantly stronger in the group expressing the highest level of SST1 mRNA. Combined treatment with pasireotide and everolimus induced a higher reduction in cell viability than that with octreotide plus everolimus. Moreover, pasireotide decreased Akt phosphorylation and reversed everolimus-induced Akt hyperphosphorylation to a higher degree than octreotide. Using 4E-BP1 siRNA (si4E-BP), we demonstrated that 4E-BP1 protein silencing significantly reversed the response to everolimus, both alone and in combination with SSAs. Moreover, si4E-BP completely reversed the inhibition of cyclin D1 expression level and the increase in p27kip1 induced by SSAs, both alone and in combination with everolimus. Our results strongly support the need for further studies on the combination of pasireotide and everolimus in medical therapy for meningiomas.
Collapse
Affiliation(s)
- Thomas Graillon
- Aix Marseille Univ, CNRS CRN2M UMR7286, Marseille, France.,APHM, La Timone Hospital, Department of Neurosurgery, Marseille, France
| | - David Romano
- Aix Marseille Univ, CNRS CRN2M UMR7286, Marseille, France
| | | | | | - Alexandru Saveanu
- Aix Marseille Univ, CNRS CRN2M UMR7286, Marseille, France.,APHM, La Conception Hospital, Molecular Biology Laboratory, Marseille, France
| | - Dominique Figarella-Branger
- APHM, La Timone Hospital, Department of Anatomopathology and Neuropathology, Marseille, France.,Aix Marseille Univ, INSERM, CRO2 UMR911, Marseille, France
| | | | - Stéphane Fuentes
- APHM, La Timone Hospital, Department of Neurosurgery, Marseille, France
| | - Olivier Chinot
- Aix Marseille Univ, INSERM, CRO2 UMR911, Marseille, France.,APHM, La Timone Hospital, Department of Neuro-oncology, Marseille, France
| | - Henry Dufour
- Aix Marseille Univ, CNRS CRN2M UMR7286, Marseille, France.,APHM, La Timone Hospital, Department of Neurosurgery, Marseille, France
| | - Anne Barlier
- Aix Marseille Univ, CNRS CRN2M UMR7286, Marseille, France.,APHM, La Conception Hospital, Molecular Biology Laboratory, Marseille, France
| |
Collapse
|
17
|
Würth R, Barbieri F, Pattarozzi A, Gaudenzi G, Gatto F, Fiaschi P, Ravetti JL, Zona G, Daga A, Persani L, Ferone D, Vitale G, Florio T. Phenotypical and Pharmacological Characterization of Stem-Like Cells in Human Pituitary Adenomas. Mol Neurobiol 2016; 54:4879-4895. [PMID: 27514754 DOI: 10.1007/s12035-016-0025-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 08/01/2016] [Indexed: 12/11/2022]
Abstract
The presence and functional role of tumor stem cells in benign tumors, and in human pituitary adenomas in particular, is a debated issue that still lacks a definitive formal demonstration. Fifty-six surgical specimens of human pituitary adenomas were processed to establish tumor stem-like cultures by selection and expansion in stem cell-permissive medium or isolating CD133-expressing cells. Phenotypic and functional characterization of these cells was performed (1) ex vivo, by immunohistochemistry analysis on paraffin-embedded tissues; (2) in vitro, attesting marker expression, proliferation, self-renewal, differentiation, and drug sensitivity; and (3) in vivo, using a zebrafish model. Within pituitary adenomas, we identified rare cell populations expressing stem cell markers but not pituitary hormones; we isolated and expanded in vitro these cells, obtaining fibroblast-free, stem-like cultures from 38 pituitary adenoma samples. These cells grow as spheroids, express stem cell markers (Oct4, Sox2, CD133, and nestin), show sustained in vitro proliferation as compared to primary cultures of differentiated pituitary adenoma cells, and are able to differentiate in hormone-expressing pituitary cells. Besides, pituisphere cells, apparently not tumorigenic in mice, engrafted in zebrafish embryos, inducing pro-angiogenic and invasive responses. Finally, pituitary adenoma stem-like cells express regulatory pituitary receptors (D2R, SSTR2, and SSTR5), whose activation by a dopamine/somatostatin chimeric agonist exerts antiproliferative effects. In conclusion, we provide evidence that human pituitary adenomas contain a subpopulation fulfilling biological and phenotypical signatures of tumor stem cells that may represent novel therapeutic targets for therapy-resistant tumors.
Collapse
Affiliation(s)
- Roberto Würth
- Pharmacology Section, Department of Internal Medicine, University of Genova, Viale Benedetto XV, 2, 16132, Genoa, Italy
| | - Federica Barbieri
- Pharmacology Section, Department of Internal Medicine, University of Genova, Viale Benedetto XV, 2, 16132, Genoa, Italy
- Centre of Excellence for Biomedical Research (CEBR), University of Genova, Genoa, Italy
| | - Alessandra Pattarozzi
- Pharmacology Section, Department of Internal Medicine, University of Genova, Viale Benedetto XV, 2, 16132, Genoa, Italy
| | - Germano Gaudenzi
- Department of Clinical Sciences and Community Health, University of Milano, Milan, Italy
| | - Federico Gatto
- Pharmacology Section, Department of Internal Medicine, University of Genova, Viale Benedetto XV, 2, 16132, Genoa, Italy
- Centre of Excellence for Biomedical Research (CEBR), University of Genova, Genoa, Italy
| | - Pietro Fiaschi
- Department of Neurosciences, University of Genova, Genoa, Italy
- Department of Neurosurgery, IRCCS-AOU San Martino-IST, Genoa, Italy
| | | | - Gianluigi Zona
- Department of Neurosciences, University of Genova, Genoa, Italy
- Department of Neurosurgery, IRCCS-AOU San Martino-IST, Genoa, Italy
| | - Antonio Daga
- Laboratory of Gene Transfer, IRCCS-AOU San Martino-IST, Genoa, Italy
| | - Luca Persani
- Department of Clinical Sciences and Community Health, University of Milano, Milan, Italy
- Endocrine and Metabolic Research Laboratory, Istituto Auxologico Italiano-IRCCS, Milan, Italy
| | - Diego Ferone
- Pharmacology Section, Department of Internal Medicine, University of Genova, Viale Benedetto XV, 2, 16132, Genoa, Italy
- Centre of Excellence for Biomedical Research (CEBR), University of Genova, Genoa, Italy
| | - Giovanni Vitale
- Department of Clinical Sciences and Community Health, University of Milano, Milan, Italy
- Endocrine and Metabolic Research Laboratory, Istituto Auxologico Italiano-IRCCS, Milan, Italy
| | - Tullio Florio
- Pharmacology Section, Department of Internal Medicine, University of Genova, Viale Benedetto XV, 2, 16132, Genoa, Italy.
- Centre of Excellence for Biomedical Research (CEBR), University of Genova, Genoa, Italy.
| |
Collapse
|
18
|
Berardi R, Morgese F, Torniai M, Savini A, Partelli S, Rinaldi S, Caramanti M, Ferrini C, Falconi M, Cascinu S. Medical treatment for gastro-entero-pancreatic neuroendocrine tumours. World J Gastrointest Oncol 2016; 8:389-401. [PMID: 27096034 PMCID: PMC4824717 DOI: 10.4251/wjgo.v8.i4.389] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2015] [Accepted: 02/16/2016] [Indexed: 02/05/2023] Open
Abstract
Gastro-entero-pancreatic neuroendocrine neoplasms (GEP-NENs) represents a various family of rare tumours. Surgery is the first choice in GEP-NENs patients with localized disease whilst in the metastatic setting many other treatment options are available. Somatostatin analogues are indicated for symptoms control in functioning tumours. Furthermore they may be effective to inhibit tumour progression. GEP-NENs pathogenesis has been extensively studied in the last years therefore several driver mutations pathway genes have been identified as crucial factors in their tumourigenesis. GEP-NENs can over-express vascular endothelial growth factor (VEGF), basic-fibroblastic growth factor, transforming growth factor (TGF-α and -β), platelet derived growth factor (PDGF), insulin-like growth factor-1 (IGF-1) and their receptors PDGF receptor, IGF-1 receptor, epidermal growth factor receptor, VEGF receptor, and c-kit (stem cell factor receptor) that can be considered as potential targets. The availability of new targeted agents, such as everolimus and sunitinib that are effective in advanced and metastatic pancreatic neuroendocrine tumours, has provided new treatment opportunities. Many trials combing new drugs are ongoing.
