1
|
Nerubenko E, Ryazanov P, Kuritsyna N, Paltsev A, Ivanova O, Grineva E, Kostareva A, Dmitrieva R, Tsoy U. Cushing's Disease Manifestation in USP8-Mutated Corticotropinoma May Be Mediated by Interactions Between WNT Signaling and SST Trafficking. Int J Mol Sci 2024; 25:12886. [PMID: 39684597 DOI: 10.3390/ijms252312886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/25/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
In the current work, we aimed to evaluate the association of clinical data of Cushing's disease (CD) patients with USP8 mutation status and to study USP8-related molecular mechanisms connected to the regulation of corticotropinoma growth and activity. 35 CD patients were enrolled; the sequencing of exon 14 in USP8 revealed variants in eighteen adenomas, two of which were described for the first time in CD. USP8 variants were more common in women (94% vs. 76%; p = 0.001), and microadenomas and tumor recurrence were prevalent in the USP8-mutant group (44% vs. 29%; p = 0.04 and 44% vs. 22%; p = 0.0015). Preoperative ACTH and serum cortisol did not differ in the USP8-WT and USP8-mutant patients. All USP8-mutant adenomas were SST5-positive, and 73% of them were double-positive (SST5+/SST2+). A total of 50% of USP8-WT adenomas were double-negative (SST5-/SST2-), and 40% of them were SST5-positive. Analysis of transcriptome was performed for nine USP8-mutant and six USP8-WT adenomas and revealed the that the bidirectional dysregulation of Wnt signaling, including both the agonist RSPO2 and antagonist SFRP1, in the USP8-mutant corticotropinomas was downregulated. These alterations may indicate the existence of regulatory connections between USP8 enzyme activity, Wnt signaling, EGFR signaling and somatostatin receptors' trafficking, which can explain, at least in part, the clinical manifestations of CD in patients with corticotropinomas harboring USP8 variants.
Collapse
Affiliation(s)
- Elena Nerubenko
- Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia
| | - Pavel Ryazanov
- Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia
| | - Natalia Kuritsyna
- Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia
| | - Artem Paltsev
- Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia
| | - Oksana Ivanova
- Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia
| | - Elena Grineva
- Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia
| | - Anna Kostareva
- Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia
| | - Renata Dmitrieva
- Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia
| | - Uliana Tsoy
- Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia
| |
Collapse
|
2
|
Periferakis A, Tsigas G, Periferakis AT, Tone CM, Hemes DA, Periferakis K, Troumpata L, Badarau IA, Scheau C, Caruntu A, Savulescu-Fiedler I, Caruntu C, Scheau AE. Agonists, Antagonists and Receptors of Somatostatin: Pathophysiological and Therapeutical Implications in Neoplasias. Curr Issues Mol Biol 2024; 46:9721-9759. [PMID: 39329930 PMCID: PMC11430067 DOI: 10.3390/cimb46090578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/29/2024] [Accepted: 08/31/2024] [Indexed: 09/28/2024] Open
Abstract
Somatostatin is a peptide that plays a variety of roles such as neurotransmitter and endocrine regulator; its actions as a cell regulator in various tissues of the human body are represented mainly by inhibitory effects, and it shows potent activity despite its physiological low concentrations. Somatostatin binds to specific receptors, called somatostatin receptors (SSTRs), which have different tissue distributions and associated signaling pathways. The expression of SSTRs can be altered in various conditions, including tumors; therefore, they can be used as biomarkers for cancer cell susceptibility to certain pharmacological agents and can provide prognostic information regarding disease evolution. Moreover, based on the affinity of somatostatin analogs for the different types of SSTRs, the therapeutic range includes conditions such as tumors, acromegaly, post-prandial hypotension, hyperinsulinism, and many more. On the other hand, a number of somatostatin antagonists may prove useful in certain medical settings, based on their differential affinity for SSTRs. The aim of this review is to present in detail the principal characteristics of all five SSTRs and to provide an overview of the associated therapeutic potential in neoplasias.
Collapse
Affiliation(s)
- Argyrios Periferakis
- Department of Physiology, The "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Elkyda, Research & Education Centre of Charismatheia, 17675 Athens, Greece
- Akadimia of Ancient Greek and Traditional Chinese Medicine, 16675 Athens, Greece
| | - Georgios Tsigas
- Department of Physiology, The "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Aristodemos-Theodoros Periferakis
- Department of Physiology, The "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Elkyda, Research & Education Centre of Charismatheia, 17675 Athens, Greece
| | - Carla Mihaela Tone
- Department of Physiology, The "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Daria Alexandra Hemes
- Department of Physiology, The "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Konstantinos Periferakis
- Akadimia of Ancient Greek and Traditional Chinese Medicine, 16675 Athens, Greece
- Pan-Hellenic Organization of Educational Programs, 17236 Athens, Greece
| | - Lamprini Troumpata
- Department of Physiology, The "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Ioana Anca Badarau
- Department of Physiology, The "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Cristian Scheau
- Department of Physiology, The "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Radiology and Medical Imaging, "Foisor" Clinical Hospital of Orthopaedics, Traumatology and Osteoarticular TB, 030167 Bucharest, Romania
| | - Ana Caruntu
- Department of Oral and Maxillofacial Surgery, The "Carol Davila" Central Military Emergency Hospital, 010825 Bucharest, Romania
- Department of Oral and Maxillofacial Surgery, Faculty of Dental Medicine, "Titu Maiorescu" University, 031593 Bucharest, Romania
| | - Ilinca Savulescu-Fiedler
- Department of Internal Medicine, The "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Internal Medicine and Cardiology, Coltea Clinical Hospital, 030167 Bucharest, Romania
| | - Constantin Caruntu
- Department of Physiology, The "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Dermatology, "Prof. N.C. Paulescu" National Institute of Diabetes, Nutrition and Metabolic Diseases, 011233 Bucharest, Romania
| | - Andreea-Elena Scheau
- Department of Radiology and Medical Imaging, Fundeni Clinical Institute, 022328 Bucharest, Romania
| |
Collapse
|
3
|
Oron-Herman M, Kirmayer D, Lupp A, Schulz S, Kostenich G, Afargan M. Expression prevalence and dynamics of GPCR somatostatin receptors 2 and 3 as cancer biomarkers beyond NET: a paired immunohistochemistry approach. Sci Rep 2023; 13:20857. [PMID: 38012197 PMCID: PMC10682014 DOI: 10.1038/s41598-023-47877-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 11/16/2023] [Indexed: 11/29/2023] Open
Abstract
Somatostatin receptors are clinically validated GPCR biomarkers for diagnosis and treatment of various neuroendocrine tumors (NET). Among the five somatostatin receptors, SST2 and SST3 are associated with apoptosis and cell cycle arrest, making these receptor subtypes better differentiated targets in precision oncology. In this study we performed immunohistochemistry of paired tissue microarrays containing 1125 cores, representing 43 tumor types, each stained for SST2 and SST3. A 12-point immunoreactive scoring (IRS) range was used for interpretation of the staining results. We analyzed the results twice, using the conventional positivity IRS cutoffs ≥ 3 and more stringent ≥ 6. Evaluation of receptors expression dynamics was performed for tumor-nodes-metastases (TNM) defined subgroups (ovarian and hepatocellular adenocarcinomas) as a function of their tumor stage. Our results indicate that two-thirds of tested cores exhibit clinically significant expression of at least SST2 or SST3 (IRS ≥ 6). The expression prevalence of both receptors tends to decline with tumor progression. However, an unexpected upregulation of both SST2 and SST3 reemerged in metastases suggesting conserved receptors genetic potential during tumor life cycle. We suggest that SST2 and SST3 should be further explored as potential biomarkers and therapeutic tools for maximizing the efficiency of somatostatin-based precision oncology of solid tumors beyond NET.
Collapse
Affiliation(s)
- Mor Oron-Herman
- Starget Pharma, 26 Snir st., 4704086, Ramat Hasharon, Israel.
| | - David Kirmayer
- Starget Pharma, 26 Snir st., 4704086, Ramat Hasharon, Israel
| | - Amelie Lupp
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Drackendorfer Str. 1, 07747, Jena, Germany
| | - Stefan Schulz
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Drackendorfer Str. 1, 07747, Jena, Germany
| | - Genady Kostenich
- Starget Pharma, 26 Snir st., 4704086, Ramat Hasharon, Israel
- The Advanced Technology Center, Sheba Medical Center, Tel Hashomer, 5262000, Ramat Gan, Israel
| | - Michel Afargan
- Starget Pharma, 26 Snir st., 4704086, Ramat Hasharon, Israel
| |
Collapse
|
4
|
Czajkowski M, Kaemmerer D, Sänger J, Sauter G, Wirtz RM, Schulz S, Lupp A. Comparative evaluation of somatostatin and CXCR4 receptor expression in different types of thyroid carcinoma using well-characterised monoclonal antibodies. BMC Cancer 2022; 22:740. [PMID: 35799158 PMCID: PMC9261050 DOI: 10.1186/s12885-022-09839-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 06/30/2022] [Indexed: 12/02/2022] Open
Abstract
Background Papillary and follicular thyroid carcinomas can be treated surgically and with radioiodine therapy, whereas therapeutic options for advanced stage IV medullary and for anaplastic tumours are limited. Recently, somatostatin receptors (SSTs) and the chemokine receptor CXCR4 have been evaluated for the treatment of thyroid carcinomas, however, with contradictory results. Methods The expression of the five SSTs and of CXCR4 was assessed in 90 samples from 56 patients with follicular, papillary, medullary, or anaplastic thyroid carcinoma by means of immunohistochemistry using well-characterised monoclonal antibodies. The stainings were evaluated using the Immunoreactivity Score (IRS) and correlated to clinical data. In order to further substantiate the immunohistochemistry results, in serial sections of a subset of the samples receptor expression was additionally examined at the mRNA level using qRT-PCR. Results Overall, SST and CXCR4 protein expression was low in all four entities. In single cases, however, very high IRS values for SST2 and CXCR4 were observed. SST2 was the most frequently expressed receptor, found in 38% of cases, followed by SST5 and SST4, found in 14 and 9% of tumours, respectively. SST1 and SST3 could not be detected to any significant extent. CXCR4 was present in 12.5% of medullary and 25% of anaplastic carcinomas. Expression SST3, SST4, SST5 and CXCR4 was positively correlated with expression of the proliferation marker Ki-67. Additionally, a negative interrelationship between SST4 or SST5 expression and patient survival and a positive association between SST3 expression and tumour diameter were observed. qRT-PCR revealed a similar receptor expression pattern to that seen at the protein level. However, probably due to the low overall expression, no correlation was found for the SSTs or the CXCR4 between the IRS and the mRNA values. Conclusions SST- or CXCR4-based diagnostics or therapy in thyroid carcinomas should not be considered in general but may be feasible in single cases with high levels of expression of these receptors.
Collapse
Affiliation(s)
- Max Czajkowski
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Drackendorfer Str. 1, D-07747, Jena, Germany
| | - Daniel Kaemmerer
- Department of General and Visceral Surgery, Zentralklinik Bad Berka, Bad Berka, Germany
| | - Jörg Sänger
- Laboratory of Pathology and Cytology Bad Berka, Bad Berka, Germany
| | - Guido Sauter
- Institute of Pathology, University Medical Center, Hamburg-Eppendorf, Germany
| | - Ralph M Wirtz
- STRATIFYER Molecular Pathology GmbH Köln, Köln, Germany
| | - Stefan Schulz
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Drackendorfer Str. 1, D-07747, Jena, Germany
| | - Amelie Lupp
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Drackendorfer Str. 1, D-07747, Jena, Germany.
| |
Collapse
|
5
|
Rass L, Rahvar AH, Matschke J, Saeger W, Renné T, Aberle J, Flitsch J, Rotermund R. Differences in somatostatin receptor subtype expression in patients with acromegaly: new directions for targeted therapy? Hormones (Athens) 2022; 21:79-89. [PMID: 34674191 PMCID: PMC8818633 DOI: 10.1007/s42000-021-00327-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 10/04/2021] [Indexed: 11/02/2022]
Abstract
PURPOSE To analyze the expression of somatostatin receptor (SSTR)2a and 5 by immunohistochemistry (IHC) in surgically resected somatotrophic pituitary adenomas and to associate expression rates with tumor size and clinical, biochemical, and histological parameters and response to somatostatin analog (SA) therapy. METHODS Forty-three microsurgically treated patients with histopathologically proven growth hormone (GH)-producing pituitary adenoma were included (WHO 2017). SSTR subtype expression was analyzed in adenoma tissues using monoclonal antibodies (Abcam, SSTR2a-UMB1, SSTR5-UMB4). Expression rates were classified as low (≤ 20% staining positivity), moderate (21-50%), and high (> 50%). Furthermore, biochemical parameters such as human growth hormone (hGH) and insulin-like growth factor-1 (IGF-1) levels were measured and clinical, biochemical, radiological, and histological data were evaluated. RESULTS Of the 43 patients included in this study, 28 were female and 15 were male. The median age was 52 years (range 17-72 years). The median tumor size was 1.2 cm (range: 0.13-3.93 cm). All resected tumors showed positivity for somatotrophic hormone (STH). In all tissue samples, SSTR2a signal expression was detectable in immunohistochemistry, while only 39 samples were positive for SSTR5. Thirty-six samples had a high expression of SSTR2a, while three had a moderate and four a low SSTR2a signal. In comparison, SSTR5 signal was high in 26 out of 43 samples, while seven adenomas showed a moderate and six cases a low expression rate of SSTR5. The median IGF-1 was 714.2 µg/l and the median GH 19.6 mU/l (= 6.53 µg/l). The present study indicates that there is no significant relationship between the expression rates of receptor subtypes and the parameters we analyzed. However, our study revealed that smaller adenomas have a lower baseline GH level (p = 0.015), CONCLUSION: IHC with monoclonal antibodies appears to be a suitable method to determine the expression rates of SSTR2a and 5 at protein levels, as it is not possible to draw conclusions regarding receptor subtypes solely on the basis of the parameters analyzed.
Collapse
Affiliation(s)
- Lena Rass
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Amir-Hossein Rahvar
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jakob Matschke
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Wolfgang Saeger
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thomas Renné
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jens Aberle
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jörg Flitsch
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Roman Rotermund
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany.
| |
Collapse
|
6
|
Treppiedi D, Marra G, Di Muro G, Catalano R, Mangili F, Esposito E, Calebiro D, Arosio M, Peverelli E, Mantovani G. Dimerization of GPCRs: Novel insight into the role of FLNA and SSAs regulating SST 2 and SST 5 homo- and hetero-dimer formation. Front Endocrinol (Lausanne) 2022; 13:892668. [PMID: 35992099 PMCID: PMC9389162 DOI: 10.3389/fendo.2022.892668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 07/08/2022] [Indexed: 11/13/2022] Open
Abstract
The process of GPCR dimerization can have profound effects on GPCR activation, signaling, and intracellular trafficking. Somatostatin receptors (SSTs) are class A GPCRs abundantly expressed in pituitary tumors where they represent the main pharmacological targets of somatostatin analogs (SSAs), thanks to their antisecretory and antiproliferative actions. The cytoskeletal protein filamin A (FLNA) directly interacts with both somatostatin receptor type 2 (SST2) and 5 (SST5) and regulates their expression and signaling in pituitary tumoral cells. So far, the existence and physiological relevance of SSTs homo- and hetero-dimerization in the pituitary have not been explored. Moreover, whether octreotide or pasireotide may play modulatory effects and whether FLNA may participate to this level of receptor organization have remained elusive. Here, we used a proximity ligation assay (PLA)-based approach for the in situ visualization and quantification of SST2/SST5 dimerization in rat GH3 as well as in human melanoma cells either expressing (A7) or lacking (M2) FLNA. First, we observed the formation of endogenous SST5 homo-dimers in GH3, A7, and M2 cells. Using the PLA approach combined with epitope tagging, we detected homo-dimers of human SST2 in GH3, A7, and M2 cells transiently co-expressing HA- and SNAP-tagged SST2. SST2 and SST5 can also form endogenous hetero-dimers in these cells. Interestingly, FLNA absence reduced the basal number of hetero-dimers (-36.8 ± 6.3% reduction of PLA events in M2, P < 0.05 vs. A7), and octreotide but not pasireotide promoted hetero-dimerization in both A7 and M2 (+20.0 ± 11.8% and +44.1 ± 16.3% increase of PLA events in A7 and M2, respectively, P < 0.05 vs. basal). Finally, immunofluorescence data showed that SST2 and SST5 recruitment at the plasma membrane and internalization are similarly induced by octreotide and pasireotide in GH3 and A7 cells. On the contrary, in M2 cells, octreotide failed to internalize both receptors whereas pasireotide promoted robust receptor internalization at shorter times than in A7 cells. In conclusion, we demonstrated that in GH3 cells SST2 and SST5 can form both homo- and hetero-dimers and that FLNA plays a role in the formation of SST2/SST5 hetero-dimers. Moreover, we showed that FLNA regulates SST2 and SST5 intracellular trafficking induced by octreotide and pasireotide.
