1
|
Wahba A, Tan Z, Dillon JS. Management of functional neuroendocrine tumors. Curr Probl Cancer 2024; 52:101130. [PMID: 39213785 DOI: 10.1016/j.currproblcancer.2024.101130] [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: 02/02/2024] [Accepted: 05/22/2024] [Indexed: 09/04/2024]
Abstract
Functional neuroendocrine neoplasms (NENs) are those associated with specific symptoms related to the hormonal secretion of the NENs. Although less than 25 % of NENs are functional at diagnosis,1 the associated syndromes significantly increase the patient burden of disease. Management of hormonal NEN symptoms may involve tumor resection or other reduction strategies (e.g., chemotherapy, embolotherapy, etc), but also specific therapies directed at decreasing hormonal synthesis, secretion, or end-organ effects. In this review, we focus on specific symptomatic management of many of the NEN syndromes, which may be pursued in addition to management primarily directed at tumor bulk and growth. A continued focus on symptom management related to the hormonal secretions of NENs, in the context of other efforts to reduce tumor bulk and growth, could significantly improve patient wellbeing.
Collapse
Affiliation(s)
- Amr Wahba
- ENETS Center of Excellence, Division of Endocrinology and Metabolism, University of Iowa, 200 Hawkins Drive, Room E400 GH, Iowa City, Iowa, 52242, USA
| | - Zi Tan
- ENETS Center of Excellence, Division of Endocrinology and Metabolism, University of Iowa, 200 Hawkins Drive, Room E400 GH, Iowa City, Iowa, 52242, USA
| | - Joseph S Dillon
- ENETS Center of Excellence, Division of Endocrinology and Metabolism, University of Iowa, 200 Hawkins Drive, Room E400 GH, Iowa City, Iowa, 52242, USA.
| |
Collapse
|
2
|
Yeung HY, Ramiro IBL, Andersen DB, Koch TL, Hamilton A, Bjørn-Yoshimoto WE, Espino S, Vakhrushev SY, Pedersen KB, de Haan N, Hipgrave Ederveen AL, Olivera BM, Knudsen JG, Bräuner-Osborne H, Schjoldager KT, Holst JJ, Safavi-Hemami H. Fish-hunting cone snail disrupts prey's glucose homeostasis with weaponized mimetics of somatostatin and insulin. Nat Commun 2024; 15:6408. [PMID: 39164229 PMCID: PMC11336141 DOI: 10.1038/s41467-024-50470-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 07/04/2024] [Indexed: 08/22/2024] Open
Abstract
Venomous animals have evolved diverse molecular mechanisms to incapacitate prey and defend against predators. Most venom components disrupt nervous, locomotor, and cardiovascular systems or cause tissue damage. The discovery that certain fish-hunting cone snails use weaponized insulins to induce hypoglycemic shock in prey highlights a unique example of toxins targeting glucose homeostasis. Here, we show that, in addition to insulins, the deadly fish hunter, Conus geographus, uses a selective somatostatin receptor 2 (SSTR2) agonist that blocks the release of the insulin-counteracting hormone glucagon, thereby exacerbating insulin-induced hypoglycemia in prey. The native toxin, Consomatin nG1, exists in several proteoforms with a minimized vertebrate somatostatin-like core motif connected to a heavily glycosylated N-terminal region. We demonstrate that the toxin's N-terminal tail closely mimics a glycosylated somatostatin from fish pancreas and is crucial for activating the fish SSTR2. Collectively, these findings provide a stunning example of chemical mimicry, highlight the combinatorial nature of venom components, and establish glucose homeostasis as an effective target for prey capture.
Collapse
Affiliation(s)
- Ho Yan Yeung
- Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200, Copenhagen N, Denmark
- Department of Biochemistry, University of Utah, 15 N Medical Drive, Salt Lake City, UT, 84112, USA
| | - Iris Bea L Ramiro
- Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200, Copenhagen N, Denmark
| | - Daniel B Andersen
- Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200, Copenhagen N, Denmark
- Novo Nordisk Foundation Centre for Basic Metabolic Research, Blegdamsvej 3, DK-2200, Copenhagen N, Denmark
| | - Thomas Lund Koch
- Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200, Copenhagen N, Denmark
- Department of Biochemistry, University of Utah, 15 N Medical Drive, Salt Lake City, UT, 84112, USA
- School of Biological Sciences, University of Utah, 257 South 1400 East, Salt Lake City, UT, 84112, USA
| | - Alexander Hamilton
- Department of Biology, University of Copenhagen, Ole Maaløes Vej 5, DK-2200, Copenhagen N, Denmark
- Department of Clinical Sciences in Malmö, Islet Cell Exocytosis, Lund University, Malmö, Sweden
| | - Walden E Bjørn-Yoshimoto
- Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200, Copenhagen N, Denmark
| | - Samuel Espino
- School of Biological Sciences, University of Utah, 257 South 1400 East, Salt Lake City, UT, 84112, USA
| | - Sergey Y Vakhrushev
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, DK-2200, Copenhagen N, Denmark
| | - Kasper B Pedersen
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, DK-2200, Copenhagen N, Denmark
| | - Noortje de Haan
- Leiden University Medical Center, Center for Proteomics and Metabolomics, 2333, ZA, Leiden, The Netherlands
| | - Agnes L Hipgrave Ederveen
- Leiden University Medical Center, Center for Proteomics and Metabolomics, 2333, ZA, Leiden, The Netherlands
| | - Baldomero M Olivera
- School of Biological Sciences, University of Utah, 257 South 1400 East, Salt Lake City, UT, 84112, USA
| | - Jakob G Knudsen
- Department of Biology, University of Copenhagen, Ole Maaløes Vej 5, DK-2200, Copenhagen N, Denmark
| | - Hans Bräuner-Osborne
- Department of Drug Design and Pharmacology, University of Copenhagen, Jagtvej 160, DK-2100, Copenhagen, Denmark
| | - Katrine T Schjoldager
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, DK-2200, Copenhagen N, Denmark
| | - Jens Juul Holst
- Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200, Copenhagen N, Denmark
- Novo Nordisk Foundation Centre for Basic Metabolic Research, Blegdamsvej 3, DK-2200, Copenhagen N, Denmark
| | - Helena Safavi-Hemami
- Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200, Copenhagen N, Denmark.
- Department of Biochemistry, University of Utah, 15 N Medical Drive, Salt Lake City, UT, 84112, USA.
- School of Biological Sciences, University of Utah, 257 South 1400 East, Salt Lake City, UT, 84112, USA.
| |
Collapse
|
3
|
Chowdhury A, Tripathi NM, Jadav R, Gour V, Purohit P, Bandyopadhyay A. On-resin synthesis of Lanreotide epimers and studies of their structure-activity relationships. RSC Med Chem 2024; 15:2766-2772. [PMID: 39149098 PMCID: PMC11324058 DOI: 10.1039/d4md00338a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 06/27/2024] [Indexed: 08/17/2024] Open
Abstract
Peptide drugs often accompany epimeric impurities (isomers). Therefore, efficient chemical synthesis of epimers is critical to identify them correctly and investigate their biological activities. Here, we report the rapid synthesis and structure-activity relationship (SAR) studies of eight possible epimers of a somatostatin synthetic analog (SSA), lanreotide (LAN). SPPS and the subsequent on-resin rapid disulfide closure method offered >90% conversion yield for all epimers (P1-P8). Further, we developed an analytical method to separate these epimers, which enabled the profiling of five epimeric impurities in the API, purchased for Somatuline generic formulations. In SAR studies, most LAN epimers revealed compromised antiproliferative activity, while the P7 epimer retained antiproliferative activity similar to LAN API, as supported by in silico SAR studies in detail. Additionally, P7 showed serum stability nearly identical to LAN, suggesting that drug epimers could be a potential API. Current studies will further encourage the development of novel SSA scaffolds.