Collapse
|
19
|
Sampedro-Núñez M, Luque RM, Ramos-Levi AM, Gahete MD, Serrano-Somavilla A, Villa-Osaba A, Adrados M, Ibáñez-Costa A, Martín-Pérez E, Culler MD, Marazuela M, Castaño JP. Presence of sst5TMD4, a truncated splice variant of the somatostatin receptor subtype 5, is associated to features of increased aggressiveness in pancreatic neuroendocrine tumors. Oncotarget 2016; 7:6593-608. [PMID: 26673010 PMCID: PMC4872735 DOI: 10.18632/oncotarget.6565] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 11/21/2015] [Indexed: 12/13/2022] Open
Abstract
Purpose Gastroenteropancreatic neuroendocrine tumors (GEP-NETs) are rare and heterogeneous tumors, and their biological behavior is not well known. We studied the presence and potential functional roles of somatostatin receptors (sst1-5), focusing particularly on the truncated variants (sst5TMD4, sst5TMD5) and on their relationships with the angiogenic system (Ang/Tie-2 and VEGF) in human GEP-NETs. Experimental Design We evaluated 42 tumor tissue samples (26 primary/16 metastatic) from 26 patients with GEP-NETs, and 30 non-tumoral tissues (26 from adjacent non-tumor regions and 4 from normal controls) from a single center. Expression of sst1-5, sst5TMD4, sst5TMD5, Ang1-2, Tie-2 and VEGF was analyzed using real-time qPCR, immunofluorescence and immunohistochemistry. Expression levels were associated with tumor characteristics and clinical outcomes. Functional role of sst5TMD4 was analyzed in GEP-NET cell lines. Results sst1 exhibited the highest expression in GEP-NET, whilst sst2 was the most frequently observed sst-subtype (90.2%). Expression levels of sst1, sst2, sst3, sst5TMD4, and sst5TMD5 were significantly higher in tumor tissues compared to their adjacent non-tumoral tissue. Lymph-node metastases expressed higher levels of sst5TMD4 than in its corresponding primary tumor tissue. sst5TMD4 was also significantly higher in intestinal tumor tissues from patients with residual disease of intestinal origin compared to those with non-residual disease. Functional assays demonstrated that the presence of sst5TMD4 was associated to enhanced malignant features in GEP-NET cells. Angiogenic markers correlated positively with sst5TMD4, which was confirmed by immunohistochemical/fluorescence studies. Conclusions sst5TMD4 is overexpressed in GEP-NETs and is associated to enhanced aggressiveness, suggesting its potential value as biomarker and target in GEP-NETs.
Collapse
Affiliation(s)
- Miguel Sampedro-Núñez
- Department of Endocrinology and Nutrition, Hospital Universitario de la Princesa, Instituto de Investigación Princesa, Universidad Autónoma de Madrid, Madrid 28006, Spain
| | - Raúl M Luque
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofia, and CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba 14014, Spain
| | - Ana M Ramos-Levi
- Department of Endocrinology and Nutrition, Hospital Universitario de la Princesa, Instituto de Investigación Princesa, Universidad Autónoma de Madrid, Madrid 28006, Spain
| | - Manuel D Gahete
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofia, and CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba 14014, Spain
| | - Ana Serrano-Somavilla
- Department of Endocrinology and Nutrition, Hospital Universitario de la Princesa, Instituto de Investigación Princesa, Universidad Autónoma de Madrid, Madrid 28006, Spain
| | - Alicia Villa-Osaba
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofia, and CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba 14014, Spain
| | - Magdalena Adrados
- Department of Endocrinology and Nutrition, Hospital Universitario de la Princesa, Instituto de Investigación Princesa, Universidad Autónoma de Madrid, Madrid 28006, Spain
| | - Alejandro Ibáñez-Costa
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofia, and CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba 14014, Spain
| | - Elena Martín-Pérez
- Department of Endocrinology and Nutrition, Hospital Universitario de la Princesa, Instituto de Investigación Princesa, Universidad Autónoma de Madrid, Madrid 28006, Spain
| | | | - Mónica Marazuela
- Department of Endocrinology and Nutrition, Hospital Universitario de la Princesa, Instituto de Investigación Princesa, Universidad Autónoma de Madrid, Madrid 28006, Spain
| | - Justo P Castaño
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofia, and CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba 14014, Spain
| |
Collapse
|
20
|
Abstract
The somatostatin (SRIF) system, which includes the SRIF ligand and receptors, regulates anterior pituitary gland function, mainly inhibiting hormone secretion and to some extent pituitary tumor cell growth. SRIF-14 via its cognate G-protein-coupled receptors (subtypes 1-5) activates multiple cellular signaling pathways including adenylate cyclase/cAMP, MAPK, ion channel-dependent pathways, and others. In addition, recent data have suggested SRIF-independent constitutive SRIF receptor activity responsible for GH and ACTH inhibition in vitro. This review summarizes current knowledge on ligand-dependent and independent SRIF receptor molecular and functional effects on hormone-secreting cells in the anterior pituitary gland.
Collapse
Affiliation(s)
- Tamar Eigler
- Division of EndocrinologyDiabetes and Metabolism, Department of Medicine, Pituitary Center, Cedars Sinai Medical Center, Davis Building, Room 3066, 8700 Beverly Boulevard, Los Angeles, California 90048, USA
| | - Anat Ben-Shlomo
- Division of EndocrinologyDiabetes and Metabolism, Department of Medicine, Pituitary Center, Cedars Sinai Medical Center, Davis Building, Room 3066, 8700 Beverly Boulevard, Los Angeles, California 90048, USA
| |
Collapse
|
21
|
Bajetta E, Catena L, Fazio N, Pusceddu S, Biondani P, Blanco G, Ricci S, Aieta M, Pucci F, Valente M, Bianco N, Mauri CM, Spada F. Everolimus in combination with octreotide long-acting repeatable in a first-line setting for patients with neuroendocrine tumors: an ITMO group study. Cancer 2014; 120:2457-63. [PMID: 24752410 DOI: 10.1002/cncr.28726] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 02/27/2014] [Accepted: 02/28/2014] [Indexed: 02/06/2023]
Abstract
BACKGROUND Preclinical and clinical studies suggest synergistic activity between somatostatin analogues and mammalian target of rapamycin inhibitors. The activity and safety of everolimus was assessed in combination with octreotide long-acting repeatable (LAR) in patients with neuroendocrine tumors (NETs) of gastroenteropancreatic and lung origin. METHODS This was a phase 2, multicenter trial using a Simon's 2-stage minimax design. Treatment-naive patients with advanced well-differentiated NETs of gastroenteropancreatic tract and lung origin received everolimus 10 mg daily, in combination with octreotide LAR 30 mg every 28 days. The primary endpoint was objective response rate (ORR). RESULTS A total of 50 patients (median age, 60.5 years) were enrolled. Primary tumor sites were: pancreas (14 patients), lung (11 patients), ileum (9 patients), jejunum and duodenum (2 patients), and unknown (14 patients). Thirteen patients (26%) had carcinoid syndrome. Treatment-related adverse events (AEs) were mostly grade 1 or 2; the only grade 4 AE was mucositis in 1 patient, whereas grade 3 AEs included skin rash in 1 case (2%), stomatitis in 4 cases (8%), and diarrhea in 11 cases (22%). The ORR was 18%; 2% of patients had a complete response (CR), 16% a partial response (PR) and 74% achieved stable disease (SD). All CRs and all PRs as well as 92% of SDs had a duration ≥ 6 months. The clinical benefit (CR+PR+SD) was 92%. At a median follow-up of 277 days, median time to progression and overall survival were not reached. CONCLUSIONS The everolimus-octreotide LAR combination was active and well tolerated in these previously treated patients with advanced NETs, suggesting a possible role as first-line treatment in patients with NET.