Collapse
Affiliation(s)
- Donatella Treppiedi
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Giusy Marra
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Genesio Di Muro
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- University Sapienza of Rome, Rome, Italy
| | - Rosa Catalano
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Federica Mangili
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Emanuela Esposito
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Davide Calebiro
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
- Centre of Membrane Proteins and Receptors, Universities of Birmingham and Nottingham, Birmingham, United Kingdom
| | - Maura Arosio
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- Endocrinology Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Erika Peverelli
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- *Correspondence: Erika Peverelli,
| | - Giovanna Mantovani
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- Endocrinology Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
7
|
Mansi R, Nicolas GP, Del Pozzo L, Abid KA, Grouzmann E, Fani M. Evaluation of a New 177Lu-Labeled Somatostatin Analog for the Treatment of Tumors Expressing Somatostatin Receptor Subtypes 2 and 5. Molecules 2020; 25:E4155. [PMID: 32932783 PMCID: PMC7570871 DOI: 10.3390/molecules25184155] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/03/2020] [Accepted: 09/09/2020] [Indexed: 12/12/2022] Open
Abstract
Targeted radionuclide therapy of somatostatin receptor (SST)-expressing tumors is only partially addressed by the established somatostatin analogs having an affinity for the SST subtype 2 (SST2). Aiming to target a broader spectrum of tumors, we evaluated the bis-iodo-substituted somatostatin analog ST8950 ((4-amino-3-iodo)-d-Phe-c[Cys-(3-iodo)-Tyr-d-Trp-Lys-Val-Cys]-Thr-NH2), having subnanomolar affinity for SST2 and SST5, labeled with [177Lu]Lu3+ via the chelator DOTA (1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid). Human Embryonic Kidney (HEK) cells stably transfected with the human SST2 (HEK-SST2) and SST5 (HEK-SST5) were used for in vitro and in vivo evaluation on a dual SST2- and SST5-expressing xenografted mouse model. natLu-DOTA-ST8950 showed nanomolar affinity for both subtypes (IC50 (95% confidence interval): 0.37 (0.22-0.65) nM for SST2 and 3.4 (2.3-5.2) for SST5). The biodistribution of [177Lu]Lu-DOTA-ST8950 was influenced by the injected mass, with 100 pmol demonstrating lower background activity than 10 pmol. [177Lu]Lu-DOTA-ST8950 reached its maximal uptake on SST2- and SST5-tumors at 1 h p.i. (14.17 ± 1.78 and 1.78 ± 0.35%IA/g, respectively), remaining unchanged 4 h p.i., with a mean residence time of 8.6 and 0.79 h, respectively. Overall, [177Lu]Lu-DOTA-ST8950 targets SST2-, SST5-expressing tumors in vivo to a lower extent, and has an effective dose similar to clinically used radiolabeled somatostatin analogs. Its main drawbacks are the low uptake in SST5-tumors and the persistent kidney uptake.
Collapse
Affiliation(s)
- Rosalba Mansi
- Division of Radiopharmaceutical Chemistry, Clinic of Radiology and Nuclear Medicine, University Hospital Basel, 4031 Basel, Switzerland; (R.M.); (L.D.P.)
| | - Guillaume Pierre Nicolas
- Division of Nuclear Medicine, Clinic of Radiology and Nuclear Medicine, University Hospital Basel, 4031 Basel, Switzerland;
| | - Luigi Del Pozzo
- Division of Radiopharmaceutical Chemistry, Clinic of Radiology and Nuclear Medicine, University Hospital Basel, 4031 Basel, Switzerland; (R.M.); (L.D.P.)
| | - Karim Alexandre Abid
- Catecholamine and Peptides Laboratory, Department of Laboratories, University Hospital of Lausanne, 1011 Lausanne, Switzerland; (K.A.A.); (E.G.)
| | - Eric Grouzmann
- Catecholamine and Peptides Laboratory, Department of Laboratories, University Hospital of Lausanne, 1011 Lausanne, Switzerland; (K.A.A.); (E.G.)
| | - Melpomeni Fani
- Division of Radiopharmaceutical Chemistry, Clinic of Radiology and Nuclear Medicine, University Hospital Basel, 4031 Basel, Switzerland; (R.M.); (L.D.P.)
| |
Collapse
|
8
|
Werner C, Dirsch O, Dahmen U, Grimm MO, Schulz S, Lupp A. Evaluation of Somatostatin and CXCR4 Receptor Expression in a Large Set of Prostate Cancer Samples Using Tissue Microarrays and Well-Characterized Monoclonal Antibodies. Transl Oncol 2020; 13:100801. [PMID: 32460182 PMCID: PMC7249232 DOI: 10.1016/j.tranon.2020.100801] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 05/11/2020] [Accepted: 05/12/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND: Prostate cancer (PCa) is the most common type of cancer among men in Western countries. Despite numerous therapeutic options, few treatments are available for patients with end-stage disease. In the present study, different somatostatin receptors (SSTs) and the chemokine receptor CXCR4 were evaluated for their suitability as novel therapeutic targets in PCa. MATERIALS AND METHODS: The expression of SST subtypes 1, 2A, 3, and 5 and of CXCR4 was evaluated in 276 PCa tumor samples on a tissue microarray (TMA) in 23 whole-block tumor samples and in 3 PCa cell lines by immunohistochemistry using well-characterized monoclonal antibodies. RESULTS: Overall, the frequency and intensity of expression of SSTs and CXCR4 were very low among the PCa samples investigated. Specifically, SST5, SST2A, and SST3 were expressed, albeit at low intensity, in 10.5%, 9.1%, and 0.7% of the TMA samples, respectively. None of the TMA samples showed SST1 or CXCR4 expression. Only a single small-cell-type neuroendocrine carcinoma that was coincidentally included among the whole-block samples exhibited strong SST2A, SST5, and CXCR4 and moderate SST3 expression. Independent of the tumor cells, the tumor capillaries in many of the PCa samples were strongly positive for SST2A, SST3, SST5, or CXCR4 expression. SST expression in the tumor cells was associated with advanced tumor grade and stage. CONCLUSION: Overall, SST and CXCR4 expression levels are clearly of no therapeutic relevance in PCa. SST- or CXCR4-based therapy might be feasible, however, in rare cases of small-cell-type neuroendocrine carcinoma of the prostate.
Collapse
Affiliation(s)
- Christoph Werner
- Department of Internal Medicine III, Jena University Hospital, Jena, Germany; Institute of Pharmacology and Toxicology, Jena University Hospital, Jena, Germany
| | - Olaf Dirsch
- Institute of Pathology, Jena University Hospital, Jena, Germany; Institute of Pathology, Klinikum Chemnitz, Chemnitz, Germany
| | - Uta Dahmen
- Department of General, Visceral and Vascular Surgery, Jena University Hospital, Jena, Germany
| | | | - Stefan Schulz
- Institute of Pharmacology and Toxicology, Jena University Hospital, Jena, Germany
| | - Amelie Lupp
- Institute of Pharmacology and Toxicology, Jena University Hospital, Jena, Germany.
| |
Collapse
|
9
|
Ehrstedt C, Ahlsten G, Strömberg B, Lindskog C, Casar-Borota O. Somatostatin receptor expression and mTOR pathway activation in glioneuronal tumours of childhood. Seizure 2020; 76:123-130. [PMID: 32062323 DOI: 10.1016/j.seizure.2020.01.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 12/15/2019] [Accepted: 01/16/2020] [Indexed: 11/29/2022] Open
Abstract
PURPOSE To investigate the expression of somatostatin receptors (SSTRs) and markers of mTOR pathway in paediatric glioneuronal tumours and correlate these findings with tumour type, BRAFV600E mutational status and clinical characteristics such as tumour location, seizure frequency and duration, and age. METHOD 37 children and adolescents with a neuropathological diagnosis of glioneuronal tumour were identified over a 22-year period. Immunohistochemical analyses for SSTRs type 1, 2A, 3, 5 and ezrin-radixin-moesin (ERM) and phosphorylated S6 (pS6), which are indicators of mTOR pathway activation, were performed in tumour specimens from 33 patients and evaluated using the immunoreactive score (IRS). The IRS were compared to tumour type, BRAFV600E status and clinical characteristics. RESULTS Ganglioglioma (GG) was the most frequently encountered subgroup (n = 27), followed by dysembryoplastic neuroepithelial tumour (DNET; n = 4). GGs expressed SSTR2A and SSTR3 to a high extent, 56 % and 44 % respectively. Expression of SSTR2A was also found in DNETs. Signs of mTOR pathway activation were abundant in GGs, but only present in one DNET. No correlations with BRAFV600E presence or clinical characteristics were found. CONCLUSIONS Expression of SSTRs and activation of mTOR pathway in paediatric glioneuronal tumour suggest that somatostatin analogues and mTOR inhibitors may have potential therapeutic implications in a subset of inoperable childhood glioneuronal tumours causing medically refractory epilepsy and/or tumour growth. Further clinical studies are warranted to validate these findings.
Collapse
Affiliation(s)
- Christoffer Ehrstedt
- Department of Women´s and Children´s Health, Section for Paediatrics, Uppsala University, Sweden; Uppsala University Children´s Hospital, Uppsala, Sweden.
| | - Gunnar Ahlsten
- Department of Women´s and Children´s Health, Section for Paediatrics, Uppsala University, Sweden; Uppsala University Children´s Hospital, Uppsala, Sweden
| | - Bo Strömberg
- Department of Women´s and Children´s Health, Section for Paediatrics, Uppsala University, Sweden; Uppsala University Children´s Hospital, Uppsala, Sweden
| | - Cecilia Lindskog
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Olivera Casar-Borota
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden; Department of Clinical Pathology, Uppsala University Hospital, Uppsala, Sweden
| |
Collapse
|
10
|
Kajtazi Y, Kaemmerer D, Sänger J, Schulz S, Lupp A. Somatostatin and chemokine CXCR4 receptor expression in pancreatic adenocarcinoma relative to pancreatic neuroendocrine tumours. J Cancer Res Clin Oncol 2019; 145:2481-2493. [PMID: 31451931 DOI: 10.1007/s00432-019-03011-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 08/21/2019] [Indexed: 02/07/2023]
Abstract
PURPOSE Pancreatic adenocarcinoma (PAC) represents one of the most fatal types of cancer with an exceptionally poor prognosis, underscoring the need for improved diagnostic and treatment approaches. An over-expression of somatostatin receptors (SST) as well as of the chemokine receptor CXCR4 has been shown for many tumour entities. Respective expression data for PAC, however, are scarce and contradictory. METHODS Overall, 137 tumour samples from 70 patients, 26 of whom were diagnosed with PAC and 44 with pancreatic neuroendocrine tumour (PanNET), were compared in terms of SST and CXCR4 expression by immunohistochemical analysis using well-characterized rabbit monoclonal antibodies. RESULTS Only SST1 and CXCR4 expression was detected in PAC tumours, with SST1 present in 42.3% and CXCR4 in 7.7% of cases. However, the overall staining intensity was very weak. In contrast to the tumour cells, in many PAC cases, tumour capillaries exhibited strong SST3, SST5, or CXCR4 expression. In PanNETs, SST2 was the most-prominently expressed receptor, observed in 75.0% of the tumours at medium-strong intensity. SST5, SST1, and CXCR4 expression was detected in 20.5%, 15.9%, and 11.4% of PanNET cases, respectively, but the staining intensity was only weak. SST2 positivity in PanNET, but not in PAC, was associated with favourable patient outcomes. CONCLUSIONS SST or CXCR4 expression in PAC is clearly of no therapeutic relevance. However, indirect targeting of these tumours via SST3, SST5, or CXCR4 on tumour microvessels may represent a promising additional therapeutic strategy.
Collapse
Affiliation(s)
- Ylberta Kajtazi
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Drackendorfer Str. 1, 07747, Jena, Germany
| | - Daniel Kaemmerer
- Department of General and Visceral Surgery, Zentralklinik Bad Berka, Bad Berka, Germany
| | - Jörg Sänger
- Laboratory of Pathology and Cytology Bad Berka, Bad Berka, Germany
| | - Stefan Schulz
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Drackendorfer Str. 1, 07747, Jena, Germany
| | - Amelie Lupp
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Drackendorfer Str. 1, 07747, Jena, Germany.
| |
Collapse
|
11
|
Storvall S, Leijon H, Ryhänen E, Louhimo J, Haglund C, Schalin-Jäntti C, Arola J. Somatostatin receptor expression in parathyroid neoplasms. Endocr Connect 2019; 8:1213-1223. [PMID: 31336364 PMCID: PMC6709562 DOI: 10.1530/ec-19-0260] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 07/23/2019] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Parathyroid carcinoma represents a rare cause of primary hyperparathyroidism. Distinguishing carcinoma from the benign tumors underlying primary hyperparathyroidism remains challenging. The diagnostic criteria for parathyroid carcinoma are local and/or metastatic spreading. Atypical parathyroid adenomas share other histological features with carcinomas but lack invasive growth. Somatostatin receptors are commonly expressed in different neuroendocrine tumors, but whether this also holds for parathyroid tumors remains unknown. AIM Our aim is to examine the immunohistochemical expression of somatostatin receptor 1-5 in parathyroid typical adenomas, atypical adenomas and carcinomas. METHODS We used a tissue microarray construct from a nationwide cohort of parathyroid carcinomas (n = 32), age- and gender-matched typical parathyroid adenomas (n = 72) and atypical parathyroid adenomas (n = 27) for immunohistochemistry of somatostatin receptor subtypes 1-5. We separately assessed cytoplasmic, membrane and nuclear expression and also investigated the associations with histological, biochemical and clinical characteristics. RESULTS All parathyroid tumor subgroups expressed somatostatin receptors, although membrane expression appeared negligible. Except for somatostatin receptor 1, expression patterns differed between the three tumor types. Adenomas exhibited the weakest and carcinomas the strongest expression of somatostatin receptor 2, 3, 4 and 5. We observed the largest difference for cytoplasmic somatostatin receptor 5 expression. CONCLUSIONS Parathyroid adenomas, atypical adenomas and carcinomas all express somatostatin receptor subtypes 1-5. Somatostatin receptor 5 may serve as a potential tumor marker for malignancy. Studies exploring the role of somatostatin receptor imaging and receptor-specific therapies in patients with parathyroid carcinomas are needed.