Collapse
Affiliation(s)
- Arnab Chowdhury
- Biomimetic Peptide Engineering Laboratory, Department of Chemistry, Indian Institute of Technology Ropar Rupnagar Punjab-140001 India
| | - Nitesh Mani Tripathi
- Biomimetic Peptide Engineering Laboratory, Department of Chemistry, Indian Institute of Technology Ropar Rupnagar Punjab-140001 India
| | - Rohit Jadav
- Kashiv BioSciences Pvt Ltd. Block-B, Sardar Patel Ring Rd, opp. Applewoods Township Ahmedabad Gujarat-382210 India
| | - Vinod Gour
- Biomimetic Peptide Engineering Laboratory, Department of Chemistry, Indian Institute of Technology Ropar Rupnagar Punjab-140001 India
| | - Parva Purohit
- Kashiv BioSciences Pvt Ltd. Block-B, Sardar Patel Ring Rd, opp. Applewoods Township Ahmedabad Gujarat-382210 India
| | - Anupam Bandyopadhyay
- Biomimetic Peptide Engineering Laboratory, Department of Chemistry, Indian Institute of Technology Ropar Rupnagar Punjab-140001 India
| |
Collapse
|
4
|
Immler M, Wolfram M, Oevermann A, Walter I, Wolfesberger B, Tichy A, Gradner G. Expression of somatostatin receptors in canine and feline meningioma. Vet Med Sci 2024; 10:e1537. [PMID: 39011594 PMCID: PMC11250153 DOI: 10.1002/vms3.1537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 06/11/2024] [Accepted: 06/14/2024] [Indexed: 07/17/2024] Open
Abstract
OBJECTIVES The standard treatment for canine and feline meningiomas includes radiotherapy, surgical excision or combined therapy. However, new therapeutic approaches are required due to the possible recurrence or progression of meningiomas despite initial therapy. Adjunctive therapy with synthetic long-acting somatostatin (SST) analogues has been described in humans with SST-expressing tumours. The expression of SST receptors (SSTRs) by feline meningiomas is currently unknown, and there are little data about canine meningiomas. We hypothesized that SSTR is expressed by canine and feline meningiomas (S1). METHODS Seven canines and 11 felines with histologically confirmed meningiomas underwent STTR screening. RNA expressions of SSTR1, SSTR2, SSTR3 and SSTR5 (canine) and SSTR1-SSTR 5 (feline) in fresh frozen and formalin-fixed and paraffin-embedded (FFPE) samples were investigated using real-time (RT)-qPCR. The expression of SSTR1 and SSTR2 in FFPE samples was evaluated using immunohistochemistry (IHC). The specificity of applied antibodies for canine and feline species was confirmed by western blotting. RESULTS In canine meningiomas (n = 7), RNA expression of SSTR1, SSTR2 and SSTR5 was detected in all samples; SSTR3 RNA expression was detected in only 33% of samples. In feline meningiomas (n = 12), RNA expression of SSTR1, SSTR4, SSTR5 and SSTR2 was detected in 91%, 46%, 46% and 36% of samples, respectively; SSTR3 was not expressed. Overall, the detection rate was lower in FFPE samples. IHC revealed the expression of SSTR1 and SSTR2 in all samples from both species. However, it is important to exercise caution when interpreting IHC results due to the presence of diffuse background staining. CONCLUSIONS SSTRs are widely expressed in canine and feline meningiomas, thereby encouraging further studies investigating SSTR expression to conduct trials about the effect of adjunctive therapy with long-acting SST-analogues.
Collapse
Affiliation(s)
- Martin Immler
- University of Veterinary Medicine Vienna (Vetmeduni), Veterinaerplatz 1ViennaAustria
| | - Michael Wolfram
- University of Veterinary Medicine Vienna (Vetmeduni), Veterinaerplatz 1ViennaAustria
| | - Anna Oevermann
- Division of Neurological SciencesVetsuisse Faculty, University of Bern, Bremgartenstrasse 109a, 3012BernSwitzerland
| | - Ingrid Walter
- University of Veterinary Medicine Vienna (Vetmeduni), Veterinaerplatz 1ViennaAustria
| | - Birgitt Wolfesberger
- University of Veterinary Medicine Vienna (Vetmeduni), Veterinaerplatz 1ViennaAustria
| | - Alexander Tichy
- University of Veterinary Medicine Vienna (Vetmeduni), Veterinaerplatz 1ViennaAustria
| | - Gabriele Gradner
- University of Veterinary Medicine Vienna (Vetmeduni), Veterinaerplatz 1ViennaAustria
| |
Collapse
|
5
|
Bjørn-Yoshimoto WE, Ramiro IBL, Koch TL, Engholm E, Yeung HY, Sørensen KK, Goddard CM, Jensen KL, Smith NA, Martin LF, Smith BJ, Madsen KL, Jensen KJ, Patwardhan A, Safavi-Hemami H. Venom-inspired somatostatin receptor 4 (SSTR4) agonists as new drug leads for peripheral pain conditions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.29.591104. [PMID: 38746149 PMCID: PMC11092515 DOI: 10.1101/2024.04.29.591104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Persistent pain affects one in five people worldwide, often with severely debilitating consequences. Current treatment options, which can be effective for mild or acute pain, are ill-suited for moderate-to-severe persistent pain, resulting in an urgent need for new therapeutics. In recent years, the somatostatin receptor 4 (SSTR 4 ), which is expressed in sensory neurons of the peripheral nervous system, has emerged as a promising target for pain relief. However, the presence of several closely related receptors with similar ligand-binding surfaces complicates the design of receptor-specific agonists. In this study, we report the discovery of a potent and selective SSTR 4 peptide, consomatin Fj1, derived from extensive venom gene datasets from marine cone snails. Consomatin Fj1 is a mimetic of the endogenous hormone somatostatin and contains a minimized binding motif that provides stability and drives peptide selectivity. Peripheral administration of synthetic consomatin Fj1 provided analgesia in mouse models of postoperative and neuropathic pain. Using structure-activity studies, we designed and functionally evaluated several Fj1 analogs, resulting in compounds with improved potency and selectivity. Our findings present a novel avenue for addressing persistent pain through the design of venom-inspired SSTR 4 -selective pain therapeutics. One Sentence Summary Venom peptides from predatory marine mollusks provide new leads for treating peripheral pain conditions through a non-opioid target.