Collapse
Affiliation(s)
- Emilio Bajetta
- Institute of Oncology, Polyclinic Hospital, Monza, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Yamamoto J, Ohnuma K, Hatano R, Okamoto T, Komiya E, Yamazaki H, Iwata S, Dang NH, Aoe K, Kishimoto T, Yamada T, Morimoto C. Regulation of somatostatin receptor 4-mediated cytostatic effects by CD26 in malignant pleural mesothelioma. Br J Cancer 2014; 110:2232-45. [PMID: 24743707 PMCID: PMC4007235 DOI: 10.1038/bjc.2014.151] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 02/26/2014] [Indexed: 02/07/2023] Open
Abstract
Background: Malignant pleural mesothelioma (MPM) is an aggressive neoplasm arising from mesothelial lining of pleura. CD26 molecules preferentially expressed on epithelioid type of MPM. This study investigates the molecular mechanisms of CD26 regulating MPM cells in vitro and in vivo. Methods: Biochemical and cell biological approaches were used for identifying a novel molecular target of MPM. Its contribution to tumour expansion has been also assessed using animal models. The clinical samples of MPM were also assessed for its expression. Results: We identify that cytostatic effects in MPM are mediated by somatostatin (SST) receptor 4 (SSTR4), being inhibited by the interaction of CD26 molecules. We also indicates that SSTR4-mediated cytostatic effects are regulated by SHP-2 PTP, and that this inhibitory effect by SST agonist is enhanced via lipid raft clustering of associated molecules following crosslinking of anti-CD26 antibody. Finally, using an in vivo xenograft model, we demonstrate that the anti-tumour effect of anti-CD26 mAb is enhanced when combined with SSTR4 agonist treatment, and that SSTR4 is highly coexpressed with CD26 on epithelioid or biphasic types of MPM tissues obtained from patients' surgical specimens. Conclusions: Combination therapy with humanised anti-CD26 mAb and SSTR4 agonist may therefore potentiate anti-tumour effect on MPM.
Collapse
Affiliation(s)
- J Yamamoto
- Department of Therapy Development and Innovation for Immune Disorders and Cancers, Graduate School of Medicine, Juntendo University, 2-1-1, Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - K Ohnuma
- Department of Therapy Development and Innovation for Immune Disorders and Cancers, Graduate School of Medicine, Juntendo University, 2-1-1, Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - R Hatano
- Department of Therapy Development and Innovation for Immune Disorders and Cancers, Graduate School of Medicine, Juntendo University, 2-1-1, Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - T Okamoto
- Department of Therapy Development and Innovation for Immune Disorders and Cancers, Graduate School of Medicine, Juntendo University, 2-1-1, Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - E Komiya
- Department of Therapy Development and Innovation for Immune Disorders and Cancers, Graduate School of Medicine, Juntendo University, 2-1-1, Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - H Yamazaki
- Department of Therapy Development and Innovation for Immune Disorders and Cancers, Graduate School of Medicine, Juntendo University, 2-1-1, Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - S Iwata
- Department of Therapy Development and Innovation for Immune Disorders and Cancers, Graduate School of Medicine, Juntendo University, 2-1-1, Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - N H Dang
- Division of Hematology/Oncology, University of Florida, 1600 SW Archer Road, Box 100278, Room MSB M410A, Gainesville, FL 32610, USA
| | - K Aoe
- Department of Medical Oncology and Clinical Research, National Hospital Organization Yamaguchi-Ube Medical Center, 685 Higashi-Kiwa, Ube, Yamaguchi 755-0241, Japan
| | - T Kishimoto
- Department of Respiratory Medicine, Okayama Rosai Hospital, 1-10-25 Chikkomidorimachi, Okayama Minami-ku, Okayama 702-8055, Japan
| | - T Yamada
- Department of Pathology, Keio University School of Medicine, 35 Shinanomachi Shinjuku-ku, Tokyo 160-8582, Japan
| | - C Morimoto
- Department of Therapy Development and Innovation for Immune Disorders and Cancers, Graduate School of Medicine, Juntendo University, 2-1-1, Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| |
Collapse
|
23
|
Barbieri F, Albertelli M, Grillo F, Mohamed A, Saveanu A, Barlier A, Ferone D, Florio T. Neuroendocrine tumors: insights into innovative therapeutic options and rational development of targeted therapies. Drug Discov Today 2014; 19:458-68. [DOI: 10.1016/j.drudis.2013.10.015] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Revised: 09/02/2013] [Accepted: 10/21/2013] [Indexed: 02/07/2023]
|
24
|
Kunjara S, Greenbaum AL, Sochor M, Flyvbjerg A, Grønbaek H, McLean P. Effects of long-acting somatostatin analogues on redox systems in rat lens in experimental diabetes. Int J Exp Pathol 2014; 95:95-100. [PMID: 24602114 DOI: 10.1111/iep.12069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Accepted: 11/26/2013] [Indexed: 10/25/2022] Open
Abstract
The effects of long-acting somatostatin analogues, angiopeptin (AGP) and Sandostatin (SMS), on the early decline in the lens content of glutathione (GSH), ATP and NADPH and increase in sorbitol were studied in STZ diabetic rats, and comparison was made with the effect of insulin. Three factors prompted this study: (i) the known increase in IGF-1 in ocular tissue in diabetes and antagonistic effect of somatostatins, (ii) the known effect of IGF-1 in increasing lens aldose reductase and (iii) the lack of effect of somatostatins on diabetic hyperglycaemia, the latter enabling a differentiation to be made between effects of hyperglycaemia per se and site(s) of IGF-1/somatostatins. All four metabolites studied showed a significant restoration towards the normal control level after 7 days of treatment with AGP and SMS, and AGP was more effective on levels of GSH and ATP. A significant correlation was found between GSH and ATP across all groups at 7 days treatment. The redox state changes in diabetes include both NADP+/NADPH and NAD+/NADH in the conversion of glucose to sorbitol and via sorbitol dehydrogenase to fructose with a linked decrease in ATP formation via NAD+/NADH regulation of the glycolytic pathway. The interlinked network of change includes the requirement for ATP in the synthesis of GSH. The present study points to possible loci of action of somatostatins in improving metabolic parameters in the diabetic rat lens via effects on aldose reductase and/or glucose transport at GLUT 3.
Collapse
Affiliation(s)
- Sirilaksana Kunjara
- Division of Biosciences, Research Department of Cell and Developmental Biology, University College London, London, UK
| | | | | | | | | | | |
Collapse
|
25
|
Peng Y, Deng L, Ding Y, Chen Q, Wu Y, Yang M, Wang Y, Fu Q. Comparative study of somatostatin-human serum albumin fusion proteins and natural somatostatin on receptor binding, internalization and activation. PLoS One 2014; 9:e89932. [PMID: 24587133 PMCID: PMC3937410 DOI: 10.1371/journal.pone.0089932] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Accepted: 01/25/2014] [Indexed: 01/08/2023] Open
Abstract
Albumin fusion technology, the combination of small molecular proteins or peptides with human serum albumin (HSA), is an effective method for improving the medicinal values of natural small molecular proteins or peptides. However, comparative studies between HSA-fusion proteins or peptides and the parent small molecules in biological and molecular mechanisms are less reported. In this study, we examined the binding property of two novel somatostatin-HSA fusion proteins, (SST14)2-HSA and (SST28)2-HSA, to human SSTRs in stably expressing SSTR1-5 HEK 293 cells; observed the regulation of receptor internalization and internalized receptor recycling; and detected the receptors activation of HSA fusion proteins in stably expressing SSTR2- and SSTR3-EGFP cells. We showed that both somatostatin-HSA fusion proteins had high affinity to all five SSTRs, stimulated the ERK1/2 phosphorylation and persistently inhibited the accumulation of forskolin-stimulated cAMP in SSTR2- and SSTR3-expressing cells; but were less potent than the synthetic somatostatin-14 (SST-14). Our experiments also showed that somatostatin-HSA fusion proteins did not induce the receptors internalization; rather, they accelerated the recycling of the internalized receptors induced by SST-14 to the plasma membrane. Our results indicated that somatostatin-HSA fusion proteins, different from SST-14, exhibit some particular properties in binding, regulating, and activating somatostatin receptors.