Collapse
Affiliation(s)
- Sara Storvall
- Department of Endocrinology, Abdominal Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Helena Leijon
- Department of Pathology and Huslab, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Eeva Ryhänen
- Department of Endocrinology, Abdominal Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Johanna Louhimo
- Department of Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Caj Haglund
- Department of Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Camilla Schalin-Jäntti
- Department of Endocrinology, Abdominal Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Johanna Arola
- Department of Pathology and Huslab, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Correspondence should be addressed to J Arola:
| |
Collapse
|
12
|
Remes SM, Leijon HL, Vesterinen TJ, Arola JT, Haglund CH. Immunohistochemical Expression of Somatostatin Receptor Subtypes in a Panel of Neuroendocrine Neoplasias. J Histochem Cytochem 2019; 67:735-743. [PMID: 31381461 DOI: 10.1369/0022155419856900] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Neuroendocrine neoplasias (NENs) are known to express somatostatin receptors (SSTRs) 1-5, which are G-protein-coupled cell membrane receptors. Somatostatin receptor imaging and therapy utilizes the SSTR expression. Synthetic somatostatin analogs with radioligands are used to detect primary tumors, metastases, and recurrent disease. Receptor analogs are also used for treating NENs. Furthermore, commercially available SSTR antibodies can be used for the immunohistochemical (IHC) detection of SSTRs. We investigated different SSTR antibody clones applying diverse IHC protocol settings to identify reliable clones and feasible protocols for NENs. A tissue microarray including NENs from 12 different primary sites were stained. Only UMB clones were able to localize SSTR on the cell membranes of NENs. SSTR2 (UMB1) emerged as the most common subtype followed by SSTR5 (UMB4) and SSTR1 (UMB7). SSTR3 (UMB5) expression was mainly cytoplasmic. Yet, SSTR4 expression was weak and located primarily in the cytoplasm. Thus, appropriate IHC protocols, including proper positive and negative controls, represent requirements for high-quality NEN diagnostics and for planning personalized therapy.
Collapse
Affiliation(s)
- Satu M Remes
- HUSLAB, Department of Pathology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Helena L Leijon
- HUSLAB, Department of Pathology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Tiina J Vesterinen
- HUSLAB, Department of Pathology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Johanna T Arola
- HUSLAB, Department of Pathology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Caj H Haglund
- Department of Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
13
|
Trouillas J, Vasiljevic A, Lapoirie M, Chinezu L, Jouanneau E, Raverot G. Pathological markers of somatotroph pituitary neuroendocrine tumors predicting the response to medical treatment. MINERVA ENDOCRINOL 2019; 44:129-136. [DOI: 10.23736/s0391-1977.18.02933-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
14
|
Treppiedi D, Giardino E, Catalano R, Mangili F, Vercesi P, Sala E, Locatelli M, Arosio M, Spada A, Mantovani G, Peverelli E. Somatostatin analogs regulate tumor corticotrophs growth by reducing ERK1/2 activity. Mol Cell Endocrinol 2019; 483:31-38. [PMID: 30611770 DOI: 10.1016/j.mce.2018.12.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 12/11/2018] [Accepted: 12/31/2018] [Indexed: 12/31/2022]
Abstract
Pasireotide has been associated with tumor shrinkage in patients with Cushing's disease subjected to long term treatment. However, to date the implicated molecular mechanisms are poorly elucidated. Here, we tested pasireotide-mediated cytostatic and cytotoxic effects in ACTH-secreting primary tumor cultures and murine corticotroph tumor cell line, AtT-20 cells. We found somatostatin receptor type 5 (SST5) expressed in 17 different ACTH-secreting tumors and SST2 detectable in 15 out of the 17 tissues. Pasireotide caused a slight but significant in vitro inhibition of cell growth in 3 out of 6 ACTH-secreting primary cultures (-12.1 ± 4.3%, P < 0.01 at 10 nM), remarkably reduced phospho-ERK1/2 levels in 5 out of 8 samples (-36.4 ± 20.5%, P < 0.01 at 1 μM) and triggered an increase of caspase 3/7 activity in 2 of 4 tumors (17 ± 3.6%, P < 0.05 at 1 μM). Accordingly, in AtT-20 cells, pasireotide significantly inhibited cell proliferation (-10.5 ± 7.7% at 10 nM, P < 0.05; -13.9 ± 10.9% at 100 nM, P < 0.05; -26.8 ± 8.9% at 1 μM, P < 0.01). Similar antiproliferative actions were exerted by BIM23206 and BIM23120 (SST5&2 selective ligands, respectively), whereas octreotide was effective when used at 1 μM (-13.3 ± 9.1%, P < 0.05). Moreover, a reduction of phospho-ERK1/2 was observed upon pasireotide and BIM23206 treatment (-8.4 ± 28.6%, P < 0.01 and -51.4 ± 15.9%, P < 0.001 at 10 nM, respectively) but not after octreotide and BIM23120 incubation. Finally, pasireotide was able to induce cell apoptosis in AtT-20 cells at lower concentration than octreotide. Altogether these data indicate a downstream implication of SST5-mediated phospho-ERK1/2 inhibition by pasireotide resulting in ACTH-secreting tumor cells proliferation reduction. Moreover, we describe for the first time a pro-apoptotic effect of pasireotide in corticotrophs.
Collapse
Affiliation(s)
- Donatella Treppiedi
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Endocrine Unit, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Elena Giardino
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Endocrine Unit, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Rosa Catalano
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Endocrine Unit, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy; PhD Program in Endocrinological Sciences, Sapienza University of Rome, Rome, Italy
| | - Federica Mangili
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Endocrine Unit, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Pietro Vercesi
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Endocrine Unit, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Elisa Sala
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Endocrine Unit, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Marco Locatelli
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neurosurgery Unit, Milan, Italy
| | - Maura Arosio
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Endocrine Unit, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Anna Spada
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Endocrine Unit, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Giovanna Mantovani
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Endocrine Unit, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy.
| | - Erika Peverelli
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Endocrine Unit, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| |
Collapse
|
15
|
Günther T, Tulipano G, Dournaud P, Bousquet C, Csaba Z, Kreienkamp HJ, Lupp A, Korbonits M, Castaño JP, Wester HJ, Culler M, Melmed S, Schulz S. International Union of Basic and Clinical Pharmacology. CV. Somatostatin Receptors: Structure, Function, Ligands, and New Nomenclature. Pharmacol Rev 2019; 70:763-835. [PMID: 30232095 PMCID: PMC6148080 DOI: 10.1124/pr.117.015388] [Citation(s) in RCA: 152] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Somatostatin, also known as somatotropin-release inhibitory factor, is a cyclopeptide that exerts potent inhibitory actions on hormone secretion and neuronal excitability. Its physiologic functions are mediated by five G protein-coupled receptors (GPCRs) called somatostatin receptor (SST)1-5. These five receptors share common structural features and signaling mechanisms but differ in their cellular and subcellular localization and mode of regulation. SST2 and SST5 receptors have evolved as primary targets for pharmacological treatment of pituitary adenomas and neuroendocrine tumors. In addition, SST2 is a prototypical GPCR for the development of peptide-based radiopharmaceuticals for diagnostic and therapeutic interventions. This review article summarizes findings published in the last 25 years on the physiology, pharmacology, and clinical applications related to SSTs. We also discuss potential future developments and propose a new nomenclature.
Collapse
Affiliation(s)
- Thomas Günther
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Giovanni Tulipano
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Pascal Dournaud
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Corinne Bousquet
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Zsolt Csaba
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Hans-Jürgen Kreienkamp
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Amelie Lupp
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Márta Korbonits
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Justo P Castaño
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Hans-Jürgen Wester
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Michael Culler
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Shlomo Melmed
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| | - Stefan Schulz
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P.C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.)
| |
Collapse
|
16
|
Yamamoto M, Ben-Shlomo A, Kameda H, Fukuoka H, Deng N, Ding Y, Melmed S. Somatostatin receptor subtype 5 modifies hypothalamic-pituitary-adrenal axis stress function. JCI Insight 2018; 3:122932. [PMID: 30282821 PMCID: PMC6237446 DOI: 10.1172/jci.insight.122932] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 08/29/2018] [Indexed: 11/17/2022] Open
Abstract
Pituitary corticotroph somatostatin receptor subtype 5 (SSTR5) signals to inhibit adrenocorticotrophin (ACTH) secretion. As ACTH deficiency results in attenuated adrenal cortisol production and an impaired stress response, we sought to clarify the role of SSTR5 in modifying the hypothalamic/pituitary/adrenal (HPA) axis. We generated Tg HP5 mice overexpressing SSTR5 in pituitary corticotrophs that produce the ACTH precursor proopiomelanocortin (POMC). Basal ACTH and corticosterone were similar in HP5 and WT mice, while HP5 mice showed attenuated ACTH and corticosterone responses to corticotrophin releasing hormone (CRH). HP5 mice exhibited attenuated corticosterone responses upon a restraint stress test and inflammatory stress following LPS injection, as well as increased anxiety-like and depressive-like behavior on open field and forced swim tests. Pituitary corticotroph CRH receptor subtype 1 (CRHR1) mRNA expression and ACTH responses to CRH were also attenuated in HP5 mice. In AtT20 cells stably overexpressing SSTR5, CRHR1 expression and cAMP response to CRH were reduced, whereas both were increased after SSTR5 KO. In elucidating mechanisms for these observations, we show that SSTR5-induced miR-449c suppresses both CRHR1 expression and function. We conclude that corticotroph SSTR5 attenuates HPA axis responses via CRHR1 downregulation, suggesting a role for SSTR5 in the pathogenesis of secondary adrenal insufficiency.
Collapse
Affiliation(s)
| | | | | | | | - Nan Deng
- Biostatistics and Bioinformatics Research Center, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Yan Ding
- Pituitary Center, Department of Medicine, and
| | | |
Collapse
|
17
|
Stumpf C, Kaemmerer D, Neubauer E, Sänger J, Schulz S, Lupp A. Somatostatin and CXCR4 expression patterns in adenocarcinoma and squamous cell carcinoma of the lung relative to small cell lung cancer. J Cancer Res Clin Oncol 2018; 144:1921-1932. [PMID: 30076481 DOI: 10.1007/s00432-018-2722-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 07/30/2018] [Indexed: 02/06/2023]
Abstract
PURPOSE Lung cancer is highly prevalent and has an especially poor prognosis. Thus, new diagnostic and therapeutic targets are necessary. Two potential targets are somatostatin receptors (SST), which are overexpressed in well-differentiated neuroendocrine neoplasms, and the chemokine receptor CXCR4, which is present mainly in highly proliferative and advanced tumours. Although their expression is relatively well characterized in small cell lung cancer (SCLC), in non-small cell lung cancer (NSCLC), data on SST and CXCR4 expression are scarce and contradictory. METHODS We comparatively evaluated 83 tumour samples from a total of 57 lung cancer patients, of which 22 had adenocarcinoma (ADC), 21 had squamous cell carcinoma (SQC), and 15 had SCLC. Samples were evaluated for SST and CXCR4 expression using immunohistochemistry with well-characterized rabbit monoclonal antibodies. RESULTS In the samples investigated, the most prominently expressed receptors were CXCR4 and SST5. Specifically, CXCR4 was detected with high expression intensity in more than 60% of ADC samples, about 90% of SQC, and 100% of SCLC. SST5 was present in about 75% of ADC and SQC samples and in more than 90% of SCLC. Although not noticeably expressed in ADC and SQC samples, SST2 was detected in 50% of SCLC cases, with a subset of patients displaying exceptionally high expression. The comparison of the three tumour entities revealed that SCLC samples had higher SST2, SST5, and CXCR4 expression, but lower SST3 and SST1 relative to ADC or SQC samples. CONCLUSION CXCR4 may be a promising target for diagnostics and therapy in both SCLC and NSCLC.
Collapse
Affiliation(s)
- Claudia Stumpf
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Drackendorfer Str. 1, 07747, Jena, Germany
| | - Daniel Kaemmerer
- Department of General and Visceral Surgery, Zentralklinik Bad Berka, Bad Berka, Germany
| | - Elisa Neubauer
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Drackendorfer Str. 1, 07747, Jena, Germany
| | - Jörg Sänger
- Institute of Pathology and Cytology Bad Berka, Bad Berka, Germany
| | - Stefan Schulz
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Drackendorfer Str. 1, 07747, Jena, Germany
| | - Amelie Lupp
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Drackendorfer Str. 1, 07747, Jena, Germany.
| |
Collapse
|
18
|
Immunohistochemical expression of somatostatin receptors SSTR2A and SSTR5 in 299 pituitary adenomas. Pathology 2018; 50:472-474. [DOI: 10.1016/j.pathol.2017.10.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Revised: 10/14/2017] [Accepted: 10/22/2017] [Indexed: 11/23/2022]
|
19
|
Lange F, Kaemmerer D, Behnke-Mursch J, Brück W, Schulz S, Lupp A. Differential somatostatin, CXCR4 chemokine and endothelin A receptor expression in WHO grade I-IV astrocytic brain tumors. J Cancer Res Clin Oncol 2018; 144:1227-1237. [PMID: 29696364 DOI: 10.1007/s00432-018-2645-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Accepted: 04/18/2018] [Indexed: 12/13/2022]
Abstract
PURPOSE Glioblastomas represent the most common primary malignant tumor of the nervous system and the most frequent type of astrocytic tumors. Despite improved therapeutic options, prognosis has remained exceptionally poor over the last two decades. Therefore, new treatment approaches are urgently needed. An overexpression of somatostatin (SST) as well as chemokine CXCR4 and endothelin A (ETA) receptors has been shown for many types of cancer. Respective expression data for astrocytic brain tumors, however, are scarce and contradictory. METHODS SST subtype, CXCR4 and ETA expression was comparatively evaluated in a total of 57 grade I-IV astrocytic tumor samples by immunohistochemistry using well-characterized monoclonal antibodies. RESULTS Overall, receptor expression on the tumor cells was only very low. SST5 was the most prominently expressed receptor, followed by SST3, ETA, SST2 and CXCR4. In contrast, tumor capillaries displayed strong SST2, SST3, SST5, CXCR4 and ETA expression. Presence of SST5, CXCR4 and ETA on tumor cells and of SST3, CXCR4 and ETA on microvessels gradually increased from grade II to grade IV tumors. Ki-67 values correlated significantly with CXCR4 expression on tumor cells and with vascular SST3, CXCR4 or ETA positivity. SST5 or CXCR4 positivity of tumor cells and vascular SST3 or CXCR4 expression negatively correlated with patient outcome. CONCLUSIONS Though having some prognostic value, SST, CXCR4 or ETA expression on astrocytic tumor cells is clearly of no therapeutic relevance. Indirect targeting of these highly vascularized tumors via SST3, SST5, CXCR4 or ETA on the microvessels, in contrast, may represent a promising additional therapeutic strategy.