Collapse
|
6
|
Pusceddu S, Corti F, Prinzi N, Nichetti F, Ljevar S, Busico A, Cascella T, Leporati R, Oldani S, Pircher CC, Coppa J, Resi V, Milione M, Maccauro M, Miceli R, Tamborini E, Perrone F, Spreafico C, Niger M, Morano F, Pietrantonio F, Seregni E, Mariani L, Mazzaferro V, Di Liberti G, Fucà G, de Braud F, Vernieri C. Safety and antitumor activity of metformin plus lanreotide in patients with advanced gastro-intestinal or lung neuroendocrine tumors: the phase Ib trial MetNET2. J Hematol Oncol 2023; 16:119. [PMID: 38098114 PMCID: PMC10722662 DOI: 10.1186/s13045-023-01510-9] [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: 09/03/2023] [Accepted: 11/08/2023] [Indexed: 12/17/2023] Open
Abstract
In retrospective studies, metformin use has been associated with better clinical outcomes in diabetic patients with advanced, well-differentiated neuroendocrine tumors (WDNETs). However, prospective evidence of metformin safety and activity is lacking. Here, we conducted the first-in-human phase Ib MetNET2 trial to investigate the safety and antitumor activity of metformin in combination with the somatostatin analog lanreotide autogel (ATG) in both diabetic and non-diabetic patients with advanced WDNETs of the gastrointestinal (GI) or thoracic tract. Enrolled patients received lanreotide ATG 120 mg plus oral metformin, up to a maximum dosage of 2550 mg/day. We enrolled 20 patients, of whom 18 (90%) and 2 (10%) had WDNETs of the GI and thoracic tract, respectively. Fourteen patients (70%) were non-diabetic. With a 5% incidence of SAEs, the study met its primary objective of demonstrating treatment safety. With a median follow-up of 39 months (95% CI 28-NE), median PFS was 24 months (95% CI 16-NE), with 12-month and 24-month PFS probability of 75% (95% CI 58-97) and 49% (95% CI 31-77), respectively. We found no statistically significant PFS differences between diabetic and non-diabetic patients. Among exploratory analyses, the presence of tumor genomic alterations in DNA damage pathways was associated with trend towards worse PFS, whereas a precocious reduction of HOMA-IR index and plasma cholesterol concentration showed a trend towards an association with better PFS. In conclusion, metformin plus lanreotide ATG is a safe and well tolerated combination treatment that is associated with promising antitumor activity in both non-diabetic and diabetic patients with WDNETs, and that warrants further investigation in larger clinical trials.
Collapse
Affiliation(s)
- Sara Pusceddu
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Via Venezian 1, 20133, Milan, Italy.
| | - Francesca Corti
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Via Venezian 1, 20133, Milan, Italy
- SC Medical Oncology, Fondazione IRCCS San Gerardo Dei Tintori, Monza, Italy
| | - Natalie Prinzi
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Via Venezian 1, 20133, Milan, Italy
| | - Federico Nichetti
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Via Venezian 1, 20133, Milan, Italy
| | - Silva Ljevar
- Clinical Epidemiology and Trial Organization, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale Tumori Di Milano, Milan, Italy
| | - Adele Busico
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Milan, Italy
| | - Tommaso Cascella
- Department of Radiology Foundation IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Milan, Italy
| | - Rita Leporati
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Via Venezian 1, 20133, Milan, Italy
| | - Simone Oldani
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Via Venezian 1, 20133, Milan, Italy
| | - Chiara Carlotta Pircher
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Via Venezian 1, 20133, Milan, Italy
| | - Jorgelina Coppa
- Hepato-Biliary-Pancreatic and Upper G.I. Surgery, Liver Transplantation and Hepato-Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Milan, Italy
| | - Veronica Resi
- Endocrinology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Massimo Milione
- Department of the Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Milan, Italy
| | - Marco Maccauro
- Departement of Nuclear Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Milan, Italy
| | - Rosalba Miceli
- Clinical Epidemiology and Trial Organization, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale Tumori Di Milano, Milan, Italy
| | - Elena Tamborini
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Milan, Italy
| | - Federica Perrone
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Milan, Italy
| | - Carlo Spreafico
- Department of Radiology Foundation IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Milan, Italy
| | - Monica Niger
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Via Venezian 1, 20133, Milan, Italy
| | - Federica Morano
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Via Venezian 1, 20133, Milan, Italy
| | - Filippo Pietrantonio
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Via Venezian 1, 20133, Milan, Italy
| | - Ettore Seregni
- Departement of Nuclear Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Milan, Italy
| | - Luigi Mariani
- Clinical Epidemiology and Trial Organization, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale Tumori Di Milano, Milan, Italy
| | - Vincenzo Mazzaferro
- Hepato-Biliary-Pancreatic and Upper G.I. Surgery, Liver Transplantation and Hepato-Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Milan, Italy
- Department of Oncology and Hemato-Oncology, Università deli Studi di Milano, Milan, Italy
| | - Giorgia Di Liberti
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Via Venezian 1, 20133, Milan, Italy
| | - Giovanni Fucà
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Via Venezian 1, 20133, Milan, Italy
| | - Filippo de Braud
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Via Venezian 1, 20133, Milan, Italy
- Department of Oncology and Hemato-Oncology, Università deli Studi di Milano, Milan, Italy
| | - Claudio Vernieri
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Via Venezian 1, 20133, Milan, Italy.
- IFOM ETS - The AIRC Institute of Molecular Oncology, Via Ademello 16, 20139, Milan, Italy.
| |
Collapse
|
7
|
Vinzant NJ, Laporta ML, Sprung J, Atwell TD, Reisenauer CJ, Welch TL, Schulte PJ, Weingarten TN. Hemodynamic course during ablation and selective hepatic artery embolization for metastatic liver carcinoid: a retrospective observational study. BRAZILIAN JOURNAL OF ANESTHESIOLOGY (ELSEVIER) 2023; 73:603-610. [PMID: 33895218 PMCID: PMC10533977 DOI: 10.1016/j.bjane.2021.03.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 03/15/2021] [Accepted: 03/19/2021] [Indexed: 11/27/2022]
Abstract
BACKGROUND Manipulation of carcinoid tumors during ablation or selective hepatic artery embolization (transarterial embolization, TAE) can release vasoactive mediators inducing hemodynamic instability. The main aim of our study was to review hemodynamics and complications related to minimally invasive treatments of liver carcinoids with TAE or ablation. METHODS Electronic medical records of all patients with metastatic liver carcinoid undergoing ablation or TAE from 2003 to 2019 were abstracted. Noted were severe hypotension (mean arterial pressure [MAP] ..± 55.ßmmHg), severe hypertension (systolic blood pressure ... 180.ßmmHg), and perioperative complications. Associations of procedure type and pre-procedure octreotide use with intraprocedural hemodynamics were assessed using linear regression. A robust covariance approach using generalized estimating equation method was used to account for multiple observations. RESULTS A total of 161 patients underwent 98 ablations and 207 TAEs. Severe hypertension was observed in 24 (24.5%) vs. 15 (7.3%), severe hypotension in 56 (57.1%) vs. 6 (2.9%), and cutaneous flushing observed in 2 (2.0%) vs. 48 (23.2%) ablations and TAEs, respectively. After adjusting for preprocedural MAP, ablation was associated with lower intraprocedural MAP compared to TAE (estimate -27.ßmmHg, 95%CI -30 to -24.ßmmHg, p.ß<.ß0.001). Intraprocedural declines in MAP were not affected by preprocedural use of octreotide (p.ß=.ß0.7 for TAE and p.ß=.ß0.4 for ablation). CONCLUSIONS Ablation of liver carcinoids was associated with substantial hemodynamic instability, especially hypotension. In contrast, a higher number of TAE patients had cutaneous flushing. Preprocedural use of octreotide was not associated with attenuation of intraprocedural hypotension.