Collapse
Affiliation(s)
- Ying Peng
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
| | - Lili Deng
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
| | - Yuedi Ding
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
| | - Quancheng Chen
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
| | - Yu Wu
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
| | - Meilin Yang
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yaping Wang
- Wuxi Second People’s Hospital of Nanjing Medical University, Wuxi, Jiangsu, China
- * E-mail: (YW); (QF)
| | - Qiang Fu
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
- * E-mail: (YW); (QF)
| |
Collapse
|
26
|
New molecules and old drugs as emerging approaches to selectively target human glioblastoma cancer stem cells. BIOMED RESEARCH INTERNATIONAL 2014; 2014:126586. [PMID: 24527434 PMCID: PMC3909978 DOI: 10.1155/2014/126586] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Accepted: 12/04/2013] [Indexed: 02/07/2023]
Abstract
Despite relevant progress obtained by multimodal treatment, glioblastoma (GBM), the most aggressive primary brain tumor, is still incurable. The most encouraging advancement of GBM drug research derives from the identification of cancer stem cells (CSCs), since these cells appear to represent the determinants of resistance to current standard therapies. The goal of most ongoing studies is to identify drugs able to affect CSCs biology, either inducing selective toxicity or differentiating this tumor cell population into nontumorigenic cells. Moreover, the therapeutic approach for GBM could be improved interfering with chemo- or radioresistance mechanisms, microenvironment signals, and the neoangiogenic process. During the last years, molecular targeted compounds such as sorafenib and old drugs, like metformin, displayed interesting efficacy in preclinical studies towards several tumors, including GBM, preferentially affecting CSC viability. In this review, the latest experimental results, controversies, and prospective application concerning these promising anticancer drugs will be discussed.
Collapse
|
27
|
Barbieri F, Thellung S, Würth R, Gatto F, Corsaro A, Villa V, Nizzari M, Albertelli M, Ferone D, Florio T. Emerging Targets in Pituitary Adenomas: Role of the CXCL12/CXCR4-R7 System. Int J Endocrinol 2014; 2014:753524. [PMID: 25484899 PMCID: PMC4248486 DOI: 10.1155/2014/753524] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 10/21/2014] [Indexed: 12/15/2022] Open
Abstract
Chemokines are chemotactic regulators of immune surveillance in physiological and pathological conditions such as inflammation, infection, and cancer. Several chemokines and cognate receptors are constitutively expressed in the central nervous system, not only in glial and endothelial cells but also in neurons, controlling neurogenesis, neurite outgrowth, and axonal guidance during development. In particular, the chemokine CXCL12 and its receptors, CXCR4 and CXCR7, form a functional network that controls plasticity in different brain areas, influencing neurotransmission, neuromodulation, and cell migration, and the dysregulation of this chemokinergic axis is involved in several neurodegenerative, neuroinflammatory, and malignant diseases. CXCR4 primarily mediates the transduction of proliferative signals, while CXCR7 seems to be mainly responsible for scavenging CXCL12. Importantly, the multiple intracellular signalling generated by CXCL12 interaction with its receptors influences hypothalamic modulation of neuroendocrine functions, although a direct modulation of pituitary functioning via autocrine/paracrine mechanisms was also reported. Both CXCL12 and CXCR4 are constitutively overexpressed in pituitary adenomas and their signalling induces cell survival and proliferation, as well as hormonal hypersecretion. In this review we focus on the physiological and pathological functions of immune-related cyto- and chemokines, mainly focusing on the CXCL12/CXCR4-7 axis, and their role in pituitary tumorigenesis. Accordingly, we discuss the potential targeting of CXCR4 as novel pharmacological approach for pituitary adenomas.
Collapse
Affiliation(s)
- Federica Barbieri
- Department of Internal Medicine and Medical Specialties and Center of Excellence for Biomedical Research (CEBR), University of Genova, Viale Benedetto XV, 2-16132 Genova, Italy
- *Federica Barbieri:
| | - Stefano Thellung
- Department of Internal Medicine and Medical Specialties and Center of Excellence for Biomedical Research (CEBR), University of Genova, Viale Benedetto XV, 2-16132 Genova, Italy
| | - Roberto Würth
- Department of Internal Medicine and Medical Specialties and Center of Excellence for Biomedical Research (CEBR), University of Genova, Viale Benedetto XV, 2-16132 Genova, Italy
| | - Federico Gatto
- Department of Internal Medicine and Medical Specialties and Center of Excellence for Biomedical Research (CEBR), University of Genova, Viale Benedetto XV, 2-16132 Genova, Italy
| | - Alessandro Corsaro
- Department of Internal Medicine and Medical Specialties and Center of Excellence for Biomedical Research (CEBR), University of Genova, Viale Benedetto XV, 2-16132 Genova, Italy
| | - Valentina Villa
- Department of Internal Medicine and Medical Specialties and Center of Excellence for Biomedical Research (CEBR), University of Genova, Viale Benedetto XV, 2-16132 Genova, Italy
| | - Mario Nizzari
- Department of Internal Medicine and Medical Specialties and Center of Excellence for Biomedical Research (CEBR), University of Genova, Viale Benedetto XV, 2-16132 Genova, Italy
| | - Manuela Albertelli
- Department of Internal Medicine and Medical Specialties and Center of Excellence for Biomedical Research (CEBR), University of Genova, Viale Benedetto XV, 2-16132 Genova, Italy
| | - Diego Ferone
- Department of Internal Medicine and Medical Specialties and Center of Excellence for Biomedical Research (CEBR), University of Genova, Viale Benedetto XV, 2-16132 Genova, Italy
| | - Tullio Florio
- Department of Internal Medicine and Medical Specialties and Center of Excellence for Biomedical Research (CEBR), University of Genova, Viale Benedetto XV, 2-16132 Genova, Italy
| |
Collapse
|
28
|
Reubi JC, Schonbrunn A. Illuminating somatostatin analog action at neuroendocrine tumor receptors. Trends Pharmacol Sci 2013; 34:676-88. [PMID: 24183675 DOI: 10.1016/j.tips.2013.10.001] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 09/26/2013] [Accepted: 10/03/2013] [Indexed: 02/08/2023]
Abstract
Somatostatin analogs for the diagnosis and therapy of neuroendocrine tumors (NETs) have been used in clinical applications for more than two decades. Five somatostatin receptor subtypes have been identified and molecular mechanisms of somatostatin receptor signaling and regulation have been elucidated. These advances increased understanding of the biological role of each somatostatin receptor subtype, their distribution in NETs, as well as agonist-specific regulation of receptor signaling, internalization, and phosphorylation, particularly for the sst2 receptor subtype, which is the primary target of current somatostatin analog therapy for NETs. Various hypotheses exist to explain differences in patient responsiveness to somatostatin analog inhibition of tumor secretion and growth as well as differences in the development of tumor resistance to therapy. In addition, we now have a better understanding of the action of both first generation (octreotide, lanreotide, Octreoscan) and second generation (pasireotide) FDA-approved somatostatin analogs, including the biased agonistic character of some agonists. The increased understanding of somatostatin receptor pharmacology provides new opportunities to design more sophisticated assays to aid the future development of somatostatin analogs with increased efficacy.
Collapse
Affiliation(s)
- Jean Claude Reubi
- Cell Biology and Experimental Cancer Research, Institute of Pathology, University of Berne, Berne, Switzerland.
| | | |
Collapse
|
29
|
|
30
|
Gadelha MR, Kasuki L, Korbonits M. Novel pathway for somatostatin analogs in patients with acromegaly. Trends Endocrinol Metab 2013; 24:238-46. [PMID: 23270713 DOI: 10.1016/j.tem.2012.11.007] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Revised: 11/21/2012] [Accepted: 11/24/2012] [Indexed: 01/11/2023]
Abstract
Acromegaly is a chronic disease with increased morbidity and mortality, where usually multiple treatment modalities are used. The somatostatin analogs (SSAs) are the mainstay of medical therapy but, in many patients, including those with a germline mutation in the aryl hydrocarbon receptor-interacting protein (AIP) gene, disease activity cannot be controlled with these drugs. Previous data have suggested the involvement of the tumor-suppressor gene ZAC1 in the mechanism of action of SSAs, and more recent findings suggested that SSAs could regulate AIP, which in turn can stimulate ZAC1, therefore suggesting the existence of a SSA-AIP-ZAC1-somatostatin effect pathway. The current review discusses these novel observations, highlighting their significance in the treatment of sporadic and familial somatotroph adenomas.