Collapse
Affiliation(s)
- Franziska Lange
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Drackendorfer Str. 1, 07747, Jena, Germany
| | - Daniel Kaemmerer
- Department of General and Visceral Surgery, Zentralklinik Bad Berka, Bad Berka, Germany
| | | | - Wolfgang Brück
- Institute of Pathology, University Medical Centre Göttingen, University of Göttingen, Göttingen, Germany
| | - Stefan Schulz
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Drackendorfer Str. 1, 07747, Jena, Germany
| | - Amelie Lupp
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Drackendorfer Str. 1, 07747, Jena, Germany.
| |
Collapse
|
20
|
Werner C, Lupp A, Drescher R, Freesmeyer M, Mireskandari M, Stoykow C, Bauschke A, Müller UA. Morphologically ‘invisible’ proinsulin - secreting adenoma detected by Ga-68 Exendin-4 (GLP-1 Receptor) positron emission tomography/CT. J Med Imaging Radiat Oncol 2018; 62:370-374. [DOI: 10.1111/1754-9485.12704] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Accepted: 12/27/2017] [Indexed: 11/30/2022]
Affiliation(s)
- Christoph Werner
- Division of Endocrinology and Metabolic Diseases; Clinic of Internal Medicine III; Jena University Hospital; Jena Germany
| | - Amelie Lupp
- Institute of Pharmacology and Toxicology; Jena University Hospital; Jena Germany
| | - Robert Drescher
- Clinic of Nuclear Medicine; Jena University Hospital; Jena Germany
| | | | | | - Christian Stoykow
- Department of Nuclear Medicine; University Hospital Freiburg; Freiburg Germany
| | - Astrid Bauschke
- Clinic of General-, Abdominal and Vascular Surgery; Jena University Hospital; Jena Germany
| | - Ulrich A Müller
- Division of Endocrinology and Metabolic Diseases; Clinic of Internal Medicine III; Jena University Hospital; Jena Germany
| |
Collapse
|
21
|
Kaemmerer D, Schindler R, Mußbach F, Dahmen U, Altendorf-Hofmann A, Dirsch O, Sänger J, Schulz S, Lupp A. Somatostatin and CXCR4 chemokine receptor expression in hepatocellular and cholangiocellular carcinomas: tumor capillaries as promising targets. BMC Cancer 2017; 17:896. [PMID: 29282035 PMCID: PMC5745780 DOI: 10.1186/s12885-017-3911-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 12/13/2017] [Indexed: 12/15/2022] Open
Abstract
Background Hepatocellular (HCC) and cholangiocellular carcinomas (CCC) display an exceptionally poor prognosis. Especially for advanced disease no efficient standard therapy is currently available. Recently, somatostatin analogs have been evaluated for the treatment of HCC, however, with contradictory results. Besides, for both malignancies the chemokine receptor CXCR4 has been discussed as a possible new target structure. Methods Expression of somatostatin receptor (SSTR) subtypes 1, 2A, 3, 4, and 5, and of CXCR4 was evaluated in a total of 71 HCCs and 27 CCCs by immunohistochemistry using well-characterized novel monoclonal antibodies. Results In HCC tumor cells, frequency and intensity of expression of SSTRs and CXCR4 were only low. CXCR4 was present in about 40% of the HCCs, although at a low intensity. SSTR5, SSTR2, and SSTR3 were detected in about 15%, 8%, and 5% of the HCC tumors, respectively. SSTR and CXCR4 expression was much higher in CCC than in HCC. CXCR4 and SSTR1 were present in 60% and 67% of the CCC samples, respectively, followed by SSTR2 and SSTR5, which were detected in 30% and 11% of the tumors, respectively. Most notably, CXCR4 was intensely expressed on the tumor capillaries in about 50% of the HCCs and CCCs. CXCR4 expression on tumor vessels was associated with poor patient outcomes. Conclusions CCC, but not HCC, may be suitable for SSTR-based treatments. Because of the predominant expression of SSTR1, pan-somatostatin analogs should be preferred. In both HCC and CCC, indirect targeting of tumors via the CXCR4-positive tumor capillaries may represent a promising additional therapeutic strategy. Electronic supplementary material The online version of this article (10.1186/s12885-017-3911-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Daniel Kaemmerer
- Department of General and Visceral Surgery, Zentralklinik Bad Berka, Bad Berka, Germany
| | - Robin Schindler
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Drackendorfer Str. 1, D-07747, Jena, Germany
| | - Franziska Mußbach
- Department of General, Visceral and Vascular Surgery, Jena University Hospital, Jena, Germany
| | - Uta Dahmen
- Department of General, Visceral and Vascular Surgery, Jena University Hospital, Jena, Germany
| | | | - Olaf Dirsch
- Institute of Pathology, Jena University Hospital, Jena, Germany
| | - Jörg Sänger
- Institute of Pathology and Cytology Bad Berka, Bad Berka, Germany
| | - Stefan Schulz
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Drackendorfer Str. 1, D-07747, Jena, Germany
| | - Amelie Lupp
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Drackendorfer Str. 1, D-07747, Jena, Germany.
| |
Collapse
|
22
|
Abstract
The carcinoid as originally described is part of the relatively large family of neuroendocrine neoplasia found in almost every organ. Historical reasons back their current definitions. Neuroendocrine cancer is most frequently observed in the lung and the digestive tract. In the lung is defined as carcinoid (typical and atypical) for well differentiated, low to intermediate grade, and small cell and large cell neuroendocrine carcinoma for poorly differentiated, high grade. In the digestive system are respectively defined as neuroendocrine tumor (NET) and neuroendocrine carcinoma (NEC) of small and large cell types. Grading and staging are developed for their clinical classification by the World Health Organization (WHO) and the American Joint Committee on Cancer (AJCC). In both anatomical sites the morphological features are overlapping, with bland histology for carcinoid and NET, and aggressive features with extensive necrosis, severe atypia and abundant, atypical mitoses for high grade cancer types. Such features are also essential diagnostic clues in cytological preparations. The confirmation of the neuroendocrine signature by immunohistochemistry is mandatory for the diagnosis; a minimum panel comprising chromogranin A and synaptophysin is recommended in the digestive system. In addition, the application of grading requires the mitotic count and or spotty necrosis assessment for lung, or the mitotic count and the Ki67 assessment in the digestive system.
Collapse
Affiliation(s)
- Frediano Inzani
- Institute of Anatomic Pathology, Università Cattolica del Sacro Cuore-Fondazione Policlinico Universitario Agostino Gemelli, Largo A. Gemelli, 8, 00168, Rome, Italy
- Roma European NeuroEndocrine Tumor Society (ENETS) Center of Excellence, Rome, Italy
| | - Gianluigi Petrone
- Institute of Anatomic Pathology, Università Cattolica del Sacro Cuore-Fondazione Policlinico Universitario Agostino Gemelli, Largo A. Gemelli, 8, 00168, Rome, Italy
- Roma European NeuroEndocrine Tumor Society (ENETS) Center of Excellence, Rome, Italy
| | - Guido Fadda
- Institute of Anatomic Pathology, Università Cattolica del Sacro Cuore-Fondazione Policlinico Universitario Agostino Gemelli, Largo A. Gemelli, 8, 00168, Rome, Italy
- Roma European NeuroEndocrine Tumor Society (ENETS) Center of Excellence, Rome, Italy
| | - Guido Rindi
- Institute of Anatomic Pathology, Università Cattolica del Sacro Cuore-Fondazione Policlinico Universitario Agostino Gemelli, Largo A. Gemelli, 8, 00168, Rome, Italy.
- Roma European NeuroEndocrine Tumor Society (ENETS) Center of Excellence, Rome, Italy.
- Institute of Anatomic Pathology, Università Cattolica - Policlinico A. Gemelli, Largo A. Gemelli, 8, I-00168, Rome, Italy.
| |
Collapse
|
23
|
Kasajima A, Papotti M, Ito W, Brizzi MP, La Salvia A, Rapa I, Tachibana T, Yazdani S, Sasano H, Volante M. High interlaboratory and interobserver agreement of somatostatin receptor immunohistochemical determination and correlation with response to somatostatin analogs. Hum Pathol 2017; 72:144-152. [PMID: 29180250 DOI: 10.1016/j.humpath.2017.11.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 11/13/2017] [Accepted: 11/17/2017] [Indexed: 01/12/2023]
Abstract
Monoclonal antibodies to somatostatin receptors 2A (SSTR2A, UMB-1) and 5 (SSTR5, UMB-4) were reported to be highly reliable for immunohistochemical detection of these receptors in neuroendocrine neoplasms. However, the standardization of either the immunohistochemical procedure and the methods of evaluation has yet to be established. Fifty-two tissues from 38 patients with neuroendocrine neoplasm were retrieved from 2 institutions in Italy and Japan. The tissues were immunostained using 3 staining methodologies: 1 automated and 2 manual protocols from the Italian and Japanese institutions. The slides were independently evaluated by 3 observers (2 experienced pathologists and 1 medical student) using 3 scoring systems (Volante-Score, HER2-Score, and H-Score). The scores obtained from the staining methods were highly correlated with each other (r>0.85, P<.0001). Especially, the Volante- and HER2-Scores were highly concordant (r≥0.95, P≤.0001). Very high interobserver agreement was obtained irrespective of the method used and the experience of the evaluator, with the best concordance obtained by experienced pathologists evaluating automated system-stained slides (SSTR2A, r>0.97; SSTR5, r>0.96). HER2- and H-Scores were reliable to represent the characteristics of the patients. SSTR2A expression evaluated by the HER2-Score was significantly associated with clinical efficacy to somatostatin analogs (P=.04). SSTRs determination is an easily standardizable tool in different laboratories and is highly reproducible irrespective of the method of evaluation used. Given the positive association with clinical efficacy to somatostatin analogs, as well as the simple and widespread use, HER2-Score can be proposed as a standard evaluation procedure of SSTR2A and SSTR5 expression in neuroendocrine neoplasms.
Collapse
Affiliation(s)
- Atsuko Kasajima
- Department of Pathology, Tohoku University Graduate School of Medicine, 980-8574 Sendai, Japan; Department of Pathology, Technical University Munich, 81675 Munich, Germany.
| | - Mauro Papotti
- Department of Oncology, University of Turin at San Luigi Hospital, Orbassano, 10043 Turin, Italy; Città della Salute Hospital, Orbassano, 10126 Turin, Italy.
| | - Wataru Ito
- Department of Pathology, Tohoku University Graduate School of Medicine, 980-8574 Sendai, Japan.
| | - Maria Pia Brizzi
- Oncology Unit, San Luigi Hospital, Orbassano, 10043 Turin, Italy.
| | - Anna La Salvia
- Oncology Unit, San Luigi Hospital, Orbassano, 10043 Turin, Italy.
| | - Ida Rapa
- Department of Oncology, University of Turin at San Luigi Hospital, Orbassano, 10043 Turin, Italy.
| | - Tomoyoshi Tachibana
- Department of Pathology, Tohoku University Graduate School of Medicine, 980-8574 Sendai, Japan.
| | - Samaneh Yazdani
- Department of Pathology, Tohoku University Graduate School of Medicine, 980-8574 Sendai, Japan.
| | - Hironobu Sasano
- Department of Pathology, Tohoku University Graduate School of Medicine, 980-8574 Sendai, Japan.
| | - Marco Volante
- Department of Oncology, University of Turin at San Luigi Hospital, Orbassano, 10043 Turin, Italy.
| |
Collapse
|
24
|
Kaemmerer D, Sänger J, Arsenic R, D’Haese JG, Neumann J, Schmitt-Graeff A, Wirtz RM, Schulz S, Lupp A. Evaluation of somatostatin, CXCR4 chemokine and endothelin A receptor expression in a large set of paragangliomas. Oncotarget 2017; 8:89958-89969. [PMID: 29163802 PMCID: PMC5685723 DOI: 10.18632/oncotarget.21194] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 08/17/2017] [Indexed: 12/11/2022] Open
Abstract
Paragangliomas are predominantly benign tumors, but in some cases invasive growth and also metastasis are observed. Given the limited number of nonsurgical treatment options, novel target structures for diagnostics and therapy of this tumor entity are urgently needed. In the present study, expression of all five somatostatin receptor (SST) subtypes, chemokine receptor CXCR4 and endothelin receptor type A (ETA) was assessed by means of immunohistochemistry in a total of 66 paraffin-embedded paraganglioma samples from 55 patients. The stainings were rated by means of the Immunoreactive Score and correlated to clinical data and to succinate dehydrogenase subunit B (SDHB) expression. SST2A was by far the most prominent receptor in the paragangliomas investigated. It was present in 89% of the tumors at a high intensity, followed by SST5, SST3, SST1 and SST4, which were detected in 47%, 35%, 35% and 13% of the samples, respectively. SDHB positive tumors exhibited significantly higher SST2A and SST3 expression as compared to SDHB negative cases. There was no correlation between SST and Ki-67 expression or grading of the tumors and no difference in SST expression between primary tumors and metastases. Cell surface expression of CXCR4 and ETA was detected only in few samples. On tumor capillaries, however, exceptionally strong staining for these two receptors was noticed in the vast majority of the tumors. In conclusion, paragangliomas are well suited for SST2A-based diagnostics and treatment modalities. An indirect targeting of these highly vascularized tumors via CXCR4 or ETA may also represent a promising future strategy.
Collapse
Affiliation(s)
- Daniel Kaemmerer
- Department of General and Visceral Surgery, Zentralklinik Bad Berka, Bad Berka, Germany
| | - Jörg Sänger
- Laboratory of Pathology and Cytology Bad Berka, Bad Berka, Germany
| | - Ruza Arsenic
- Institute of Pathology, Charité University Hospital Berlin, Berlin, Germany
| | - Jan G. D’Haese
- Department of General, Visceral, Transplantation, Vascular and Thoracic Surgery, Hospital of the University of Munich, Munich, Germany
| | - Jens Neumann
- Department of Pathology, Ludwig-Maximilians-University Munich, Munich, Germany
| | | | | | - Stefan Schulz
- Institute of Pharmacology and Toxicology, Jena University Hospital, Jena, Germany
| | - Amelie Lupp
- Institute of Pharmacology and Toxicology, Jena University Hospital, Jena, Germany
| |
Collapse
|
25
|
The components of somatostatin and ghrelin systems are altered in neuroendocrine lung carcinoids and associated to clinical-histological features. Lung Cancer 2017; 109:128-136. [PMID: 28577942 DOI: 10.1016/j.lungcan.2017.05.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 05/02/2017] [Accepted: 05/07/2017] [Indexed: 12/20/2022]
Abstract
BACKGROUND Lung carcinoids (LCs) are rare tumors that comprise 1-5% of lung malignancies but represent 20-30% of neuroendocrine tumors. Their incidence is progressively increasing and a better characterization of these tumors is required. Alterations in somatostatin (SST)/cortistatin (CORT) and ghrelin systems have been associated to development/progression of various endocrine-related cancers, wherein they may become useful diagnostic, prognostic and therapeutic biomarkers. OBJECTIVES We aimed to evaluate the expression levels of ghrelin and SST/CORT system components in LCs, as well as to explore their putative relationship with histological/clinical characteristics. PATIENTS AND METHODS An observational retrospective study was performed; 75 LC patients with clinical/histological characteristics were included. Samples from 46 patients were processed to isolate mRNA from tumor and adjacent non-tumor region, and the expression levels of SST/CORT and ghrelin systems components, determined by quantitative-PCR, were compared to those of 7 normal lung tissues. RESULTS Patient cohort was characterized by mean age 53±15 years, 48% males, 34% with tobacco exposure; 71.4/28.6% typical/atypical carcinoids, 21.7% incidental tumors, 4.3% functioning tumors, 17.7% with metastasis. SST/CORT and ghrelin system components were expressed at variable levels in a high proportion of tumors, as well as in adjacent non-tumor tissues, while a lower proportion of normal lung samples also expressed these molecules. A gradation was observed from normal non-neoplastic lung tissues, non-tumor adjacent tissue and LCs, being SST, sst4, sst5, GHS-R1a and GHS-R1b overexpressed in tumor tissue compared to normal tissue. Importantly, several SST/CORT and ghrelin system components displayed significant correlations with relevant clinical parameters, such as necrosis, peritumoral and vascular invasion, or metastasis. CONCLUSION Altogether, these data reveal a prominent, widespread expression of key SST/CORT/ghrelin system components in LCs, where they display clinical-histological correlations, which could provide novel, valuable markers for NET patient management.