Collapse
Affiliation(s)
- Nathan J Vinzant
- Mayo Clinic College of Medicine and Science, Departments of Anesthesiology and Perioperative Medicine, Rochester, United States
| | - Mariana L Laporta
- Mayo Clinic College of Medicine and Science, Departments of Anesthesiology and Perioperative Medicine, Rochester, United States.
| | - Juraj Sprung
- Mayo Clinic College of Medicine and Science, Departments of Anesthesiology and Perioperative Medicine, Rochester, United States
| | - Thomas D Atwell
- Mayo Clinic College of Medicine and Science, Department of Radiology, Rochester, United States
| | | | - Tasha L Welch
- Mayo Clinic College of Medicine and Science, Departments of Anesthesiology and Perioperative Medicine, Rochester, United States
| | - Phillip J Schulte
- Mayo Clinic College of Medicine and Science, Division of Biomedical Statistics and Informatics, Health Sciences Research, Rochester, United States
| | - Toby N Weingarten
- Mayo Clinic College of Medicine and Science, Departments of Anesthesiology and Perioperative Medicine, Rochester, United States
| |
Collapse
|
8
|
Zaki U, Shakeel AS, Rauf Y, Raza M. Pituicytoma: A rare tumor of the sella. A case report and review of literature for diagnosis and management. Surg Neurol Int 2023; 14:220. [PMID: 37404513 PMCID: PMC10316132 DOI: 10.25259/sni_248_2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 06/13/2023] [Indexed: 07/06/2023] Open
Abstract
Background Pituicytomas are rare tumors of the pituitary gland derived from the ependymal cells and line the pituitary stalk and posterior lobe. These tumors are located in the vulnerable regions of the brain: Either in the sellar or suprasellar area. The location marks the difference in the clinical features of the tumor. Here, we report a case of histopathologically diagnosed pituicytoma of the sellar region. Literature is also reviewed and discussed to gain a better understanding of this rare disease. Case Description A 24-year-old female presented to the outpatient department with complaints of headache, diplopia, dizziness, and decreased vision in the right eye for 6 months. Computed tomography scan brain without contrast showed a well-defined hyperdense lesion in the sella without associated bony erosion. Her magnetic resonance imaging showed well defined rounded lesion in the pituitary fossa which was isointense on T1-weighted image and hyperintense on T2-weighted images. A presumptive diagnosis of pituitary adenoma was made. She underwent endoscopic endonasal transsphenoidal resection of pituitary mass. Intraoperatively, normal pituitary gland was visualized and there was a grayish-green-colored, jelly like tumor which was pulled gently. On 9th postoperative day, she presented with cerebrospinal fluid (CSF) rhinorrhea. She underwent endoscopic CSF leak repair. Her histopathology was concluded to be Pituicytoma. Conclusion Pituicytoma is an uncommon diagnosis. The surgical aim is to completely excise the tumor which results in complete cure, but incomplete resection may be performed due to high vascularity of this tumor. In case of incomplete excision, recurrence is common and adjuvant radiotherapy may be administered.
Collapse
Affiliation(s)
- Umaima Zaki
- Department of Neurosurgery, Patel Hospital, Karachi, Pakistan
| | | | - Yaseen Rauf
- Department of Neurosurgery, Patel Hospital, Karachi, Pakistan
| | - Muhammad Raza
- Department of Histopathology, Aga Khan University Hospital, Karachi, Pakistan
| |
Collapse
|
9
|
La Salvia A, Modica R, Rossi RE, Spada F, Rinzivillo M, Panzuto F, Faggiano A, Cinieri S, Fazio N. Targeting neuroendocrine tumors with octreotide and lanreotide: Key points for clinical practice from NET specialists. Cancer Treat Rev 2023; 117:102560. [PMID: 37088017 DOI: 10.1016/j.ctrv.2023.102560] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 04/08/2023] [Accepted: 04/12/2023] [Indexed: 04/25/2023]
Abstract
Octreotide and lanreotide are the two somatostatin analogs (SSA) currently available in clinical practice. They have been approved first to control the clinical syndrome (mainly carcinoid syndrome) associated with functioning neuroendocrine tumors (NET) and later for tumor growth control in advanced low/intermediate grade NET. Although evidence regarding their role, especially as antiproliferative therapy, has been increasing over the years some clinical indications remain controversial. Solicited by AIOM (Italian Association of Medical Oncology) a group of clinicians from various specialties, including medical oncology, endocrinology, and gastroenterology, deeply involved in NET for their clinical and research activity, addressed eight open questions, critically reviewing evidence and guidelines and sharing clinical take-home messages. The questions regarded the use of long-acting octreotide and lanreotide in the following settings: functioning and non-functioning NET refractory to label dose, first-line metastatic pulmonary NET, combination with other therapy with an antiproliferative intent, maintenance in NET responding to other therapies, adjuvant treatment, Ki-67-related cut-off, somatostatin receptor imaging, safety, and feasibility. The level of evidence is not absolute for the majority of these clinical contexts, so it is recommended to distinguish routine versus sporadic utilization in very selected cases. Mention of such specific issues by the main European guidelines (ENETS, European Neuroendocrine Tumor Society, and ESMO, European Society for Medical Oncology) was explored and their position reported. However, different clinical decisions on single patients could be made if the case is carefully discussed within a NET-dedicated multidisciplinary team.
Collapse
Affiliation(s)
- A La Salvia
- National Center for Drug Research and Evaluation, National Institute of Health (ISS), Rome, Italy.
| | - R Modica
- Department of Clinical Medicine and Surgery, Endocrinology Unit of Federico II University of Naples, Naples, Italy.
| | - R E Rossi
- Gastroenterology and Endoscopy Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, Milan, Italy.
| | - F Spada
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, IEO, European Institute of Oncology, IRCCS, Milan, Italy.
| | - M Rinzivillo
- ENETS Center of Excellence, Disease Unit, Sant'Andrea University Hospital, Rome, Italy.
| | - F Panzuto
- ENETS Center of Excellence, Disease Unit, Sant'Andrea University Hospital, Rome, Italy; Department of Medical-Surgical Sciences and Translational Medicine, Sapienza University of Rome, Rome, Italy.
| | - A Faggiano
- Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, ENETS Center of Excellence, Sapienza University of Rome, Rome, Italy.
| | - S Cinieri
- Medical Oncology Division and Breast Unit, ASL Brindisi, Brindisi, Italy.
| | - N Fazio
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, IEO, European Institute of Oncology, IRCCS, Milan, Italy.