Collapse
Affiliation(s)
- Mônica R Gadelha
- Division of Endocrinology, Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Professor Rodolpho Paulo Rocco street 255, Rio de Janeiro, Brazil
| | | | | |
Collapse
|
31
|
Albertelli M, Arvigo M, Boschetti M, Ferone D, Gatto F, Minuto F. Somatostatin receptor pathophysiology in the neuroendocrine system. Expert Rev Endocrinol Metab 2013; 8:149-157. [PMID: 30736175 DOI: 10.1586/eem.13.7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The actions of somatostatin (SRIF) are mediated by specific G protein-coupled receptors, named SRIF receptor (SSTR) subtypes 1, 2, 3 and 5. SRIF binding to SSTR activates a series of second messenger systems, resulting in the inhibition of calcium channels and adenylate cyclase activity, ultimately leading to inhibition of hormone secretion, while stimulation of other second messengers, such as phosphotyrosine phosphatases play a role in the control of cell growth. The SSTR and dopamine receptor families share a 30% sequence homology and appear to be structurally related. The knowledge on the pathophysiology of these two families of G protein-coupled receptors in neuroendocrine tumors has progressively increased due to the new insights in receptor dimerization, internalization and trafficking. Depending on the expression of different SSTRs in tissues, their combinations and interactions affect the functionality of the subtypes expressed and the influence of the microenvironment, the response to ligands and, by consequence, the response to treatment can be very different.
Collapse
Affiliation(s)
| | - Marica Arvigo
- a Department of Internal Medicine, University of Genova, Genova, Italy
| | - Mara Boschetti
- a Department of Internal Medicine, University of Genova, Genova, Italy
- b IRCSS AOU San Martino - IST, Genova, Italy
| | - Diego Ferone
- a Department of Internal Medicine, University of Genova, Genova, Italy
- b IRCSS AOU San Martino - IST, Genova, Italy
| | - Federico Gatto
- a Department of Internal Medicine, University of Genova, Genova, Italy
| | - Francesco Minuto
- a Department of Internal Medicine, University of Genova, Genova, Italy
- b IRCSS AOU San Martino - IST, Genova, Italy
- c Department of Internal Medicine, University of Genova, Genova, Italy.
| |
Collapse
|
32
|
Peptide receptor targeting in cancer: the somatostatin paradigm. INTERNATIONAL JOURNAL OF PEPTIDES 2013; 2013:926295. [PMID: 23476673 PMCID: PMC3582104 DOI: 10.1155/2013/926295] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2012] [Revised: 12/10/2012] [Accepted: 12/28/2012] [Indexed: 02/06/2023]
Abstract
Peptide receptors involved in pathophysiological processes represent promising therapeutic targets. Neuropeptide somatostatin (SST) is produced by specialized cells in a large number of human organs and tissues. SST primarily acts as inhibitor of endocrine and exocrine secretion via the activation of five G-protein-coupled receptors, named sst1–5, while in central nervous system, SST acts as a neurotransmitter/neuromodulator, regulating locomotory and cognitive functions. Critical points of SST/SST receptor biology, such as signaling pathways of individual receptor subtypes, homo- and heterodimerization, trafficking, and cross-talk with growth factor receptors, have been extensively studied, although functions associated with several pathological conditions, including cancer, are still not completely unraveled. Importantly, SST exerts antiproliferative and antiangiogenic effects on cancer cells in vitro, and on experimental tumors in vivo. Moreover, SST agonists are clinically effective as antitumor agents for pituitary adenomas and gastro-pancreatic neuroendocrine tumors. However, SST receptors being expressed by tumor cells of various tumor histotypes, their pharmacological use is potentially extendible to other cancer types, although to date no significant results have been obtained. In this paper the most recent findings on the expression and functional roles of SST and SST receptors in tumor cells are discussed.
Collapse
|
33
|
Tulipano G, Faggi L, Losa M, Mortini P, Spinello M, Sibilia V, Pagani F, Cocchi D, Giustina A. Effects of AMPK activation and combined treatment with AMPK activators and somatostatin on hormone secretion and cell growth in cultured GH-secreting pituitary tumor cells. Mol Cell Endocrinol 2013; 365:197-206. [PMID: 23116772 DOI: 10.1016/j.mce.2012.10.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Revised: 10/18/2012] [Accepted: 10/18/2012] [Indexed: 12/25/2022]
Abstract
We investigated the effects of the AMPK activator AICAR as compared to somatostatin-14 on cell viability and GH secretion in human GH-secreting pituitary adenomas in vitro and in rat GH3 cells. Overnight treatment with AICAR increased phospho-(threonine-172) AMPK levels (activated AMPK) in cultured human adenomas. As to the effects on cell viability, four adenomas out of 15 were responsive to AICAR (0.4mM) and five adenomas were responsive to SS-14 (100 nM). One adenoma was responsive to both somatostatin and AICAR. The effects of cotreatment with SS-14 and AICAR were investigated in eight adenomas. In two adenomas, the effects of AICAR+SS-14 did not exceed the effect of AICAR. In two adenomas which were not responsive to either AICAR or SS-14, the cotreatment was able to reduce cell viability versus control. Two adenomas were not responsive to any treatment. As to the effects on GH secretion, nine adenomas out of 15 were responsive to AICAR. Twelve adenomas were responsive to SS-14. Eight adenomas were responsive to both AICAR and SS-14. Cotreatment exceeded the effect of single treatments in 4 out of 10 adenomas. In GH3 cells, AICAR reduced the activity of p70S6 kinase, which plays an important role in cell growth. SS-14 did not affect significantly AMPK phosphorylation and p70S6K activity but it was able to enhance the inhibitory effect of AICAR on phospho-S6 levels. Moreover, AICAR and SS-14 reduced ERK phosphorylation with a different time course. The combined treatment reduced phospho-ERK levels at any time point.
Collapse
Affiliation(s)
- Giovanni Tulipano
- Pharmacology Unit, Department of Biomedical Sciences and Biotechnologies, University of Brescia, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Antiangiogenic role of miR-361 in human umbilical vein endothelial cells: functional interaction with the peptide somatostatin. Naunyn Schmiedebergs Arch Pharmacol 2012; 386:15-27. [PMID: 23128854 DOI: 10.1007/s00210-012-0808-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Accepted: 10/22/2012] [Indexed: 12/15/2022]
Abstract
Somatostatin (SRIF) acts as antiangiogenic factor, but its role in the regulation of microRNAs (miRNAs) targeting proangiogenic factors is unknown. We used human umbilical vein endothelial cells (HUVEC) to investigate whether (1) miRNAs targeting proangiogenic factors are influenced by hypoxia, (2) their expression is regulated by SRIF, and (3) SRIF-regulated miRNAs affect HUVEC angiogenic phenotype. The involvement of signal transducer and activator of transcription (STAT) 3 and hypoxia inducible factor (HIF)-1 in miRNA effects was studied. Quantitative real-time PCR, Western blot, cell proliferation assays, and enzyme-linked immunosorbent assay (ELISA) were used. Using specific algorithms, three miRNAs (miR-17, miR-18b, and miR-361) were predicted to bind angiogenesis-associated factors including STAT3, HIF-1α, and vascular endothelial growth factor (VEGF). Hypoxia downregulates miR-17 and miR-361 without affecting miR-18b. SRIF restored decreased levels of miR-361 acting at the SRIF receptor sst(1). Downregulated miR-361 was also restored by HIF-1α inhibition with YC-1. Combined application of SRIF did not influence YC-1-induced miR-361 downregulation, suggesting that YC-1 and SRIF modulate miR-361 through a common mechanism involving HIF-1α. This possibility was confirmed by the result that HIF-1α activation in normoxia-downregulated miR-361 and that this downregulation was prevented by SRIF. miR-361 overexpression reduced hypoxia-induced cell proliferation and VEGF release indicating miR-361 involvement in the acquisition of an angiogenic phenotype by HUVEC. miR-361 effects on VEGF were enhanced by the coadministration of SRIF. Our results suggest that (1) SRIF regulates miR-361 expression through a control on HIF-1, (2) miR-361 affects HUVEC angiogenic phenotype, and (3) SRIF and miR-361 act cooperatively in limiting hypoxia-induced VEGF release.