Collapse
|
26
|
Gatto F, Feelders RA, Franck SE, van Koetsveld PM, Dogan F, Kros JM, Neggers SJCMM, van der Lely AJ, Lamberts SWJ, Ferone D, Hofland LJ. In Vitro Head-to-Head Comparison Between Octreotide and Pasireotide in GH-Secreting Pituitary Adenomas. J Clin Endocrinol Metab 2017; 102:2009-2018. [PMID: 28323931 DOI: 10.1210/jc.2017-00135] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Accepted: 03/08/2017] [Indexed: 11/19/2022]
Abstract
CONTEXT First-generation somatostatin analogs (SSAs), such as octreotide (OCT), are the first line medical therapy for acromegaly. Pasireotide (PAS), a newly developed SSA, has shown promising results in the treatment of acromegaly. OBJECTIVE To compare the antisecretory effect of OCT and PAS in primary cultures of growth hormone (GH)-secreting pituitary adenomas (GH-omas). To correlate responses with the adenoma somatostatin receptor (SSTR) profile. DESIGN The effect of OCT and PAS on GH (and PRL) secretion was tested in 33 GH-oma cultures. SSTR expression was evaluated in adenoma samples. SETTING AND PATIENTS Patients with acromegaly referred to the Erasmus Medical Center (Rotterdam, The Netherlands). INTERVENTIONS OCT and PAS treatment for 72 hours (10 nM). MAIN OUTCOME MEASURES GH (and PRL) concentrations in cell culture media. SSTR expression in adenoma samples. RESULTS The overall effect of OCT (-36.8%) and PAS (-37.1%) on GH secretion was superimposable. We identified three adenoma groups: PAS+ (PAS more effective than OCT), n = 6; PAS = OCT, n = 22; and OCT+ (OCT more effective than PAS), n = 5. PAS+ adenomas showed lower somatostatin receptor subtype (sst)2 messenger RNA (mRNA) and sst2/sst5 mRNA ratio, compared with the other groups (P < 0.05). PAS inhibited PRL hypersecretion more than OCT (P < 0.01). CONCLUSIONS Overall, OCT and PAS equally reduced GH secretion in vitro. Adenomas with lower sst2 mRNA expression and lower sst2/sst5 mRNA ratio were better responders to PAS compared with OCT. SSTR evaluation in GH-omas may become a tool for tailored SSA treatment in acromegaly.
Collapse
Affiliation(s)
- Federico Gatto
- Division of Endocrinology, Department of Internal Medicine, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands
| | - Richard A Feelders
- Division of Endocrinology, Department of Internal Medicine, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands
- Pituitary Center Rotterdam, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands
| | - Sanne E Franck
- Division of Endocrinology, Department of Internal Medicine, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands
| | - Peter M van Koetsveld
- Division of Endocrinology, Department of Internal Medicine, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands
| | - Fadime Dogan
- Division of Endocrinology, Department of Internal Medicine, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands
| | - Johan M Kros
- Pituitary Center Rotterdam, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands
- Department of Pathology, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands
| | - Sebastian J C M M Neggers
- Division of Endocrinology, Department of Internal Medicine, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands
- Pituitary Center Rotterdam, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands
| | - Aart-Jan van der Lely
- Division of Endocrinology, Department of Internal Medicine, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands
- Pituitary Center Rotterdam, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands
| | - Steven W J Lamberts
- Division of Endocrinology, Department of Internal Medicine, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands
| | - Diego Ferone
- Section of Endocrinology, Department of Internal Medicine and Medical Specialties, Center of Excellence for Biomedical Research, University of Genova, Genova 16126, Italy
| | - Leo J Hofland
- Division of Endocrinology, Department of Internal Medicine, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands
- Pituitary Center Rotterdam, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands
| |
Collapse
|
27
|
Stollberg S, Kämmerer D, Neubauer E, Schulz S, Simonitsch-Klupp I, Kiesewetter B, Raderer M, Lupp A. Differential somatostatin and CXCR4 chemokine receptor expression in MALT-type lymphoma of gastric and extragastric origin. J Cancer Res Clin Oncol 2016; 142:2239-47. [PMID: 27544389 DOI: 10.1007/s00432-016-2220-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 08/15/2016] [Indexed: 12/17/2022]
Abstract
PURPOSE Whereas the different somatostatin receptor (SSTR) subtypes and the chemokine receptor CXCR4 are known to be expressed in a wide variety of human malignancies, comprehensive data are still lacking for MALT-type lymphomas. METHODS Overall, 55 cases of MALT-type lymphoma of both gastric and extragastric origin were evaluated for the SSTR subtype and CXCR4 expression by means of immunohistochemistry using novel monoclonal rabbit antibodies. The stainings were rated by means of the immunoreactive score and correlated with clinical data. RESULTS While the CXCR4 was detected in 92 % of the cases investigated, the SSTR subtypes were much less frequently present. The SSTR5 was expressed in about 50 % of the cases, followed by the SSTR3, the SSTR2A, the SSTR4 and the SSTR1, which were present in 35, 27, 18 or 2 %, respectively, of the tumors only. Gastric lymphomas displayed a significantly higher SSTR3, SSTR4 and SSTR5 expression than extragastric tumors. A correlation between CXCR4 and Ki-67 expression was seen in gastric lymphomas, whereas primarily in extragastric tumors SSTR5 negativity was associated with poor patient outcome. CONCLUSIONS The CXCR4 may serve as a promising target for diagnostics and therapy of MALT-type lymphomas, while the SSTRs appear not suitable in this respect.
Collapse
Affiliation(s)
- Susann Stollberg
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Drackendorfer Str. 1, 07747, Jena, Germany
| | - Daniel Kämmerer
- Department of General and Visceral Surgery, Zentralklinik Bad Berka, Bad Berka, Germany
| | - Elisa Neubauer
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Drackendorfer Str. 1, 07747, Jena, Germany
| | - Stefan Schulz
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Drackendorfer Str. 1, 07747, Jena, Germany
| | | | | | - Markus Raderer
- Department of Internal Medicine I, University of Vienna, Vienna, Austria
| | - Amelie Lupp
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Drackendorfer Str. 1, 07747, Jena, Germany.
| |
Collapse
|
28
|
Kaemmerer D, Träger T, Hoffmeister M, Sipos B, Hommann M, Sänger J, Schulz S, Lupp A. Inverse expression of somatostatin and CXCR4 chemokine receptors in gastroenteropancreatic neuroendocrine neoplasms of different malignancy. Oncotarget 2016; 6:27566-79. [PMID: 26259237 PMCID: PMC4695009 DOI: 10.18632/oncotarget.4491] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 07/03/2015] [Indexed: 12/30/2022] Open
Abstract
INTRODUCTION Somatostatin receptors (SSTR) are widely distributed in well-differentiated neuroendocrine neoplasms (NEN) and serve as primary targets for diagnostics and treatment. An overexpression of the chemokine receptor CXCR4, in contrast, is considered to be present mainly in highly proliferative and advanced tumors. Comparative data are still lacking, however, for neuroendocrine carcinomas (NEC). METHODS SSTR subtype (1, 2A, 3, 5) and CXCR4 expression was evaluated in G1 (n = 31), G2 (n = 47), and low (G3a; Ki-67: 21-49%; n = 21) and highly proliferative (G3b; Ki-67: >50%, n = 22) G3 (total n = 43) gastroenteropancreatic NEN samples by performing immunohistochemistry with monoclonal rabbit anti-human anti-SSTR and anti-CXCR4 antibodies, respectively, and was correlated with clinical data. RESULTS Both CXCR4 and SSTR were widely expressed in all tumors investigated. CXCR4 expression differed significantly between the G1 and G3 specimens and within the G3 group (G3a to G3b), and was positively correlated with Ki-67 expression. SSTR2A, in contrast, exhibited an inverse association with Ki-67. SSTR2A was highly expressed in G1 and G2 tumors, but was significantly less abundant in G3 carcinomas. Additionally, SSTR1 expression was higher in G3a than in G3b tumors. CONCLUSION We observed an elevation in CXCR4 and a decrease in SSTR2A expression with increasing malignancy. Interestingly, 23% of the G3 specimens had strong SSTR2A expression. Because CXCR4 was strongly expressed in highly proliferative G3 carcinomas, it is an interesting new target and needs to be validated in larger studies.
Collapse
Affiliation(s)
- Daniel Kaemmerer
- Department of General and Visceral Surgery, Zentralklinik Bad Berka, Bad Berka, Germany
| | - Tina Träger
- Department of General and Visceral Surgery, Zentralklinik Bad Berka, Bad Berka, Germany.,Department of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | | | - Bence Sipos
- Institute of Pathology, University Hospital Tuebingen, Germany
| | - Merten Hommann
- Department of General and Visceral Surgery, Zentralklinik Bad Berka, Bad Berka, Germany
| | - Jörg Sänger
- Institute of Pathology and Cytology, Bad Berka, Germany
| | - Stefan Schulz
- Department of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - Amelie Lupp
- Department of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| |
Collapse
|
29
|
Gatto F, Biermasz NR, Feelders RA, Kros JM, Dogan F, van der Lely AJ, Neggers SJCMM, Lamberts SWJ, Pereira AM, Ferone D, Hofland LJ. Low beta-arrestin expression correlates with the responsiveness to long-term somatostatin analog treatment in acromegaly. Eur J Endocrinol 2016; 174:651-62. [PMID: 26888629 DOI: 10.1530/eje-15-0391] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 02/17/2016] [Indexed: 12/18/2022]
Abstract
OBJECTIVE The high expression of somatostatin receptor subtype 2 (SSTR2 also known as sst2) usually present in growth hormone (GH)-secreting adenomas is the rationale for therapy with somatostatin analogs (SSAs) in acromegaly. Although SSTR2 expression is a good predictor for biochemical response to SSA treatment, we still face tumors resistant to SSAs despite high SSTR2 expression. Recently, beta-arrestins (β-arrestins) have been highlighted as key players in the regulation of SSTR2 function. DESIGN To investigate whether β-arrestins might be useful predictors of responsiveness to long-term SSA treatment in acromegaly, we retrospectively evaluated 35 patients with acromegaly who underwent adenomectomy in two referral centers in The Netherlands. METHODS β-arrestin mRNA levels were evaluated in adenoma samples, together with SSTR2 (and SSTR5) mRNA and protein expression. Biochemical response to long-term SSA treatment (median 12 months) was assessed in 32 patients. RESULTS β-arrestin 1 and 2 mRNA was significantly lower in adenoma tissues from patients who achieved insulin-like growth factor 1 normalization (P = 0.024 and P = 0.047) and complete biochemical control (P = 0.047 and P = 0.039). The SSTR2 mRNA was higher in SSA responder patients compared with the resistant ones (P = 0.026). This difference was more evident when analyzing the SSTR2/β-arrestin 1 and SSTR2/β-arrestin 2 ratio (P = 0.011 and P = 0.010). β-arrestin 1 and 2 expression showed a significant trend of higher median values from full responders, partial responders to resistant patients (P = 0.045 and P = 0.021, respectively). Interestingly, SSTR2 protein expression showed a strong inverse correlation with both β-arrestin 1 and 2 mRNA (ρ = -0.69, P = 0.0011 and ρ = -0.67, P = 0.0016). CONCLUSIONS Low β-arrestin expression and high SSTR2/β-arrestin ratio correlate with the responsiveness to long-term treatment with SSAs in patients with acromegaly.
Collapse
Affiliation(s)
- Federico Gatto
- Department of Internal MedicineDivision Endocrinology, Erasmus MC, Rotterdam, The Netherlands
| | - Nienke R Biermasz
- Department of MedicineDivision of Endocrinology and Center for Endocrine Tumors, Leiden University Medical Center, Leiden, The Netherlands
| | - Richard A Feelders
- Department of Internal MedicineDivision Endocrinology, Erasmus MC, Rotterdam, The Netherlands Pituitary Center RotterdamErasmus MC, Rotterdam, The Netherlands
| | - Johan M Kros
- PathologyErasmus MC, Rotterdam, The Netherlands Pituitary Center RotterdamErasmus MC, Rotterdam, The Netherlands
| | - Fadime Dogan
- Department of Internal MedicineDivision Endocrinology, Erasmus MC, Rotterdam, The Netherlands
| | - Aart-Jan van der Lely
- Department of Internal MedicineDivision Endocrinology, Erasmus MC, Rotterdam, The Netherlands Pituitary Center RotterdamErasmus MC, Rotterdam, The Netherlands
| | - Sebastian J C M M Neggers
- Department of Internal MedicineDivision Endocrinology, Erasmus MC, Rotterdam, The Netherlands Pituitary Center RotterdamErasmus MC, Rotterdam, The Netherlands
| | - Steven W J Lamberts
- Department of Internal MedicineDivision Endocrinology, Erasmus MC, Rotterdam, The Netherlands
| | - Alberto M Pereira
- Department of MedicineDivision of Endocrinology and Center for Endocrine Tumors, Leiden University Medical Center, Leiden, The Netherlands
| | - Diego Ferone
- EndocrinologyDepartment of Internal Medicine and Medical Specialties (DIMI) and Center of Excellence for Biomedical Research (CEBR), IRCCS AOU San Martino-IST, University of Genoa, Genoa, Italy
| | - Leo J Hofland
- Department of Internal MedicineDivision Endocrinology, Erasmus MC, Rotterdam, The Netherlands Pituitary Center RotterdamErasmus MC, Rotterdam, The Netherlands
| |
Collapse
|
30
|
Günther T, Culler M, Schulz S. Research Resource: Real-Time Analysis of Somatostatin and Dopamine Receptor Signaling in Pituitary Cells Using a Fluorescence-Based Membrane Potential Assay. Mol Endocrinol 2016; 30:479-90. [PMID: 26967369 DOI: 10.1210/me.2015-1241] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Stable somatostatin analogues and dopamine receptor agonists are the mainstay for the pharmacological treatment of functional pituitary adenomas; however, only a few cellular assays have been developed to detect receptor activation of novel compounds without disrupting cells to obtain the second messenger content. Here, we adapted a novel fluorescence-based membrane potential assay to characterize receptor signaling in a time-dependent manner. This minimally invasive technique provides a robust and reliable read-out for ligand-induced receptor activation in permanent and primary pituitary cells. The mouse corticotropic cell line AtT-20 endogenously expresses both the somatostatin receptors 2 (sst2) and 5 (sst5). Exposure of wild-type AtT-20 cells to the sst2- and sst5-selective agonists BIM-23120 and BIM-23268, respectively, promoted a pertussis toxin- and tertiapin-Q-sensitive reduction in fluorescent signal intensity, which is indicative of activation of G protein-coupled inwardly rectifying potassium (GIRK) channels. After heterologous expression, sst1, sst3, and sst4 receptors also coupled to GIRK channels in AtT-20 cells. Similar activation of GIRK channels by dopamine required overexpression of dopamine D2 receptors (D2Rs). Interestingly, the presence of D2Rs in AtT-20 cells strongly facilitated GIRK channel activation elicited by the sst2-D2 chimeric ligand BIM-23A760, suggesting a synergistic action of sst2 and D2Rs. Furthermore, stable somatostatin analogues produced strong responses in primary pituitary cultures from wild-type mice; however, in cultures from sst2 receptor-deficient mice, only pasireotide and somatoprim, but not octreotide, induced a reduction in fluorescent signal intensity, suggesting that octreotide mediates its pharmacological action primarily via the sst2 receptor.