| |
Collapse
|
10
|
Giampietro A, Menotti S, Chiloiro S, Pontecorvi A, De Marinis L, Bianchi A. De-escalation treatment with pasireotide for acromegaly: a long-term experience. Endocrine 2023; 80:505-510. [PMID: 36808072 DOI: 10.1007/s12020-023-03325-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 02/08/2023] [Indexed: 02/21/2023]
Abstract
INTRODUCTION Pasireotide long-acting release (LAR) is approved for second-line treatment of acromegaly. Starting pasireotide LAR 40 mg every 4 weeks is recommended and then up-titrate to 60 mg monthly in case of IGF-I uncontrolled levels. We present three patients treated with a de-escalation approach with pasireotide LAR. CASE 1: A 61-year-old female diagnosed with resistant acromegaly was treated with pasireotide LAR 60 mg every 28 days. When IGF-I reached the lower age range, therapy was decreased to pasireotide LAR 40 mg and then to 20 mg. In 2021 and 2022, IGF-I value remained within the normal range. CASE 2: A 40-year-old female diagnosed with resistant acromegaly underwent three neurosurgeries. In 2011, she was enrolled in the PAOLA study and assigned to pasireotide LAR 60 mg. Due to IGF-I overcontrol and radiological stability, therapy was downscaled to 40 mg in 2016 and to 20 mg in 2019. The patient developed hyperglycemia, which was treated with metformin. CASE 3: A 37-year-old male diagnosed with resistant acromegaly was treated with pasireotide LAR 60 mg in 2011. In 2018, therapy was decreased to 40 mg due to IGF-I overcontrol and in 2022 to 20 mg. He developed hyperglycemia, but HbA1c values remained under 48 nmol/L for 7 years. CONCLUSION De-escalation treatment with pasireotide LAR may allow a greater proportion of patients to achieve control of acromegaly, particularly in selected cases of clinically aggressive acromegaly potentially responsive to pasireotide (high IGF-I values, invasion of the cavernous sinuses, partial resistance to first-line somatostatin analogues and positive expression of somatostatin receptor 5). Another benefit may be IGF-I oversuppression overtime. The major risk seems to be hyperglycemia.
Collapse
Affiliation(s)
- Antonella Giampietro
- UOC Endocrinologia, Fondazione Policlinico A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Sara Menotti
- UOC Endocrinologia, Fondazione Policlinico A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Sabrina Chiloiro
- UOC Endocrinologia, Fondazione Policlinico A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168, Rome, Italy.
| | - Alfredo Pontecorvi
- UOC Endocrinologia, Fondazione Policlinico A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Laura De Marinis
- UOC Endocrinologia, Fondazione Policlinico A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Antonio Bianchi
- UOC Endocrinologia, Fondazione Policlinico A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| |
Collapse
|
11
|
Csaba Z, Dournaud P. Internalization of somatostatin receptors in brain and periphery. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 196:43-57. [PMID: 36813365 DOI: 10.1016/bs.pmbts.2022.09.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Somatostatin (SRIF) is a neuropeptide that acts as an important regulator of both endocrine and exocrine secretion and modulates neurotransmission in the central nervous system (CNS). SRIF also regulates cell proliferation in normal tissues and tumors. The physiological actions of SRIF are mediated by a family of five G protein-coupled receptors, called somatostatin receptor (SST) SST1, SST2, SST3, SST4, SST5. These five receptors share similar molecular structure and signaling pathways but they display marked differences in their anatomical distribution, subcellular localization and intracellular trafficking. The SST subtypes are widely distributed in the CNS and peripheral nervous system, in many endocrine glands and tumors, particularly of neuroendocrine origin. In this review, we focus on the agonist-dependent internalization and recycling of the different SST subtypes in vivo in the CNS, peripheral organs and tumors. We also discuss the physiological, pathophysiological and potential therapeutic effects of the intracellular trafficking of SST subtypes.
Collapse
Affiliation(s)
- Zsolt Csaba
- Université Paris Cité, NeuroDiderot, Inserm UMR, Paris, France
| | - Pascal Dournaud
- Université Paris Cité, NeuroDiderot, Inserm UMR, Paris, France.
| |
Collapse
|
12
|
Combined anti-tumor efficacy of somatostatin fusion protein and vaccinia virus on tumor cells with high expression of somatostatin receptors. Sci Rep 2022; 12:16885. [PMID: 36207478 PMCID: PMC9547013 DOI: 10.1038/s41598-022-21506-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 09/28/2022] [Indexed: 11/30/2022] Open
Abstract
Somatostatin, a growth hormone-release inhibiting peptide, exerts antiproliferative and antiangiogenic effects on tumor cells. However, the short half-life of somatostatin limits its application in human therapy, and long-acting somatostatin fusion protein is also limited by its severe terminal degradation. Therefore, oncolytic virus delivery system was introduced to express somatostatin fusion protein and the anti-tumor effects of both somatostatin and oncolytic virus were combined to destroy tumor tissues. Here, a vaccinia VG9/(SST-14)2-HSA recombinant was constructed by replacing somatostatin fusion gene into TK locus of attenuated VG9 strain via homologous recombination. Results showed that vaccinia VG9/(SST-14)2-HSA possessed a combined anti-tumor effect on sstr-positive tumor cells in vitro. In the tumor burden models, BALB/c mice with complete immunity are most suitable for evaluating tumor regression and immune activation. Complete tumor regression was observed in 3 out of 10 mice treated with vaccinia VG9/TK− or VG9/(SST-14)2-HSA, and the survival of all mice in both groups was significantly prolonged. Besides, vaccinia VG9/(SST-14)2-HSA is more effective in prolonging survival than VG9/TK−. Vaccinia VG9/(SST-14)2-HSA exerts a combined anti-tumor efficacy including the oncolytic ability provided by the virus and the anti-tumor effect contributed by (SST-14)2-HSA, which is expected to become a potent therapeutic agent for cancer treatment.
Collapse
|
13
|
Liu B, Li P, He S, Xing S, Chen C, Liu L, Li ZH. Chronic exposure to tralopyril induced abnormal growth and calcium regulation of turbot (Scophthalmus maximus). CHEMOSPHERE 2022; 299:134405. [PMID: 35364078 DOI: 10.1016/j.chemosphere.2022.134405] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/12/2022] [Accepted: 03/21/2022] [Indexed: 06/14/2023]
Abstract
Tralopyril is an emerging marine antifouling agent with limited data on its effects on fish growth and calcium regulation. To determine the changes induced by long-term exposure to tralopyril, multi-levels (such as molecular, biochemical, and individual levels) responses were measured in turbot at different concentrations (1 μg/L, 20 μg/L). The results showed that 1 μg/L mainly affected the immune response, while 20 μg/L affected the synthesis and metabolism of steroids and fat. However, different concentrations of tralopyril affected the synthesis, secretion and action of parathyroid hormone and growth hormone. The expression of GH/IGF axis gene and the level of growth hormone increased significantly, leading to abnormal growth. The energy tradeoff between immunity and growth at 1 μg/L tralopyril pressure may inhibit growth. The change of Ca2+ level was accompanied by the disturbance of PTH-related gene expression. The results of molecular docking showed that the disturbance of Ca2+ regulation might be attributed to the inhibition of vitamin D receptor by tralopyril, which affected the vitamin D signaling pathway. This study provides scientific data for the in-depth understanding and risk assessment of the toxicological effects of tralopyril and reveals the potential threat of tralopyril to environmental health.