Collapse
|
35
|
Shpakov AO. Somatostatin receptors and signaling cascades coupled to them. J EVOL BIOCHEM PHYS+ 2012. [DOI: 10.1134/s0022093012040020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
36
|
Hernández-Pinto AM, Puebla-Jiménez L, Perianes-Cachero A, Arilla-Ferreiro E. Vitamin E deficiency impairs the somatostatinergic receptor-effector system and leads to phosphotyrosine phosphatase overactivation and cell death in the rat hippocampus. J Nutr Biochem 2012; 24:848-58. [PMID: 22902329 DOI: 10.1016/j.jnutbio.2012.05.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Revised: 04/17/2012] [Accepted: 05/01/2012] [Indexed: 11/30/2022]
Abstract
Vitamin E plays an essential role in maintaining the structure and function of the nervous system, and its deficiency, commonly associated with fat malabsorption diseases, may reduce neuronal survival. We previously demonstrated that the somatostatinergic system, implicated in neuronal survival control, can be modulated by α-tocopherol in the rat dentate gyrus, increasing cyclic adenosine monophosphate response element binding protein phosphorylation. To gain a better understanding of the molecular actions of tocopherols and examine the link among vitamin E, somatostatin and neuronal survival, we have investigated the effects of a deficiency and subsequent administration of tocopherol on the somatostatin signaling pathway and neuronal survival in the rat hippocampus. No changes in somatostatin expression were detected in vitamin-E-deficient rats. These rats, however, showed a significant increase in the somatostatin receptor density and dissociation constant, which correlated with a significant increase in the protein levels of somatostatin receptors. Nevertheless, vitamin E deficiency impaired the ability of the somatostatin receptors to couple to the effectors adenylyl cyclase and phosphotyrosine phosphatase by diminishing Gi protein functionality. Furthermore, vitamin E deficiency significantly increased phosphotyrosine phosphatase activity and PTPη expression, as well as PKCδ activation, and decreased extracellular-signal-regulated kinase phosphorylation. All these changes were accompanied by an increase in neuronal cell death. Subsequent α-tocopherol administration partially or completely reversed all these values to control levels. Altogether, our results prove the importance of vitamin E homeostasis in the somatostatin receptor-effector system and suggest a possible mechanism by which this vitamin may regulate the neuronal cell survival in the adult hippocampus.
Collapse
Affiliation(s)
- Alberto M Hernández-Pinto
- Biochemical and Molecular Biology Department, Neuro-Biochemical Group, Faculty of Medicine, Universidad de Alcalá de Henares, Madrid, Spain
| | | | | | | |
Collapse
|
37
|
Parry JJ, Chen R, Andrews R, Lears KA, Rogers BE. Identification of critical residues involved in ligand binding and G protein signaling in human somatostatin receptor subtype 2. Endocrinology 2012; 153:2747-55. [PMID: 22495673 PMCID: PMC3359596 DOI: 10.1210/en.2011-1662] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
G protein signaling through human somatostatin receptor subtype 2 (SSTR2) is well known, but the amino acids involved in stimulation of intracellular responses upon ligand binding have not been characterized. We constructed a series of point mutants in SSTR2 at amino acid positions 89, 139, and 140 in attempts to disrupt G protein signaling upon ligand binding. The aspartic acid changes at position 89 to either Ala, Leu, or Arg generated mutant receptors with varying expression profiles and a complete inability to bind somatostatin-14 (SST). Mutations to Asp 139 and Arg 140 also led to varying expression profiles with some mutants maintaining their affinity for SST. Mutation of Arg 140 to Ala resulted in a mutated receptor that had a B(max) and dissociation constant (K(d)) similar to wild-type receptor but was still coupled to the G protein as determined in both a cAMP assay and a calcium-release assay. In contrast, mutation of Asp 139 to Asn resulted in a mutated receptor with B(max) and K(d) values that were similar to wild type but was uncoupled from G protein-mediated cAMP signaling, but not calcium release. Thus, we identified mutations in SSTR2 that result in either receptor expression levels that are similar to wild type but is completely ablated for ligand binding or a receptor that maintains affinity for SST and is uncoupled from G protein-mediated cAMP signaling.
Collapse
Affiliation(s)
- Jesse J Parry
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, Missouri 63108-8224, USA
| | | | | | | | | |
Collapse
|
38
|
Annunziata M, Luque RM, Durán-Prado M, Baragli A, Grande C, Volante M, Gahete MD, Deltetto F, Camanni M, Ghigo E, Castaño JP, Granata R. Somatostatin and somatostatin analogues reduce PDGF-induced endometrial cell proliferation and motility. Hum Reprod 2012; 27:2117-29. [PMID: 22588000 DOI: 10.1093/humrep/des144] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Endometriosis is characterized by ectopic implantation of endometrial cells, which show increased proliferation and migration. Somatostatin (SST) and its analogues inhibit normal and cancer cell growth and motility through the SST receptors, sst1-5. Cortistatin (CST), which displays high structural and functional homology with SST, binds all ssts, as well as MrgX2. Our objective was to investigate the gene expression of the SST/CST system and to determine the effect of SST and its analogues on platelet-derived growth factor (PDGF)-induced proliferation and motility in telomerase-immortalized human endometrial stromal cell (T HESC) line and in primary endometrial stromal cell (ESCs) isolated from human endometriotic tissues. METHODS Ectopic endometrial tissues were collected from women (n= 23) undergoing laparoscopic surgery for endometriosis (Stage III/IV). Gene expression was evaluated by real-time PCR, cell motility by wound healing assay, protein expression and β-actin rearrangement by immunofluorescence, cell proliferation by the Alamar blue assay and ERK1/2 and Akt phosphorylation by western blot. RESULTS Human endometriotic tissues, primary ESCs and T HESCs expressed SST, CST and ssts. SST, its analogues SOM230 and octreotide, as well as CST, counteracted PDGF-induced proliferation and migration in both ESCs and T HESCs. SST also inhibited vascular endothelial growth factor and metalloprotease-2 mRNA expression, and reduced basal and PDGF-induced ERK1/2 phosphorylation. CONCLUSION These results indicate that the SST/CST system is expressed in endometriotic tissues and cells. The inhibitory effects of SST and its analogues on PDGF-induced proliferation and motility suggest that these peptides may represent promising tools in the treatment of endometriosis.
Collapse
Affiliation(s)
- Marta Annunziata
- Laboratory of Molecular and Cellular Endocrinology, Department of Internal Medicine, University of Turin, Corso Dogliotti 14, 10126 Turin, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Mei S, Cammalleri M, Azara D, Casini G, Bagnoli P, Dal Monte M. Mechanisms underlying somatostatin receptor 2 down-regulation of vascular endothelial growth factor expression in response to hypoxia in mouse retinal explants. J Pathol 2012; 226:519-533. [DOI: 10.1002/path.3006] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
|
40
|
A switch of G protein-coupled receptor binding preference from phosphoinositide 3-kinase (PI3K)-p85 to filamin A negatively controls the PI3K pathway. Mol Cell Biol 2011; 32:1004-16. [PMID: 22203038 DOI: 10.1128/mcb.06252-11] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Frequent oncogenic alterations occur in the phosphoinositide 3-kinase (PI3K) pathway, urging identification of novel negative controls. We previously reported an original mechanism for restraining PI3K activity, controlled by the somatostatin G protein-coupled receptor (GPCR) sst2 and involving a ligand-regulated interaction between sst2 with the PI3K regulatory p85 subunit. We here identify the scaffolding protein filamin A (FLNA) as a critical player regulating the dynamic of this complex. A preexisting sst2-p85 complex, which was shown to account for a significant basal PI3K activity in the absence of ligand, is disrupted upon sst2 activation. FLNA was here identified as a competitor of p85 for direct binding to two juxtaposed sites on sst2. Switching of GPCR binding preference from p85 toward FLNA is determined by changes in the tyrosine phosphorylation of p85- and FLNA-binding sites on sst2 upon activation. It results in the disruption of the sst2-p85 complex and the subsequent inhibition of PI3K. Knocking down FLNA expression, or abrogating FLNA recruitment to sst2, reversed the inhibition of PI3K and of tumor growth induced by sst2. Importantly, we report that this FLNA inhibitory control on PI3K can be generalized to another GPCR, the mu opioid receptor, thereby providing an unprecedented mechanism underlying GPCR-negative control on PI3K.