Collapse
Affiliation(s)
- Thomas Günther
- Department of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, D-07749 Jena, Germany
| | - Michael Culler
- Department of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, D-07749 Jena, Germany
| | - Stefan Schulz
- Department of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, D-07749 Jena, Germany
| |
Collapse
|
31
|
Iacovazzo D, Carlsen E, Lugli F, Chiloiro S, Piacentini S, Bianchi A, Giampietro A, Mormando M, Clear AJ, Doglietto F, Anile C, Maira G, Lauriola L, Rindi G, Roncaroli F, Pontecorvi A, Korbonits M, De Marinis L. Factors predicting pasireotide responsiveness in somatotroph pituitary adenomas resistant to first-generation somatostatin analogues: an immunohistochemical study. Eur J Endocrinol 2016; 174:241-50. [PMID: 26586796 DOI: 10.1530/eje-15-0832] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Accepted: 11/19/2015] [Indexed: 01/02/2023]
Abstract
AIM To gather data regarding factors predicting responsiveness to pasireotide in acromegaly. PATIENTS AND METHODS SSTR2a, SSTR3, SSTR5, AIP, Ki-67 and the adenoma subtype were evaluated in somatotroph adenomas from 39 patients treated post-operatively with somatostatin analogues (SSAs). A standardized SSTR scoring system was applied (scores 0-3). All patients received first-generation SSAs, and 11 resistant patients were subsequently treated with pasireotide LAR. RESULTS None of the patients with negative or cytoplasmic-only SSTR2a expression (scores 0-1) were responsive to first-generation SSAs, as opposed to 20% (score 2) and 50% of patients with a score of 3 (P=0.04). None of the patients with an SSTR5 score of 0-1 were responsive to pasireotide, as opposed to 5/7 cases with a score of 2 or 3 (P=0.02). SSTR3 expression did not influence first-generation SSAs or pasireotide responsiveness. Tumours with low AIP were resistant to first-generation SSAs (100 vs 60%; P=0.02), while they had similar responsiveness to pasireotide compared to tumours with conserved AIP expression (50 vs 40%; P=0.74). Tumours with low AIP displayed reduced SSTR2 (SSTR2a scores 0-1 44.4 vs 6.7%; P=0.006) while no difference was seen in SSTR5 (SSTR5 scores 0-1 33.3 vs 23.3%; P=0.55). Sparsely granulated adenomas responded better to pasireotide compared to densely granulated ones (80 vs 16.7%; P=0.04). CONCLUSION The expression of SSTR5 might predict responsiveness to pasireotide in acromegaly. AIP deficient and sparsely granulated adenomas may benefit from pasireotide treatment. These results need to be confirmed in larger series of pasireotide-treated patients.
Collapse
Affiliation(s)
- Donato Iacovazzo
- EndocrinologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKPathologySTHF, N-3710 Skien, NorwayEndocrinologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyHaemato-OncologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKNeurosurgeryUniversità di Brescia, 25121 Brescia, ItalyNeurosurgeryUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyNeurosurgeryHumanitas, 20089 Milan, ItalyPathologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyBrainBehaviour and Mental Health, University of Manchester, M13 9PT Manchester, UK EndocrinologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKPathologySTHF, N-3710 Skien, NorwayEndocrinologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyHaemato-OncologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKNeurosurgeryUniversità di Brescia, 25121 Brescia, ItalyNeurosurgeryUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyNeurosurgeryHumanitas, 20089 Milan, ItalyPathologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyBrainBehaviour and Mental Health, University of Manchester, M13 9PT Manchester, UK
| | - Eivind Carlsen
- EndocrinologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKPathologySTHF, N-3710 Skien, NorwayEndocrinologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyHaemato-OncologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKNeurosurgeryUniversità di Brescia, 25121 Brescia, ItalyNeurosurgeryUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyNeurosurgeryHumanitas, 20089 Milan, ItalyPathologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyBrainBehaviour and Mental Health, University of Manchester, M13 9PT Manchester, UK
| | - Francesca Lugli
- EndocrinologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKPathologySTHF, N-3710 Skien, NorwayEndocrinologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyHaemato-OncologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKNeurosurgeryUniversità di Brescia, 25121 Brescia, ItalyNeurosurgeryUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyNeurosurgeryHumanitas, 20089 Milan, ItalyPathologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyBrainBehaviour and Mental Health, University of Manchester, M13 9PT Manchester, UK
| | - Sabrina Chiloiro
- EndocrinologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKPathologySTHF, N-3710 Skien, NorwayEndocrinologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyHaemato-OncologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKNeurosurgeryUniversità di Brescia, 25121 Brescia, ItalyNeurosurgeryUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyNeurosurgeryHumanitas, 20089 Milan, ItalyPathologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyBrainBehaviour and Mental Health, University of Manchester, M13 9PT Manchester, UK
| | - Serena Piacentini
- EndocrinologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKPathologySTHF, N-3710 Skien, NorwayEndocrinologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyHaemato-OncologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKNeurosurgeryUniversità di Brescia, 25121 Brescia, ItalyNeurosurgeryUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyNeurosurgeryHumanitas, 20089 Milan, ItalyPathologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyBrainBehaviour and Mental Health, University of Manchester, M13 9PT Manchester, UK
| | - Antonio Bianchi
- EndocrinologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKPathologySTHF, N-3710 Skien, NorwayEndocrinologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyHaemato-OncologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKNeurosurgeryUniversità di Brescia, 25121 Brescia, ItalyNeurosurgeryUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyNeurosurgeryHumanitas, 20089 Milan, ItalyPathologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyBrainBehaviour and Mental Health, University of Manchester, M13 9PT Manchester, UK
| | - Antonella Giampietro
- EndocrinologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKPathologySTHF, N-3710 Skien, NorwayEndocrinologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyHaemato-OncologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKNeurosurgeryUniversità di Brescia, 25121 Brescia, ItalyNeurosurgeryUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyNeurosurgeryHumanitas, 20089 Milan, ItalyPathologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyBrainBehaviour and Mental Health, University of Manchester, M13 9PT Manchester, UK
| | - Marilda Mormando
- EndocrinologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKPathologySTHF, N-3710 Skien, NorwayEndocrinologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyHaemato-OncologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKNeurosurgeryUniversità di Brescia, 25121 Brescia, ItalyNeurosurgeryUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyNeurosurgeryHumanitas, 20089 Milan, ItalyPathologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyBrainBehaviour and Mental Health, University of Manchester, M13 9PT Manchester, UK
| | - Andrew J Clear
- EndocrinologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKPathologySTHF, N-3710 Skien, NorwayEndocrinologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyHaemato-OncologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKNeurosurgeryUniversità di Brescia, 25121 Brescia, ItalyNeurosurgeryUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyNeurosurgeryHumanitas, 20089 Milan, ItalyPathologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyBrainBehaviour and Mental Health, University of Manchester, M13 9PT Manchester, UK
| | - Francesco Doglietto
- EndocrinologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKPathologySTHF, N-3710 Skien, NorwayEndocrinologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyHaemato-OncologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKNeurosurgeryUniversità di Brescia, 25121 Brescia, ItalyNeurosurgeryUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyNeurosurgeryHumanitas, 20089 Milan, ItalyPathologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyBrainBehaviour and Mental Health, University of Manchester, M13 9PT Manchester, UK
| | - Carmelo Anile
- EndocrinologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKPathologySTHF, N-3710 Skien, NorwayEndocrinologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyHaemato-OncologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKNeurosurgeryUniversità di Brescia, 25121 Brescia, ItalyNeurosurgeryUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyNeurosurgeryHumanitas, 20089 Milan, ItalyPathologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyBrainBehaviour and Mental Health, University of Manchester, M13 9PT Manchester, UK
| | - Giulio Maira
- EndocrinologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKPathologySTHF, N-3710 Skien, NorwayEndocrinologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyHaemato-OncologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKNeurosurgeryUniversità di Brescia, 25121 Brescia, ItalyNeurosurgeryUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyNeurosurgeryHumanitas, 20089 Milan, ItalyPathologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyBrainBehaviour and Mental Health, University of Manchester, M13 9PT Manchester, UK
| | - Libero Lauriola
- EndocrinologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKPathologySTHF, N-3710 Skien, NorwayEndocrinologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyHaemato-OncologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKNeurosurgeryUniversità di Brescia, 25121 Brescia, ItalyNeurosurgeryUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyNeurosurgeryHumanitas, 20089 Milan, ItalyPathologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyBrainBehaviour and Mental Health, University of Manchester, M13 9PT Manchester, UK
| | - Guido Rindi
- EndocrinologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKPathologySTHF, N-3710 Skien, NorwayEndocrinologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyHaemato-OncologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKNeurosurgeryUniversità di Brescia, 25121 Brescia, ItalyNeurosurgeryUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyNeurosurgeryHumanitas, 20089 Milan, ItalyPathologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyBrainBehaviour and Mental Health, University of Manchester, M13 9PT Manchester, UK
| | - Federico Roncaroli
- EndocrinologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKPathologySTHF, N-3710 Skien, NorwayEndocrinologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyHaemato-OncologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKNeurosurgeryUniversità di Brescia, 25121 Brescia, ItalyNeurosurgeryUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyNeurosurgeryHumanitas, 20089 Milan, ItalyPathologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyBrainBehaviour and Mental Health, University of Manchester, M13 9PT Manchester, UK
| | - Alfredo Pontecorvi
- EndocrinologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKPathologySTHF, N-3710 Skien, NorwayEndocrinologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyHaemato-OncologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKNeurosurgeryUniversità di Brescia, 25121 Brescia, ItalyNeurosurgeryUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyNeurosurgeryHumanitas, 20089 Milan, ItalyPathologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyBrainBehaviour and Mental Health, University of Manchester, M13 9PT Manchester, UK
| | - Márta Korbonits
- EndocrinologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKPathologySTHF, N-3710 Skien, NorwayEndocrinologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyHaemato-OncologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKNeurosurgeryUniversità di Brescia, 25121 Brescia, ItalyNeurosurgeryUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyNeurosurgeryHumanitas, 20089 Milan, ItalyPathologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyBrainBehaviour and Mental Health, University of Manchester, M13 9PT Manchester, UK
| | - Laura De Marinis
- EndocrinologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKPathologySTHF, N-3710 Skien, NorwayEndocrinologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyHaemato-OncologyBarts and The London School of Medicine, Queen Mary University of London, EC1M 6BQ London, UKNeurosurgeryUniversità di Brescia, 25121 Brescia, ItalyNeurosurgeryUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyNeurosurgeryHumanitas, 20089 Milan, ItalyPathologyUniversità Cattolica del Sacro Cuore, 00168 Rome, ItalyBrainBehaviour and Mental Health, University of Manchester, M13 9PT Manchester, UK
| |
Collapse
|
32
|
Abstract
Neuroendocrine tumors (NET) are characterized by a high over-expression of many different peptide hormone receptors. These receptors represent important molecular targets for imaging and therapy, using either radiolabeled or cold peptide analogs. The clinically best established example is somatostatin receptor targeting. A relatively new application is glucagon-like peptide 1 (GLP-1) receptor-targeted imaging of insulinomas, which is highly sensitive. A potential future candidate for peptide receptor targeting is the gastric inhibitory peptide (GIP) receptor. It was recently found to exhibit a very wide expression in NET and may be a particularly suitable target in somatostatin and GLP-1 receptor negative tumors. With increasing use of peptide receptor targeting, reliable morphologic in vitro tools to assess peptide receptors in tissues are mandatory, such as in vitro receptor autoradiography or thoroughly established immunohistochemical procedures.
Collapse
Affiliation(s)
- Meike Körner
- Pathologie Länggasse, CH-3001 Bern, Switzerland.
| |
Collapse
|
33
|
Analysis of somatostatin receptor 2A immunohistochemistry, RT-qPCR, and in vivo PET/CT data in patients with pancreatic neuroendocrine neoplasm. Pancreas 2015; 44:648-54. [PMID: 25872131 DOI: 10.1097/mpa.0000000000000316] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
OBJECTIVE Gallium 68 somatostatin receptor (SSTR) positron emission tomography/computed tomography (PET/CT) is one of the most sensitive imaging methods for pancreatic neuroendocrine tumors. The aim of the study was to correlate the receptor density generated from the static PET/CT (maximum standard uptake values [SUVmax], mean standard uptake values [SUVmean]) with subtype 2A SSTR (SSTR2A) immunohistochemistry and reverse transcriptase-quantitative polymerase chain reaction (RT-qPCR) gene-expression data. METHODS Thirty-nine tumor specimens (17 primary pancreatic tumors [PTs], 22 metastases [MTS]) of 19 patients with PET/CT scans preoperatively were evaluated. Subtype 2A SSTR expression was quantified immunohistochemically (immunoreactive score [IRS]) and on messenger RNA (mRNA) level by RT-qPCR. RESULTS The PT and MTS did not differ significantly in their SUVmax (P = 0.07) but displayed a dissimilarity with respect to their SSTR2A expression (mean [SD] IRS PT, 8.8 [3.6] vs mean [SD] IRS MTS, 5.1 [4.5]; P = 0.02).The SUVmean was highly significantly correlated to SSTR2A mRNA expression (C = 0.85, P < 0.001) and moderately to SSTR2A protein expression (C = 0.53, P = 0.05). Moreover, the SUVmax correlated moderately with SSTR2A protein expression (C = 0.44, P = 0.03) and mRNA expression (C = 0.64, P = 0.042). CONCLUSIONS The SUVmax and SUVmean are reliable ex vivo parameters for in vivo quantification of SSTR expression in pancreatic neuroendocrine tumors. Both immunohistochemistry and RT-qPCR are comparable methods for SSTR2A quantification. The PT and MTS differ significantly in their SSTR2A expression. This fact should be taken into account when treating patients with somatostatin analogs or peptide receptor radionuclide therapy.
Collapse
|
34
|
Specht E, Kaemmerer D, Sänger J, Wirtz RM, Schulz S, Lupp A. Comparison of immunoreactive score, HER2/neu score and H score for the immunohistochemical evaluation of somatostatin receptors in bronchopulmonary neuroendocrine neoplasms. Histopathology 2015; 67:368-77. [PMID: 25641082 DOI: 10.1111/his.12662] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 01/28/2015] [Indexed: 01/14/2023]
Abstract
AIMS Due to the growing number of somatostatin receptor (SSTR) targeting analogues and radiopeptides used for the diagnosis and therapy of neuroendocrine neoplasms (NEN), the assessment of SSTR subtype status has increasingly gained predictive value. In pathology, the SSTR protein levels are detected routinely by immunohistochemistry (IHC); however, a lack of a standardized evaluation system persists. Thus, in the present investigation, three well-established semi-quantitative scoring systems [immunoreactive score (IRS), human epidermal growth factor receptor 2 (HER2)/neu score, H score] used commonly for SSTR-IHC evaluation in NEN were compared. METHODS AND RESULTS A total of 240 formalin-fixed, paraffin-embedded tumour samples from 90 patients with bronchopulmonary NEN were examined by IHC and quantitative reverse transcription-polymerase chain reaction (qRT-PCR) for SSTR1, 2A, 3, 4 and 5 expression. Using both methods, SSTR1, 2A and 5 were the most frequently expressed subtypes. For all SSTR subtypes, all three scores correlated well with each other and with qRT-PCR data. However, the IRS was the most meaningful score with the best correlation to mRNA levels. CONCLUSIONS Because a unified IHC scoring system for SSTR analysis is needed urgently to optimize the theranostics of NEN, among the scores tested, the IRS seems to be the most suitable according to our results. It provides sufficient accuracy combined with high practicability.