Collapse
Affiliation(s)
- Bin Liu
- Marine College, Shandong University, Weihai, Shandong, 264209, China
| | - Ping Li
- Marine College, Shandong University, Weihai, Shandong, 264209, China
| | - Shuwen He
- Marine College, Shandong University, Weihai, Shandong, 264209, China
| | - Shaoying Xing
- Marine College, Shandong University, Weihai, Shandong, 264209, China
| | - Chengzhuang Chen
- Marine College, Shandong University, Weihai, Shandong, 264209, China
| | - Ling Liu
- Marine College, Shandong University, Weihai, Shandong, 264209, China
| | - Zhi-Hua Li
- Marine College, Shandong University, Weihai, Shandong, 264209, China.
| |
Collapse
|
14
|
Coopmans EC, van der Lely AJ, Neggers SJCMM. Approach to the Patient With Treatment-resistant Acromegaly. J Clin Endocrinol Metab 2022; 107:1759-1766. [PMID: 35090028 PMCID: PMC9315163 DOI: 10.1210/clinem/dgac037] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Indexed: 11/29/2022]
Abstract
Although most tumors in patients with acromegaly are benign and are cured or controlled by surgery and/or first-generation somatostatin receptor ligands therapy, some can behave more aggressively and are resistant to these standard therapies. Acromegaly, if left untreated, is a rare and chronic disorder, commonly caused by a GH-producing pituitary adenoma and is associated with significant comorbidities and an increased mortality. Transsphenoidal surgery is considered the mainstay of acromegaly management, but medical therapy has an increasingly important role. However, disease activity is not fully controlled in a significant number of patients treated with surgery and/or high-dose first-generation somatostatin receptor ligand monotherapy. In these circumstances, therefore, repeated surgery, second-line medical therapy, and radiotherapy, alone or combined as multimodal therapeutic strategies should be considered, in a patient-centered perspective.
Collapse
Affiliation(s)
- Eva C Coopmans
- Department of Medicine, Section Endocrinology, Pituitary Center Rotterdam, Erasmus University Medical Center, 3000 CA Rotterdam, the Netherlands
| | - Aart J van der Lely
- Department of Medicine, Section Endocrinology, Pituitary Center Rotterdam, Erasmus University Medical Center, 3000 CA Rotterdam, the Netherlands
| | - Sebastian J C M M Neggers
- Correspondence: S. Neggers, Erasmus University Medical Center Rotterdam, PO box 2040, 3000 CA Rotterdam, The Netherlands.
| |
Collapse
|
15
|
Role of Somatostatin Signalling in Neuroendocrine Tumours. Int J Mol Sci 2022; 23:ijms23031447. [PMID: 35163374 PMCID: PMC8836266 DOI: 10.3390/ijms23031447] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/24/2022] [Accepted: 01/25/2022] [Indexed: 02/06/2023] Open
Abstract
Somatostatin (SST) is a small peptide that exerts inhibitory effects on a wide range of neuroendocrine cells. Due to the fact that somatostatin regulates cell growth and hormone secretion, somatostatin receptors (SSTRs) have become valuable targets for the treatment of different types of neuroendocrine tumours (NETs). NETs are a heterogeneous group of tumours that can develop in various parts of the body, including the digestive system, lungs, and pituitary. NETs are usually slow growing, but they are often diagnosed in advanced stages and can display aggressive behaviour. The mortality rate of NETs is not outstandingly increased compared to other malignant tumours, even in the metastatic setting. One of the intrinsic properties of NETs is the expression of SSTRs that serve as drug targets for SST analogues (SSAs), which can delay tumour progression and downregulate hormone overproduction. Additionally, in many NETs, it has been demonstrated that the SSTR expression level provides a prognostic value in predicting a therapeutic response. Furthermore, higher a SSTR expression correlates with a better survival rate in NET patients. In recent studies, other epigenetic regulators affecting SST signalling or SSA–mTOR inhibitor combination therapy in NETs have been considered as novel strategies for tumour control. In conclusion, SST signalling is a relevant regulator of NET functionality. Alongside classical SSA treatment regimens, future advanced therapies and treatment modalities are expected to improve the disease outcomes and overall health of NET patients.
Collapse
|
16
|
Vemuri KR, Balcerek MI, McGowan TE, Love A, Chapman PR. VIPoma:
an unsuspecting culprit of severe secretory diarrhoea in a human immunodeficiency virus‐infected patient. Intern Med J 2022; 52:125-129. [DOI: 10.1111/imj.15655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 06/16/2021] [Accepted: 06/29/2021] [Indexed: 11/27/2022]
Affiliation(s)
- Kanthi R. Vemuri
- Department of Infectious Diseases Royal Brisbane and Women's Hospital Brisbane Queensland Australia
- Herston Infectious Diseases Institute (HeIDI) Metro North Hospitals and Health Services Brisbane Queensland Australia
| | - Matthew I. Balcerek
- Department of Endocrinology and Diabetes Royal Brisbane and Women's Hospital Brisbane Queensland Australia
| | - Tom E. McGowan
- Department of Infectious Diseases Royal Brisbane and Women's Hospital Brisbane Queensland Australia
| | - Amanda Love
- Department of Endocrinology and Diabetes Royal Brisbane and Women's Hospital Brisbane Queensland Australia
| | - Paul R. Chapman
- Department of Infectious Diseases Royal Brisbane and Women's Hospital Brisbane Queensland Australia
| |
Collapse
|
17
|
Dicitore A, Saronni D, Gaudenzi G, Carra S, Cantone MC, Borghi MO, Persani L, Vitale G. Long-term effects of somatostatin analogues in rat GH-secreting pituitary tumor cell lines. J Endocrinol Invest 2022; 45:29-41. [PMID: 34128215 PMCID: PMC8741688 DOI: 10.1007/s40618-021-01609-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 06/03/2021] [Indexed: 11/18/2022]
Abstract
PURPOSE First-generation somatostatin analogs, octreotide (OCT) and lanreotide, are the cornerstone for the medical treatment of growth hormone (GH)-secreting pituitary tumors. A new multireceptor analog, such as pasireotide (PAS), showed better activity than OCT in long-term treatment of patients with acromegaly, but modulation of intracellular key processes is still unclear in vitro. In this study, we evaluated the antitumor activity of OCT and PAS in two GH-secreting pituitary tumor cell lines, GH3 and GH4C1, after a long-term incubation. METHODS The effects of PAS and OCT on the cell viability, cell cycle, apoptosis, GH secretion, and tumor-induced angiogenesis have been evaluated through a colorimetric method (MTS Assay), DNA flow cytometry with propidium iodide, and Annexin V-FITC/propidium iodide staining, ELISA assay and zebrafish platform, respectively. RESULTS PAS showed a more potent antitumor activity compared to OCT in GH3 cell line exerted through inhibition of cell viability, perturbation of cell cycle progression, and induction of apoptosis after 6 days of incubation. A concomitant decrease in GH secretion has been observed after 2 days of incubation only with PAS. No effect on tumor-induced angiogenesis has been reported after treatment with OCT or PAS in zebrafish/tumor xenograft model. CONCLUSION Long-term incubation with PAS showed a more potent antitumor activity than that reported after OCT in GH3 cells, mainly modulated by a cell cycle perturbation and a relevant induction in apoptosis.