Collapse
|
41
|
Chen JH, He HC, Jiang FN, Militar J, Ran PY, Qin GQ, Cai C, Chen XB, Zhao J, Mo ZY, Chen YR, Zhu JG, Liu X, Zhong WD. Analysis of the specific pathways and networks of prostate cancer for gene expression profiles in the Chinese population. Med Oncol 2011; 29:1972-84. [PMID: 22038724 DOI: 10.1007/s12032-011-0088-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2011] [Accepted: 10/07/2011] [Indexed: 01/04/2023]
Abstract
The global physiological function of specifically expressed genes of prostate cancer in Chinese patients is unclear. This study aims to determine the genome-wide expression of genes related to prostate cancer in the Chinese population. Genes that were differentially expressed in prostate cancer were identified using DNA microarray technology. Expressions were validated by using real-time PCR. The identified genes were analyzed using the ingenuity pathway analysis (IPA) to investigate the gene ontology, functional pathway and network. A total of 1,444 genes (Fold time ≥ 1.5; P ≤ 0.05) were differentially expressed in prostate primary tumor tissue compared with benign tissue. IPA revealed a unique landscape where inductions of certain pathways were involved in Cell Cycle Regulation and proliferation. Network analysis not only confirmed that protein interactions lead to the deregulation of DNA Replication, Recombination and Repair, Cellular Compromise and Cell Cycle, Genetic Disorders and Connective Tissue Disorders, but it was also observed that many of the genes regulated by Myc contributed to the modulation of lipid Metabolism and Nucleic Acid Metabolism. Both pathway and network analysis exhibited some remarkable characteristics of prostate cancer for Chinese patients, which showed profound differences from that of other non-Chinese populations. These differences may provide new insights into the molecular cascade of prostate cancer that occurs in Chinese patients.
Collapse
Affiliation(s)
- Jia-hong Chen
- Department of Urology, Guangzhou First Municipal People's Hospital, Affiliated Guangzhou Medical College, 510180 Guangzhou, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Cakir M, Dworakowska D, Grossman A. Somatostatin receptor biology in neuroendocrine and pituitary tumours: part 1--molecular pathways. J Cell Mol Med 2011; 14:2570-84. [PMID: 20629989 PMCID: PMC4373477 DOI: 10.1111/j.1582-4934.2010.01125.x] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Neuroendocrine tumours (NETs) may occur at many sites in the body although the majority occur within the gastroenteropancreatic axis. Non-gastroenteropancreatic NETs encompass phaeochromocytomas and paragangliomas, medullary thyroid carcinoma, anterior pituitary tumour, broncho-pulmonary NETs and parathyroid tumours. Like most endocrine tumours, NETs also express somatostatin (SST) receptors (subtypes 1–5) whose ligand SST is known to inhibit endocrine and exocrine secretions and have anti-tumour effects. In the light of this knowledge, the idea of using SST analogues in the treatment of NETs has become increasingly popular and new studies have centred upon the development of new SST analogues. We attempt to review SST receptor (SSTR) biology primarily in neuroendocrine tissues, focusing on pituitary tumours. A full data search was performed through PubMed over the years 2000–2009 with keywords ‘somatostatin, molecular biology, somatostatin receptors, somatostatin signalling, NET, pituitary’ and all relevant publications have been included, together with selected publications prior to that date. SSTR signalling in non-neuroendocrine solid tumours is beyond the scope of this review. SST is a potent anti-proliferative and anti-secretory agent for some NETs. The successful therapeutic use of SST analogues in the treatment of these tumours depends on a thorough understanding of the diverse effects of SSTR subtypes in different tissues and cell types. Further studies will focus on critical points of SSTR biology such as homo- and heterodimerization of SSTRs and the differences between post-receptor signalling pathways of SSTR subtypes.
Collapse
Affiliation(s)
- Mehtap Cakir
- Selcuk University, Meram School of Medicine, Division of Endocrinology and Metabolism, Konya, Turkey.
| | | | | |
Collapse
|
43
|
Branch DR. Role of G protein-coupled vasoactive intestinal peptide receptors in HIV integration. Future Virol 2011. [DOI: 10.2217/fvl.11.42] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The pathogenesis of HIV infection is closely linked to the replication of the virus in vivo. Even though the progress in anti-HIV-1 chemotherapy in the past several years has been dramatic, the efficient protection against HIV-1 infection still remains one of the most important global challenges. The complete blockage of AIDS progression appears to be difficult with current treatment due to the rapid occurrence of viral drug-resistance, increasing cost and the likelihood of adverse side effects. Furthermore, although originally regarded with high hope, development of a suitable vaccine appears to be years away. The purpose of this article is to describe previous findings regarding a potentially important role of the vasoactive intestinal peptide/pituitary adenylate cyclase-activating polypeptide (VPAC) family of G protein-coupled receptors in HIV-1 infection, to provide evidence for the involvement of these receptors in providing signals that can control the integration of the virus into the host DNA and to report new findings that support a role for VPAC receptors in the facilitation of HIV integration.
Collapse
Affiliation(s)
- Donald R Branch
- Research & Development, Canadian Blood Services, Immunology Hub, Toronto Centre, Toronto, Ontario M5G 2M1, Canada
| |
Collapse
|
44
|
Abstract
Somatostatin is an important regulator of endocrine and exocrine secretion, affecting the release of many hormones. The effects of somatostatin are mediated through its interaction with one of five somatostatin receptors. Gastroenteropancreatic neuroendocrine tumors (GEP-NETs) express multiple somatostatin receptors, making them excellent potential therapeutic targets. Many trials have shown that treatment with somatostatin analogs is associated with disease stabilization and prolonged survival. More recently, somatostatin analogs have been shown to have antiproliferative effects, thus broadening the scope of their uses. In this review, we update the current data on the treatment of GEP-NETs with somatostatin analogs, with particular emphasis on the results of the PROMID study. In addition, we discuss the current state of knowledge of novel therapies against GEP-NETs, including the use of somatostatin analogs with broader receptor binding profiles, chimeric somatostatin-dopamine molecules, combinations of somatostatin analogs with other active chemotherapy agents, and peptide receptor-targeted radionuclide therapy.