Collapse
Affiliation(s)
- Elisa Specht
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - Daniel Kaemmerer
- Department of General and Visceral Surgery, Zentralklinik Bad Berka, Bad Berka, Germany
| | - Jörg Sänger
- Laboratory of Pathology and Cytology, Bad Berka, Germany
| | | | - Stefan Schulz
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - Amelie Lupp
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| |
Collapse
|
35
|
Kaemmerer D, Specht E, Sänger J, Wirtz RM, Sayeg M, Schulz S, Lupp A. Somatostatin receptors in bronchopulmonary neuroendocrine neoplasms: new diagnostic, prognostic, and therapeutic markers. J Clin Endocrinol Metab 2015; 100:831-40. [PMID: 25494861 DOI: 10.1210/jc.2014-2699] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
CONTEXT AND OBJECTIVES Gastroenteropancreatic neuroendocrine neoplasms are known for their overexpression of somatostatin receptors (SSTRs), which provide the molecular basis for diagnostic and therapeutic interventions. In contrast, few data on the SSTR expression profile exist for bronchopulmonary neuroendocrine neoplasms (BP-NEN). DESIGN AND SETTINGS A total of 240 formalin-fixed, paraffin-embedded specimens from 26 typical carcinoid (TC), 30 atypical carcinoid (AC), and 34 small cell lung cancer (SCLC) patients were examined retrospectively by immunohistochemistry (IHC) using specific rabbit monoclonal antibodies and evaluated by the immunoreactive score. Adjacent slides from 20 samples of each tumor type were subjected to additional RT-quantitative PCR mRNA analysis. RESULTS With different expression patterns, SSTRs were present in most of the tumor sections, at both the protein and mRNA levels. The RT-quantitative PCR data correlated with the IHC scores. SSTR1 was detected in approximately 65% of the TC and AC, but hardly in the SCLC, whereas both SSTR2A and SSTR5 were present in approximately 45% of each entity. Furthermore, the SSTR1 expression level was positively correlated with patient survival. CONCLUSIONS Our results suggest that SSTRs can be used as novel diagnostic, prognostic, and therapeutic markers of BP-NEN. The differences in the SSTR expression profile between the three types of BP-NEN may help to set a diagnostic cutoff and predict patient prognosis. Similar to TC and AC, our results also revealed a previously unappreciated high level of SSTR2A expression in SCLC within a subgroup of patients. However, in most cases, pan-somatostatin analogs may represent an additional therapeutic option.
Collapse
Affiliation(s)
- Daniel Kaemmerer
- Department of General and Visceral Surgery (D.K.), Zentralklinik Bad Berka, 99437 Bad Berka, Germany; Institute of Pharmacology and Toxicology (E.S., S.S., A.L.), Jena University Hospital, 07747 Jena, Germany; Laboratory of Pathology and Cytology (J.S.), 99437 Bad Berka, Germany; STRATIFYER Molecular Pathology GmbH (R.M.W.), 50935 Cologne, Germany; and Department of Internal Medicine, Gastroenterology, and Endocrinology (M.S.), Zentralklinik Bad Berka, 99437 Bad Berka, Germany
| | | | | | | | | | | | | |
Collapse
|
36
|
Schulz S, Mann A, Novakhov B, Piggins HD, Lupp A. VPAC2 receptor expression in human normal and neoplastic tissues: evaluation of the novel MAB SP235. Endocr Connect 2015; 4:18-26. [PMID: 25504760 PMCID: PMC4285768 DOI: 10.1530/ec-14-0051] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The vasoactive intestinal peptide receptor 2 (VPAC2) is widely distributed throughout the body and is also overexpressed in a variety of human neoplastic tissues. However, little is known about its precise tissue distribution, regulation and function, which is in part be due to the lack of specific monoclonal anti-VPAC2 antibodies. In this study, we extensively characterised the novel rabbit monoclonal anti-VPAC2 antibody (clone SP235) using transfected cells and mouse, rat and human tissues. SP235 was then subjected to a comparative immunohistochemical study on a series of 167 histological specimens from formalin-fixed, paraffin-embedded human tumours and adjacent normal tissues. SP235 detected a broad band migrating at a molecular weight of 50-70 kDa in western blotting analyses of various mouse tissues as well as VPAC2- but not VPAC1-transfected human embryonic kidney 293 cells. SP235 yielded an efficient immunostaining of distinct cell populations in human tissue samples with a predominance of plasma membrane staining, which was completely abolished by preadsorption with its immunising peptide. SP235 immunohistochemistry detected VPAC2 receptors in lymphocytes present in spleen, tonsils, lymph nodes and Peyer's patches, chief cells of gastric mucosa, exocrine and endocrine pancreas, kidney tubules and blood vessels. In addition, VPAC2 was observed in thyroid, gastric and lung carcinomas, pancreatic adenocarcinomas, sarcomas and neuroendocrine tumours. SP235 may prove of great value in the identification of VPAC2 receptors during routine histopathological examination. VPAC2 visualisation with this simple and rapid immunohistochemical method will facilitate identification of candidate tumours for vasoactive intestinal peptide (VIP)-based diagnostics or therapeutic interventions.
Collapse
Affiliation(s)
- Stefan Schulz
- Institute of Pharmacology and ToxicologyJena University Hospital, Friedrich Schiller University Jena, Drackendorfer Straße 1, D-07747 Jena, GermanyFaculty of Life SciencesUniversity of Manchester, Manchester M13 9PT, UK
| | - Anika Mann
- Institute of Pharmacology and ToxicologyJena University Hospital, Friedrich Schiller University Jena, Drackendorfer Straße 1, D-07747 Jena, GermanyFaculty of Life SciencesUniversity of Manchester, Manchester M13 9PT, UK
| | - Benjamin Novakhov
- Institute of Pharmacology and ToxicologyJena University Hospital, Friedrich Schiller University Jena, Drackendorfer Straße 1, D-07747 Jena, GermanyFaculty of Life SciencesUniversity of Manchester, Manchester M13 9PT, UK
| | - Hugh D Piggins
- Institute of Pharmacology and ToxicologyJena University Hospital, Friedrich Schiller University Jena, Drackendorfer Straße 1, D-07747 Jena, GermanyFaculty of Life SciencesUniversity of Manchester, Manchester M13 9PT, UK
| | - Amelie Lupp
- Institute of Pharmacology and ToxicologyJena University Hospital, Friedrich Schiller University Jena, Drackendorfer Straße 1, D-07747 Jena, GermanyFaculty of Life SciencesUniversity of Manchester, Manchester M13 9PT, UK
| |
Collapse
|
37
|
Gardair C, Samimi M, Touzé A, Coursaget P, Lorette G, Caille A, Wierzbicka E, Croué A, Avenel-Audran M, Aubin F, Kerdraon R, Estève E, Beneton N, Guyétant S. Somatostatin Receptors 2A and 5 Are Expressed in Merkel Cell Carcinoma with No Association with Disease Severity. Neuroendocrinology 2015; 101:223-35. [PMID: 25765179 DOI: 10.1159/000381062] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 02/18/2015] [Indexed: 11/19/2022]
Abstract
BACKGROUND/AIMS Merkel cell carcinoma (MCC) is a rare high-grade neuroendocrine tumour of the skin. It has been speculated that MCCs express somatostatin receptors (SSTRs), but this has never been assessed in a large series of MCCs. The main aim of this study was to assess the expression of SSTR2A and SSTR5 in MCC tumours. The secondary aims were to assess whether expression of SSTR was associated with the Ki67 proliferative index, Merkel cell polyomavirus (MCPyV) status, clinical characteristics and outcome. METHODS Clinical data and tumours were collected from an ongoing cohort of French patients with MCC. Immunohistochemistry was performed with anti-SSTR2A and anti-SSTR5 monoclonal antibodies, and tumours were classified into 3 groups: 'no expression', 'low expression' and 'moderate expression' using an SSTR staining score. RESULTS SSTR expression was assessed for 105 MCC tissue samples from 98 patients, and clinical characteristics were available for 87 of them. SSTR expression was consistent between the primary skin tumour and the corresponding metastases for SSTR2A and SSTR5 in 3/7 and 6/7 cases, respectively. SSTR2A and SSTR5 were expressed in 58 cases (59.2%) and in 44 cases (44.9%), respectively. Overall, at least one SSTR was expressed in 75 tumours (76.5%). SSTR expression was not associated with clinical characteristics, Ki67 proliferative index, recurrence-free survival or MCC-specific survival. Expression of SSTR2A was associated with MCPyV status in MCC tumours but not SSTR5. CONCLUSION SSTRs were expressed in a high proportion of MCCs, although expression was heterogeneous between tumours and was not associated with disease severity.
Collapse
Affiliation(s)
- Charlotte Gardair
- Department of Pathology, University Hospital of Tours, Tours, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Schulz S, Lehmann A, Kliewer A, Nagel F. Fine-tuning somatostatin receptor signalling by agonist-selective phosphorylation and dephosphorylation: IUPHAR Review 5. Br J Pharmacol 2014; 171:1591-9. [PMID: 24328848 PMCID: PMC3966740 DOI: 10.1111/bph.12551] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Revised: 10/08/2013] [Accepted: 10/31/2013] [Indexed: 12/28/2022] Open
Abstract
The biological actions of somatostatin are mediated by a family of five GPCRs, named sst1 to sst5 . Somatostatin receptors exhibit equally high-binding affinities to their natural ligand somatostatin-14 and largely overlapping distributions. The overexpression of somatostatin receptors in human tumours is the molecular basis for diagnostic and therapeutic application of the stable somatostatin analogues octreotide, lanreotide and pasireotide. The efficiency of somatostatin receptor signalling is tightly regulated and ultimately limited by the coordinated phosphorylation and dephosphorylation of intracellular carboxyl-terminal serine and threonine residues. Here, we review and discuss recent progress in the generation and application of phosphosite-specific antibodies for human sst2 and sst5 receptors. These phosphosite-specific antibodies are unique tools to monitor the spatial and temporal dynamics of receptors phosphorylation and dephosphorylation. Using a combined approach of phosphosite-specific antibodies and siRNA knock-down screening, relevant kinases and phosphatases were identified. Emerging evidence suggests distinct mechanisms of agonist-selective fine-tuning for individual somatostatin receptors. The recently uncovered differences in phosphorylation and dephosphorylation of these receptors may hence be of physiological significance in mediating responses to acute, persistent or repeated stimuli in a variety of target tissues.
Collapse
Affiliation(s)
- Stefan Schulz
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-UniversityJena, Germany
| | - Andreas Lehmann
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-UniversityJena, Germany
| | - Andrea Kliewer
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-UniversityJena, Germany
| | - Falko Nagel
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-UniversityJena, Germany
| |
Collapse
|
39
|
Chalabi M, Duluc C, Caron P, Vezzosi D, Guillermet-Guibert J, Pyronnet S, Bousquet C. Somatostatin analogs: does pharmacology impact antitumor efficacy? Trends Endocrinol Metab 2014; 25:115-27. [PMID: 24405892 DOI: 10.1016/j.tem.2013.11.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Revised: 11/05/2013] [Accepted: 11/15/2013] [Indexed: 01/17/2023]
Abstract
Somatostatin is an endogenous inhibitor of secretion and cell proliferation. These features render somatostatin a logical candidate for the management of neuroendocrine tumors that express somatostatin receptors. Synthetic somatostatin analogs (SSAs) have longer half-lives than somatostatin, but have similar activities, and are used for the treatment of these types of disorders. Interest has focused on novel multireceptor analogs with broader affinity to several of the five somatostatin receptors, thereby presenting putatively higher antitumor activities. Recent evidence indicates that SSAs cannot be considered mimics of native somatostatin in regulating signaling pathways downstream of receptors. Here we review this knowledge, discuss the concept of biased agonism, and highlight what considerations need to be taken into account for the optimal clinical use of SSAs.
Collapse
Affiliation(s)
- Mounira Chalabi
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unité Mixte de Recherche (UMR) 1037, Centre de Recherche en Cancérologie de Toulouse (CRCT), Equipe labellisée Ligue Contre le Cancer and Laboratoire d'Excellence Toulouse Cancer (TOUCAN), 31432 Toulouse, France; Université Toulouse III Paul Sabatier, 31062 Toulouse, France
| | - Camille Duluc
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unité Mixte de Recherche (UMR) 1037, Centre de Recherche en Cancérologie de Toulouse (CRCT), Equipe labellisée Ligue Contre le Cancer and Laboratoire d'Excellence Toulouse Cancer (TOUCAN), 31432 Toulouse, France; Université Toulouse III Paul Sabatier, 31062 Toulouse, France
| | - Philippe Caron
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unité Mixte de Recherche (UMR) 1037, Centre de Recherche en Cancérologie de Toulouse (CRCT), Equipe labellisée Ligue Contre le Cancer and Laboratoire d'Excellence Toulouse Cancer (TOUCAN), 31432 Toulouse, France; Université Toulouse III Paul Sabatier, 31062 Toulouse, France; Service d'Endocrinologie et Maladies Métaboliques, Pôle Cardio-Vasculaire et Métabolique, Centre Hospitalier Universitaire (CHU) Larrey, 31059 Toulouse, France
| | - Delphine Vezzosi
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unité Mixte de Recherche (UMR) 1037, Centre de Recherche en Cancérologie de Toulouse (CRCT), Equipe labellisée Ligue Contre le Cancer and Laboratoire d'Excellence Toulouse Cancer (TOUCAN), 31432 Toulouse, France; Université Toulouse III Paul Sabatier, 31062 Toulouse, France; Service d'Endocrinologie et Maladies Métaboliques, Pôle Cardio-Vasculaire et Métabolique, Centre Hospitalier Universitaire (CHU) Larrey, 31059 Toulouse, France
| | - Julie Guillermet-Guibert
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unité Mixte de Recherche (UMR) 1037, Centre de Recherche en Cancérologie de Toulouse (CRCT), Equipe labellisée Ligue Contre le Cancer and Laboratoire d'Excellence Toulouse Cancer (TOUCAN), 31432 Toulouse, France; Université Toulouse III Paul Sabatier, 31062 Toulouse, France
| | - Stéphane Pyronnet
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unité Mixte de Recherche (UMR) 1037, Centre de Recherche en Cancérologie de Toulouse (CRCT), Equipe labellisée Ligue Contre le Cancer and Laboratoire d'Excellence Toulouse Cancer (TOUCAN), 31432 Toulouse, France; Université Toulouse III Paul Sabatier, 31062 Toulouse, France
| | - Corinne Bousquet
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unité Mixte de Recherche (UMR) 1037, Centre de Recherche en Cancérologie de Toulouse (CRCT), Equipe labellisée Ligue Contre le Cancer and Laboratoire d'Excellence Toulouse Cancer (TOUCAN), 31432 Toulouse, France; Université Toulouse III Paul Sabatier, 31062 Toulouse, France.
| |
Collapse
|
40
|
Abstract
This paper summarizes the current understanding of the biology of somatostatin receptor (sst), role of immunotherapy in neuroendocrine tumor (NET), new agents for PPRT, and methods to assess response and clinical benefit in NET. One of the most interesting aspects of sst biology is the recent discovery of truncated variants of the sst5 receptor subtype with unique tissue distribution and response to somatostatin (SST). These truncated receptors are associated with bad patient prognosis, decreased response to SST analogs, and may be new targets for diagnoses and treatment. IFN remains a cost-effective agent, particularly in classic mid gut carcinoids, and there is interest to continue examining immunotherapy's in this disease. PRRT remains a key strategy for treatment and imaging. In addition to the classic agents, there are a series of new agents targeting other receptors such as the incretin receptors (GLP-1R; GIPR) and other G-protein coupled receptors with great potential. With regards to therapy monitoring, the most commonly used criteria are Response Criteria Evaluation in Solid Tumors (RECIST). However, for different reasons, these criteria are not very useful in NET. Incorporation of other criteria such as Choi as well as functional imaging assessment with PET would be of great interest in this area.