Collapse
Affiliation(s)
- A Dicitore
- Laboratory of Geriatric and Oncologic Neuroendocrinology Research, Istituto Auxologico Italiano, IRCCS, Via Zucchi 18, 20095, Cusano Milanino, MI, Italy
| | - D Saronni
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - G Gaudenzi
- Laboratory of Geriatric and Oncologic Neuroendocrinology Research, Istituto Auxologico Italiano, IRCCS, Via Zucchi 18, 20095, Cusano Milanino, MI, Italy
| | - S Carra
- Laboratory of Endocrine and Metabolic Research, Istituto Auxologico Italiano, IRCCS, Milan, Italy
| | - M C Cantone
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - M O Borghi
- Experimental Laboratory of Immuno-rheumatology, Istituto Auxologico Italiano, IRCCS, Milan, Italy
- Department of Clinical Sciences and Community Health (DISCCO), University of Milan, Milan, Italy
| | - L Persani
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
- Laboratory of Endocrine and Metabolic Research, Istituto Auxologico Italiano, IRCCS, Milan, Italy
| | - G Vitale
- Laboratory of Geriatric and Oncologic Neuroendocrinology Research, Istituto Auxologico Italiano, IRCCS, Via Zucchi 18, 20095, Cusano Milanino, MI, Italy.
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy.
| |
Collapse
|
18
|
Jimenez-Fonseca P, Carmona-Bayonas A, Lamarca A, Barriuso J, Castaño A, Benavent M, Alonso V, Riesco MDC, Alonso-Gordoa T, Custodio A, Sanchez Canovas M, Hernando J, López C, La Casta A, Fernandez Montes A, Marazuela M, Crespo G, Diaz JA, Feliciangeli E, Gallego J, Llanos M, Segura A, Vilardell F, Percovich JC, Grande E, Capdevila J, Valle J, Garcia-Carbonero R. External Validity of Somatostatin Analogs Trials in Advanced Neuroendocrine Neoplasms: The GETNE-TRASGU Study. Neuroendocrinology 2022; 112:88-100. [PMID: 33508849 DOI: 10.1159/000514808] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/27/2021] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Somatostatin analogs (SSA) prolong progression-free survival (PFS) in patients with well-differentiated gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs). However, the eligibility criteria in randomized clinical trials (RCTs) have been restricted, which contrasts with the vast heterogeneity found in NENs. METHODS We identified patients with well-differentiated (Ki-67% ≤20%), metastatic GEP-NENs treated in first line with SSA monotherapy from the Spanish R-GETNE registry. The therapeutic effect was evaluated using a Bayesian Cox model. The objective was to compare survival-based outcomes from real-world clinical practice versus RCTs. RESULTS The dataset contained 535 patients with a median age of 62 years (range: 26-89). The median Ki-67% was 4 (range: 0-20). The most common primary tumor sites were as follows: midgut, 46%; pancreas, 34%; unknown primary, 10%; and colorectal, 10%. Half of the patients received octreotide LAR (n = 266) and half, lanreotide autogel (n = 269). The median PFS was 28.0 months (95% CI: 22.1-32.0) for octreotide versus 30.1 months (95% CI: 23.1-38.0) for lanreotide. The overall hazard ratio for lanreotide versus octreotide was 0.90 (95% credible interval: 0.71-1.12). The probability of effect sizes >30% with lanreotide versus octreotide was 2 and 6% for midgut and foregut NENs, respectively. CONCLUSION Our study evaluated the external validity of RCTs examining SSAs in the real world, as well as the main effect-modifying factors (progression status, symptoms, tumor site, specific metastases, and analytical data). Our results indicate that both octreotide LAR and lanreotide autogel had a similar effect on PFS. Consequently, both represent valid alternatives in patients with well-differentiated, metastatic GEP-NENs.
Collapse
Affiliation(s)
- Paula Jimenez-Fonseca
- Medical Oncology Department, Hospital Universitario Central de Asturias, ISPA, Oviedo, Spain
| | - Alberto Carmona-Bayonas
- Hematology and Medical Oncology Department, Hospital Universitario Morales Meseguer, UMU, IMIB, Murcia, Spain
| | - Angela Lamarca
- Medical Oncology Department, The Christie NHS Foundation Trust, ENETS Centre of Excellence, Manchester, United Kingdom
- Division of Cancer Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Jorge Barriuso
- Medical Oncology Department, The Christie NHS Foundation Trust, ENETS Centre of Excellence, Manchester, United Kingdom
- Division of Cancer Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Angel Castaño
- Pathology Department, Hospital Universitario de Fuenlabrada, Madrid, Spain
| | - Marta Benavent
- Medical Oncology Department, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS), Sevilla, Spain
| | - Vicente Alonso
- Medical Oncology Department, Hospital Universitario Miguel Servet, Zaragoza, Spain
| | - Maria Del Carmen Riesco
- Medical Oncology Department, Hospital Universitario Doce de Octubre, IIS imas12, UCM, CNIO, CIBERONC, Madrid, Spain
| | - Teresa Alonso-Gordoa
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Ana Custodio
- Medical Oncology Department, Hospital Universitario La Paz, CIBERONC CB16/12/00398, Madrid, Spain
| | - Manuel Sanchez Canovas
- Hematology and Medical Oncology Department, Hospital Universitario Morales Meseguer, UMU, IMIB, Murcia, Spain
| | - Jorge Hernando
- Medical Oncology Department, Hospital Universitario Vall d'Hebron, Vall Hebron Institute of Oncology (VHIO), Autonomous University of Barcelona, Barcelona, Spain
| | - Carlos López
- Medical Oncology Department, Hospital Universitario Marqués de Valdecilla, IDIVAL, Santander, Spain
| | - Adelaida La Casta
- Medical Oncology Department, Hospital Universitario Donostia, San Sebastián, Spain
| | - Ana Fernandez Montes
- Medical Oncology Department, Complexo Hospitalario Universitario de Ourense, Ourense, Spain
| | - Mónica Marazuela
- Endocrinology Department, Hospital Universitario de la Princesa, Madrid, Spain
| | - Guillermo Crespo
- Medical Oncology Department, Complejo Asistencial Universitario de Burgos, Burgos, Spain
| | - Jose Angel Diaz
- Endocrinology Department, Hospital Universitario Clínico San Carlos, Madrid, Spain
| | - Eduardo Feliciangeli
- Medical Oncology Department, Hospital Universitario Santa Lucia, Cartagena, Spain
| | - Javier Gallego
- Medical Oncology Department, Hospital General Universitario de Elche, Elche, Spain
| | - Marta Llanos
- Medical Oncology Department, Hospital Universitario de Canarias, Universidad de La Laguna, Tenerife, Spain
| | - Angel Segura
- Medical Oncology Department, Hospital Universitario La Fe, Valencia, Spain
| | - Felip Vilardell
- Pathology Department, Hospital Universitari Arnau de Vilanova, Lleida, Spain
| | | | - Enrique Grande
- Medical Oncology Department, MD Anderson Cancer Center Madrid, Madrid, Spain
| | - Jaume Capdevila
- Medical Oncology Department, Hospital Universitario Vall d'Hebron, Vall Hebron Institute of Oncology (VHIO), Autonomous University of Barcelona, Barcelona, Spain
| | - Juan Valle
- Medical Oncology Department, The Christie NHS Foundation Trust, ENETS Centre of Excellence, Manchester, United Kingdom
- Division of Cancer Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Rocio Garcia-Carbonero
- Medical Oncology Department, Hospital Universitario Doce de Octubre, IIS imas12, UCM, CNIO, CIBERONC, Madrid, Spain
| |
Collapse
|
19
|
Yu F, Zhang T, Fu F, Wang A, Liu X. Preparation of Long-acting Somatostatin and GnRH Analogues and their Applications in Tumor Therapy. Curr Drug Deliv 2021; 19:5-16. [PMID: 34951573 DOI: 10.2174/1567201819666211224113311] [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: 10/18/2021] [Revised: 11/24/2021] [Accepted: 12/05/2021] [Indexed: 11/22/2022]
Abstract
Hormonal drugs are essential treatment options for some hormone-dependent or hormone-sensitive tumors. The common dosage forms of hormonal drugs have a short half-life. Hence, frequent administration is needed, which results in poor patient compliance. Nevertheless, using drug delivery technology, somatostatin analogues (SSAs) and gonadotropin-releasing hormone (GnRH) analogues are prepared into long-acting formulations that can significantly prolong the action time of these drugs, reducing medication frequency and increasing patient compliance. Such drugs are advantageous when treating acromegaly, gastroenteropancreatic neuroendocrine tumors (GEP-NETs), breast cancer, prostate cancer, and other diseases having a relatively long course. SSAs and GnRH analogues are two typical hormonal drugs, the long-acting formulations of which are essential in clinical practice. This review summarized the preparation methods and clinical application of long-acting formulations in cancer. Further, the action mechanism and new research of SSAs and GnRH analogues were discussed, and suggestions related to the development of long-acting SSAs and GnRH analogues were provided.