Collapse
|
45
|
Strosberg JR, Cheema A, Kvols LK. A Review of Systemic and Liver-Directed Therapies for Metastatic Neuroendocrine Tumors of the Gastroenteropancreatic Tract. Cancer Control 2011; 18:127-137. [DOI: 10.1177/107327481101800207] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
Affiliation(s)
- Jonathan R. Strosberg
- Department of Gastrointestinal Oncology at the H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Asima Cheema
- Department of Gastrointestinal Oncology at the H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Larry K. Kvols
- Department of Gastrointestinal Oncology at the H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| |
Collapse
|
46
|
Fiorentini C, Mattanza C, Collo G, Savoia P, Spano P, Missale C. The tyrosine phosphatase Shp-2 interacts with the dopamine D(1) receptor and triggers D(1) -mediated Erk signaling in striatal neurons. J Neurochem 2011; 117:253-63. [PMID: 21272002 DOI: 10.1111/j.1471-4159.2011.07196.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We report a novel mechanism for dopamine D(1) receptor (D(1) R)-mediated extracellular signal-regulated kinases (Erk) activation in rat striatum. Erk signaling depends on phosphorylation and dephosphorylation events mediated by specific kinases and phosphatases. The tyrosine phosphatase Shp-2, that is required for Erk activation by tyrosine kinase receptors, has been recently shown to regulate signaling downstream of few G protein-coupled receptors. We show that the D(1) R interacts with Shp-2, that D(1) R stimulation results in Shp-2 tyrosine phosphorylation and activation in primary striatal neuronal cultures and that D(1) R/Shp-2 interaction is required for transmitting D(1) R-dependent signaling to Erk1/2 activation. D(1) R-mediated Erk1/2 phosphorylation in cultured striatal neurons is in fact abolished by over-expression of the inactive Shp-2(C/S) mutant and by small interfering RNA-induced Shp-2 silencing. Moreover, by using selective inhibitors we show that both D(1) R-induced Shp-2 activation and Erk1/2 phosphorylation are dependent on the cyclic AMP/protein kinase A pathway and require Src. These results, which were substantiated also in transfected human embryonic kidney 293 cells, provide a novel mechanism by which to converge D(1) R signaling to the Erk pathway and suggest that Shp-2 or the D(1) R/Shp-2 interface could represent a potential drug target for disorders of dopamine transmission involving malfunctioning of D(1) R signaling.
Collapse
Affiliation(s)
- Chiara Fiorentini
- Division of Pharmacology, Department of Biomedical Sciences and Biotechnology and National Institute of Neuroscience, University of Brescia, Brescia, Italy.
| | | | | | | | | | | |
Collapse
|
47
|
Somatostatin Receptors in Lung Cancer: From Function to Molecular Imaging and Therapeutics. ACTA ACUST UNITED AC 2011; 10:69-76. [PMID: 25663834 DOI: 10.6058/jlc.2011.10.2.69] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Lung cancer is a deadly disease that is difficult to diagnose and even more difficult to treat effectively. Many pathways are known to affect tumor growth, and targeting these pathways provides the cornerstone by which cancer is treated. Somatostatin receptors (SSTR) are a family of G protein coupled receptors that signal to alter hormonal secretion, increase apoptosis, and decrease cellular proliferation. These receptors are expressed in many normal and malignant cells, including both small cell and non-small cell lung cancer. Synthetic analogs of SSTRs are commercially available, but their effects in lung cancer are still largely uncertain. Signaling pathway studies have shown that SSTRs signal through phosphotyrosine phosphatases to induce apoptosis as well as to decrease cell proliferation. Radiolabeled SSTR2 analogs are utilized for radiographic imaging of tumors, which, when combined with positron emission tomography-computed tomography (PET-CT) may improve detection of lung cancer. These radiolabeled SSTR2 analogs also hold promise for targeted chemotherapy as well as radiotherapy. In this review, we summarize what is known about SSTRs and focus our discussion on the knowledge as it relates to lung cancer biology, as well as discuss current and future uses of these receptors for imaging and therapy of lung cancer.
Collapse
|
48
|
Castillo V, Theodoropoulou M, Stalla J, Gallelli MF, Cabrera-Blatter MF, Haedo MR, Labeur M, Schmid HA, Stalla GK, Arzt E. Effect of SOM230 (pasireotide) on corticotropic cells: action in dogs with Cushing's disease. Neuroendocrinology 2011; 94:124-36. [PMID: 21525729 DOI: 10.1159/000327429] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2010] [Accepted: 03/13/2011] [Indexed: 11/19/2022]
Abstract
SOM230 (pasireotide) is a multiligand somatostatin (SRIF) analog able to bind to somatostatin receptor (SSTR) subtypes 1, 2, 3 and 5, and trigger antisecretory and antiproliferative signaling cascades. Canines have become in vivo models to test the pharmacological treatment of corticotropinomas because they frequently develop Cushing's disease in a spontaneous manner, due to adrenocorticotropic hormone (ACTH)-producing pituitary adenomas. Different levels of expression of SSTR2 and SSTR5 have been shown in both mouse AtT20 cells and canine tumoral corticotropinoma cells. The objective of this study was to evaluate whether SOM230 controls both tumor cell growth and hormone synthesis, therefore controlling the disease. SOM230 was tested in dogs suffering from Cushing's disease (10 animals were treated continuously during 6 months, and another 10 were treated with 3 cycles consisting of 2 months of treatment followed by a 2-month rest period). A significant decrease in ACTH, urinary cortisol creatinine ratio, adenoma size (magnetic nuclear resonance) and improvement of clinical signs were obtained, without side effects. AtT20 cells treated with SOM230 suppressed pro-opiomelanocortin (POMC) promoter activity through SSTR2, via the G(i) α-subunit, and reduced Nur77/Nurr1 transcriptional activity. We conclude that SOM230, in addition to its well-described antisecretory effects, inhibits, as shown in AtT20 cells, ACTH synthesis at the POMC transcriptional level, an effect mediated mainly through SSTR2, and limits tumor growth. The controlled Cushing's disease in the dogs that received the treatment indicates that SOM230 has a potential therapeutic use in humans suffering from Cushing's disease.
Collapse
Affiliation(s)
- Victor Castillo
- Unidad de Endocrinología, Hospital Escuela, Facultad de Ciencias Veterinarias, Universidad de Buenos Aires, Buenos Aires, Argentina
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Strosberg J, Kvols L. Antiproliferative effect of somatostatin analogs in gastroenteropancreatic neuroendocrine tumors. World J Gastroenterol 2010; 16:2963-70. [PMID: 20572298 PMCID: PMC2890935 DOI: 10.3748/wjg.v16.i24.2963] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Somatostatin analogs were initially developed for the control of hormonal syndromes associated with neuroendocrine tumors (NETs). In recent years, accumulating data has supported their role as antiproliferative agents, capable of stabilizing tumor growth in patients with metastatic neuroendocrine malignancies, including carcinoid and pancreatic endocrine tumors. A phase III, randomized, placebo-controlled trial has now demonstrated that octreotide long-acting repeatable (LAR) 30 mg can significantly prolong time to tumor progression among patients with metastatic midgut NETs regardless of functional status, chromogranin A level or age. In addition to significantly lengthening time to tumor progression in the overall study population, subset analysis suggests that patients with low tumor burden are most likely to experience disease stabilization with octreotide LAR 30 mg, supporting the early use of octreotide LAR in patients with metastatic disease. Further research efforts are underway to evaluate the use of somatostatin analogs as antiproliferative agents in other types of gastroenteropancreatic-NETs. Ongoing studies are also evaluating novel somatostatin analogs and somatostatin analogs in combination with other anti-tumor therapies.
Collapse
|
50
|
Ben-Shlomo A, Melmed S. Pituitary somatostatin receptor signaling. Trends Endocrinol Metab 2010; 21:123-33. [PMID: 20149677 PMCID: PMC2834886 DOI: 10.1016/j.tem.2009.12.003] [Citation(s) in RCA: 134] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2009] [Revised: 12/09/2009] [Accepted: 12/10/2009] [Indexed: 12/27/2022]
Abstract
Somatotropin-release inhibitory factor (SRIF) is a major regulator of pituitary function, mostly inhibiting hormone secretion and to a lesser extent pituitary cell growth. Five SRIF receptor subtypes (SSTR1-5) are ubiquitously expressed G-protein coupled receptors. In the pituitary, SSTR1, 2, 3 and 5 are expressed, with SSTR2 and SSTR5 predominating. As new SRIF analogs have recently been introduced for treatment of pituitary disease, we evaluate the current knowledge of cell-specific pituitary SRIF receptor signaling and highlight areas of future research for comprehensive understanding of these mechanisms. Elucidating pituitary SRIF receptor signaling enables understanding of pituitary hormone secretion and cell growth, and also encourages future therapeutic development for pituitary disorders.
Collapse
Affiliation(s)
- Anat Ben-Shlomo
- Pituitary Center, Department of Medicine, Cedars Sinai Medical Center, Los Angeles, California 90048, USA
| | | |
Collapse
|