Collapse
|
41
|
Chinezu L, Vasiljevic A, Jouanneau E, François P, Borda A, Trouillas J, Raverot G. Expression of somatostatin receptors, SSTR2A and SSTR5, in 108 endocrine pituitary tumors using immunohistochemical detection with new specific monoclonal antibodies. Hum Pathol 2014; 45:71-7. [DOI: 10.1016/j.humpath.2013.08.007] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2013] [Revised: 08/02/2013] [Accepted: 08/02/2013] [Indexed: 11/28/2022]
|
42
|
Abstract
The peptide hormone somatostatin (SST) is produced in the brain, the gut, and in δ-cells in pancreatic islets of Langerhans. SST secretion from δ-cells is stimulated by glucose, amino acids, and glucagon-like peptide-1. Exogenous SST strongly inhibits the secretion of the blood glucose-regulating hormones insulin and glucagon from pancreatic β-cells and α-cells, respectively. Endogenous SST secreted from δ-cells is a paracrine regulator of insulin and glucagon secretion, although the exact physiological significance of this regulation is unclear. Secreted SST binds to specific receptors (SSTRs), which are coupled to Gi/o proteins. In both β- and α-cells, activation of SSTRs suppresses hormone secretion by reducing cAMP levels, inhibiting electrical activity, decreasing Ca²⁺ influx through voltage-gated Ca²⁺ channels and directly reducing exocytosis in a Ca²⁺ and cAMP-independent manner. In rodents, β-cells express predominantly SSTR5, whereas α-cells express SSTR2. In human islets, SSTR2 is the dominant receptor in both β- and α-cells, but other isoforms also contribute to the SST effects. Evidence from rodent models suggests that SST secretion from δ-cells is dysregulated in diabetes mellitus, which may contribute to the metabolic disturbances in this disease. SST analogues are currently used for the treatment of hyperinsulinism and other endocrine disorders, including acromegaly and Cushing's syndrome.
Collapse
Affiliation(s)
- Matthias Braun
- Alberta Diabetes Institute, Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
43
|
Peineau S, Guimiot F, Csaba Z, Jacquier S, Fafouri A, Schwendimann L, de Roux N, Schulz S, Gressens P, Auvin S, Dournaud P. Somatostatin receptors type 2 and 5 expression and localization during human pituitary development. Endocrinology 2014; 155:33-9. [PMID: 24189142 DOI: 10.1210/en.2013-1683] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Somatostatin (SRIF), by acting mainly through sst2 and sst5 receptors, is a potent inhibitor of hormonal secretion by the human anterior pituitary gland. However, the pattern of protein expression of these SRIF receptors remains unknown during pituitary development. To get further insights into the physiological role of SRIF receptors in human development and pituitary function, the present study examined the developmental expression of the sst2 and sst5 receptors in the individual cell types of the anterior human pituitary. Thirteen fetal human pituitaries were investigated between 13 to 38 weeks of gestation (WG) by double-labeling immunofluorescence with antibodies raised against sst2 or sst5 receptors and GH, LH, FSH, TSH, or pro-opiomelanocortin proteins. SRIF immunoreactivity in the hypothalamus and median eminence was investigated at the same developmental ages. Immunoreactivity for the sst2 receptor was evident as early as 13 to 15 WG and onward mainly in TSH-, LH-, and FSH-expressing cells, whereas sst5 immunoreactivity was apparent at the late development stages (35-38 WG). GH-expressing cells mainly expressed sst5 immunoreactivity. SRIF-positive fibers and cells were detected as soon as 13 to 16 WG in the hypothalamus and median eminence and their densities increased with gestational age. The early appearance of hypothalamic SRIF cells and fibers suggests a physiological link between SRIF and its receptors during pituitary development. Whereas sst2 receptors might play a primary role in the differentiation and regulation of TSH, LH, and FSH cells, sst5 receptors appear to be mainly involved in GH regulation from birth onward.
Collapse
Affiliation(s)
- Stéphane Peineau
- Medical Research Council Centre for Synaptic Plasticity (S.P.), University of Bristol, School of Physiology and Pharmacology, Bristol BS8 1TD, United Kingdom; Inserm (S.P., F.G., Z.C., S.J., A.F., L.S., N.d.R., P.G., S.A., P.D.), U676, 75019 Paris, France; University Paris Diderot (S.P., F.G., Z.C., S.J., A.F., L.S., N.d.R., P.G., S.A., P.D.), Sorbonne Paris Cité, UMR676, 75019 Paris, France; and Institute of Pharmacology and Toxicology (S.S.), Jena University Hospital, Friedrich Schiller University Jena, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Gastrointestinal Hormones and Their Targets. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 817:157-75. [DOI: 10.1007/978-1-4939-0897-4_7] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
45
|
Lambertini C, Barzaghi-Rinaudo P, D'Amato L, Schulz S, Nuciforo P, Schmid HA. Evaluation of somatostatin receptor subtype expression in human neuroendocrine tumors using two sets of new monoclonal antibodies. ACTA ACUST UNITED AC 2013; 187:35-41. [PMID: 24188818 DOI: 10.1016/j.regpep.2013.10.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Revised: 10/29/2013] [Accepted: 10/29/2013] [Indexed: 11/28/2022]
Abstract
INTRODUCTION The expression and reliable detection of somatostatin receptor subtypes (SSTR1-5) is a prerequisite for the successful use of somatostatin analogs in neuroendocrine tumors (NETs). Two sets of monoclonal antibodies (mAbs) against human SSTR1, 2A, 3 and 5 have recently been developed by two independent laboratories using rabbit and mouse hybridomas. Our aim was to evaluate the usefulness of both sets of mAbs for detection of SSTRs in NET samples as they are routinely collected in clinical practice. METHODS Mouse and rabbit mAbs were characterized in SSTR1, 2A, 3 and 5-transfected HEK293 cells and human archival samples of pancreatic tissue and NET. Comparative analysis of mAbs was also conducted by immunostaining of a tissue microarray composed of 75 cores of NET. RESULTS Immunohistochemical analysis of HEK293 cells showed that both rabbit and mouse mAbs specifically detect their cognate receptor subtype, with mild cytoplasmic cross-reactivity observed for rabbit mAbs. Both sets of mAbs labeled normal pancreatic islets and showed similar patterns of immunoreactivity in NET controls. Direct comparison of mAb sets using a NET tissue microarray revealed strong correlation between rabbit and mouse mAbs against SSTR1 and 5, and moderate correlation for SSTR3. The rabbit mAb against SSTR2A showed higher affinity for its cognate receptor than the corresponding mouse mAb, resulting in a more reliable detection of this SSTR. CONCLUSIONS mAbs from both sets are reliable tools for the detection of SSTR1, 3 and 5, whereas the rabbit mAb against SSTR2A is recommended for use in routine clinical testing due to its superior binding affinity.
Collapse
|
46
|
Casar-Borota O, Heck A, Schulz S, Nesland JM, Ramm-Pettersen J, Lekva T, Alafuzoff I, Bollerslev J. Expression of SSTR2a, but not of SSTRs 1, 3, or 5 in somatotroph adenomas assessed by monoclonal antibodies was reduced by octreotide and correlated with the acute and long-term effects of octreotide. J Clin Endocrinol Metab 2013; 98:E1730-9. [PMID: 24092823 DOI: 10.1210/jc.2013-2145] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
CONTEXT Reduced expression of somatostatin receptors (SSTRs) in somatotroph adenomas and their potential down-regulation after medical treatment may explain the unsatisfactory response to octreotide in particular acromegalic patients. The expression of SSTRs other than SSTR2a has not been studied in large, unselected cohorts using novel rabbit monoclonal antibodies. OBJECTIVE We aimed to determine the expression of SSTRs 1, 2a, 3, and 5 in somatotroph adenomas, to correlate expression with clinical characteristics and the response to octreotide, and to ascertain whether preoperative octreotide treatment affected SSTR expression. DESIGN, SETTING, PATIENTS The study included 78 adenomas from patients operated on consecutively during 2000 to 2010. After exclusion of 13 patients, immunohistochemical analysis with rabbit monoclonal antibodies against SSTRs 1, 2a, 3, and 5 (clones UMB-7, -1, -5, and -4) was performed on 65 adenomas. INTERVENTION Twenty-eight patients received preoperative octreotide, and 37 patients were operated on without pretreatment. Twenty-six patients were randomized to direct surgery (n = 13) or to octreotide pretreatment (n = 13). MAIN OUTCOME MEASURE SSTR expression was evaluated using a 12-grade scoring system. The responses to the octreotide test dose (GH reduction) and to 6 months of octreotide (IGF-I reduction) were measured. RESULTS The majority of adenomas showed membranous expression of SSTRs 2a and 5. SSTR2a expression was reduced in the pretreated group and correlated with the acute octreotide test results and the effect of octreotide treatment. In a linear regression model with SSTR2a expression as the determinant, the correlation with the acute test response improved after adjustment for medical pretreatment. CONCLUSION Rabbit monoclonal antibodies are reliable markers of SSTRs in somatotroph adenomas. SSTR2a expression correlated with the response to octreotide and was reduced after octreotide treatment, indicating the need for adjustment when SSTR2a expression is correlated with baseline characteristics. Evaluation of SSTR subtypes may be an important aspect of improving the medical treatment for acromegaly.
Collapse
Affiliation(s)
- Olivera Casar-Borota
- Section of Specialized Endocrinology, Oslo University Hospital, Rikshospitalet, PO Box 4950 Nydalen, 0424 Oslo, Norway.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
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
|
48
|
Lupp A, Nagel F, Schulz S. Reevaluation of sst₁ somatostatin receptor expression in human normal and neoplastic tissues using the novel rabbit monoclonal antibody UMB-7. ACTA ACUST UNITED AC 2013; 183:1-6. [PMID: 23466804 DOI: 10.1016/j.regpep.2013.02.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Revised: 12/20/2012] [Accepted: 02/13/2013] [Indexed: 11/17/2022]
Abstract
BACKGROUND The somatostatin receptor 1 (sst1) is widely distributed throughout the body and is also present in neoplastic tissues. However, little is known about its precise tissue distribution, regulation and function, which may in part be due to the lack of specific monoclonal anti-sst1 antibodies. METHODS We have characterized the novel rabbit monoclonal anti-human sst1 antibody UMB-7 using sst1-expressing cells and human pituitary samples. The antibody was then used for immunohistochemical staining of a large panel of formalin-fixed, paraffin-embedded human tissues. RESULTS Western blot analyses of BON-1 cells and human pituitary revealed a broad band migrating at a molecular weight of 45,000-60,000. After enzymatic deglycosylation the size of this band decreased to a molecular weight of 45,000. UMB-7 yielded an efficient immunostaining of distinct cell populations in the human tissue samples with a predominance of plasma membrane staining, which was completely abolished by preadsorption of UMB-7 with its immunizing peptide. The sst1 receptor was detected in anterior pituitary, pancreatic islets, distal tubules, enteric ganglion cells and nerve fibers, chief cells of the gastric mucosa, macrophages and mast cells. In addition, sst1 was observed in pituitary adenomas, gastrointestinal neuroendocrine tumors and pheochromocytoma as well as in pancreatic adenocarcinomas, gastric carcinomas, urinary bladder carcinomas and sarcomas. CONCLUSIONS UMB-7 may prove of great value in the identification of sst1-expressing tumors during routine histopathological examinations. This may open up new routes for diagnostic and therapeutic intervention.
Collapse
Affiliation(s)
- Amelie Lupp
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Germany
| | - Falko Nagel
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Germany
| | - Stefan Schulz
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Germany.
| |
Collapse
|
49
|
Petrich A, Mann A, Kliewer A, Nagel F, Strigli A, Märtens JC, Pöll F, Schulz S. Phosphorylation of threonine 333 regulates trafficking of the human sst5 somatostatin receptor. Mol Endocrinol 2013; 27:671-82. [PMID: 23418396 DOI: 10.1210/me.2012-1329] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The frequent overexpression of the somatostatin receptors sst2 and sst5 in neuroendocrine tumors provides the molecular basis for therapeutic application of novel multireceptor somatostatin analogs. Although the phosphorylation of the carboxyl-terminal region of the sst2 receptor has been studied in detail, little is known about the agonist-induced regulation of the human sst5 receptor. Here, we have generated phosphosite-specific antibodies for the carboxyl-terminal threonines 333 (T333) and 347 (T347), which enabled us to selectively detect either the T333-phosphorylated or the T347-phosphorylated form of sst5. We show that agonist-mediated phosphorylation occurs at T333, whereas T347 is constitutively phosphorylated in the absence of agonist. We further demonstrate that the multireceptor somatostatin analog pasireotide and the sst5-selective ligand L-817,818 but not octreotide or KE108 were able to promote a detectable T333 phosphorylation. Interestingly, BIM-23268 was the only sst5 agonist that was able to stimulate T333 phosphorylation to the same extent as natural somatostatin. Agonist-induced T333 phosphorylation was dose-dependent and selectively mediated by G protein-coupled receptor kinase 2. Similar to that observed for the sst2 receptor, phosphorylation of sst5 occurred within seconds. However, unlike that seen for the sst2 receptor, dephosphorylation and recycling of sst5 were rapidly completed within minutes. We also identify protein phosphatase 1γ as G protein-coupled receptor phosphatase for the sst5 receptor. Together, we provide direct evidence for agonist-selective phosphorylation of carboxyl-terminal T333. In addition, we identify G protein-coupled receptor kinase 2-mediated phosphorylation and protein phosphatase 1γ-mediated dephosphorylation of T333 as key regulators of rapid internalization and recycling of the human sst5 receptor.
Collapse
Affiliation(s)
- Aline Petrich
- Department of Pharmacology and Toxicology, Jena University Hospital–Friedrich Schiller University Jena, Drackendorfer Strasse 1, D-07749 Jena, Germany
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
The neuropeptide somatostatin (SRIF) is an important modulator of neurotransmission in the central nervous system and acts as a potent inhibitor of hormone and exocrine secretion. In addition, SRIF regulates cell proliferation in normal and tumorous tissues. The six somatostatin receptor subtypes (sst1, sst2A, sst2B, sst3, sst4, and sst5), which belong to the G protein-coupled receptor (GPCR) family, share a common molecular topology: a hydrophobic core of seven transmembrane-spanning α-helices, three intracellular loops, three extracellular loops, an amino-terminus outside the cell, and a carboxyl-terminus inside the cell. For most of the GPCRs, intracytosolic sequences, and more particularly the C-terminus, are believed to interact with proteins that are mandatory for either exporting neosynthesized receptor, anchoring receptor at the plasma membrane, internalization, recycling, or degradation after ligand binding. Accordingly, most of the SRIF receptors can traffic not only in vitro within different cell types but also in vivo. A picture of the pathways and proteins involved in these processes is beginning to emerge.
Collapse
Affiliation(s)
- Zsolt Csaba
- INSERM, Unité Mixte de Recherche U676, Paris, France
| | | | | |
Collapse
|