Collapse
Affiliation(s)
- Fang Yu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Shandong University), Ministry of Education, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, China
| | - Tingting Zhang
- State Key Laboratory of Long-acting and Targeting Drug Delivery Technologies, Yantai, China
| | - Fenghua Fu
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, School of Pharmacy, Yantai University, China
| | - Aiping Wang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, School of Pharmacy, Yantai University, China
| | - Xinyong Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Shandong University), Ministry of Education, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Ji'nan, China
| |
Collapse
|
20
|
Pharmacological Characterization of Veldoreotide as a Somatostatin Receptor 4 Agonist. Life (Basel) 2021; 11:life11101075. [PMID: 34685446 PMCID: PMC8541358 DOI: 10.3390/life11101075] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 10/01/2021] [Accepted: 10/07/2021] [Indexed: 12/12/2022] Open
Abstract
Veldoreotide, a somatostatin analogue, binds to the somatostatin receptors (SSTR) 2, 4, and 5. The current aim was to assess its pharmacological activity as an SSTR4 agonist. G-protein signaling was assessed using a fluorescence-based membrane potential assay in human embryonic kidney 293 (HEK293) cells stably co-expressing G-protein-coupled inwardly rectifying potassium 2 channels and the individual SSTR2, SSTR4, and SSTR5, and in human BON-1 cells stably expressing these SSTRs. Veldoreotide effects on chromogranin A (CgA) secretion and cell proliferation were examined in BON-1 cells. In HEK293 transfected cells, veldoreotide showed a high efficacy for activating the SSTR4; octreotide and pasireotide had little activity (Emax, 99.5% vs. 27.4% and 52.0%, respectively). Veldoreotide also activated SSTR2 and SSTR5 (Emax, 98.4% and 96.9%, respectively). In BON-1 cells, veldoreotide activated SSTR2, SSTR4, and SSTR5 with high potency and efficacy. CgA secretion was decreased to a greater degree in the BON-1 cells expressing SSTR4 versus the cells expressing SSTR2 and SSTR5 (65.3% vs. 80.3% and 77.6%, respectively). In the BON-1 cells expressing SSTR4, veldoreotide inhibited cell proliferation more than somatostatin SS-14 (71.2% vs. 79.7%) and to a similar extent as the SSTR4 agonist J-2156 in the presence of SSTR2 and SSTR5 antagonists. Veldoreotide is a full agonist of SSTR2, SSTR4, and SSTR5.
Collapse
|
21
|
Song YH, Yoon J, Lee SH. The role of neuropeptide somatostatin in the brain and its application in treating neurological disorders. Exp Mol Med 2021; 53:328-338. [PMID: 33742131 PMCID: PMC8080805 DOI: 10.1038/s12276-021-00580-4] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/23/2021] [Accepted: 01/25/2021] [Indexed: 02/07/2023] Open
Abstract
Somatostatin (SST) is a well-known neuropeptide that is expressed throughout the brain. In the cortex, SST is expressed in a subset of GABAergic neurons and is known as a protein marker of inhibitory interneurons. Recent studies have identified the key functions of SST in modulating cortical circuits in the brain and cognitive function. Furthermore, reduced expression of SST is a hallmark of various neurological disorders, including Alzheimer's disease and depression. In this review, we summarize the current knowledge on SST expression and function in the brain. In particular, we describe the physiological roles of SST-positive interneurons in the cortex. We further describe the causal relationship between pathophysiological changes in SST function and various neurological disorders, such as Alzheimer's disease. Finally, we discuss potential treatments and possibility of novel drug developments for neurological disorders based on the current knowledge on the function of SST and SST analogs in the brain derived from experimental and clinical studies.
Collapse
Affiliation(s)
- You-Hyang Song
- grid.37172.300000 0001 2292 0500Department of Biological Sciences, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon 34141 Republic of Korea
| | - Jiwon Yoon
- grid.37172.300000 0001 2292 0500Department of Biological Sciences, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon 34141 Republic of Korea
| | - Seung-Hee Lee
- grid.37172.300000 0001 2292 0500Department of Biological Sciences, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon 34141 Republic of Korea
| |
Collapse
|
22
|
Spiliopoulou M, Karavassili F, Triandafillidis DP, Valmas A, Fili S, Kosinas C, Barlos K, Barlos KK, Morin M, Reinle-Schmitt ML, Gozzo F, Margiolaki I. New perspectives in macromolecular powder diffraction using single-photon-counting strip detectors: high-resolution structure of the pharmaceutical peptide octreotide. ACTA CRYSTALLOGRAPHICA A-FOUNDATION AND ADVANCES 2021; 77:186-195. [PMID: 33944797 DOI: 10.1107/s2053273321001698] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 02/11/2021] [Indexed: 11/10/2022]
Abstract
Advances in instrumentation, as well as the development of powerful crystallographic software have significantly facilitated the collection of high-resolution diffraction data and have made X-ray powder diffraction (XRPD) particularly useful for the extraction of structural information; this is true even for complex molecules, especially when combined with synchrotron radiation. In this study, in-line with past instrumental profile studies, an improved data collection strategy exploiting the MYTHEN II detector system together with significant beam focusing and tailored data collection options was introduced and optimized for protein samples at the Material Science beamline at the Swiss Light Source. Polycrystalline precipitates of octreotide, a somatostatin analog of particular pharmaceutical interest, were examined with this novel approach. XRPD experiments resulted in high angular and d-spacing (1.87 Å) resolution data, from which electron-density maps of enhanced quality were extracted, revealing the molecule's structural properties. Since microcrystalline precipitates represent a viable alternative for administration of therapeutic macromolecules, XRPD has been acknowledged as the most applicable tool for examining a wide spectrum of physicochemical properties of such materials and performing studies ranging from phase identification to complete structural characterization.
Collapse
Affiliation(s)
- Maria Spiliopoulou
- Department of Biology, Section of Genetics, Cell Biology and Development, University of Patras, Patras, GR-26500, Greece
| | - Fotini Karavassili
- Department of Biology, Section of Genetics, Cell Biology and Development, University of Patras, Patras, GR-26500, Greece
| | | | - Alexandros Valmas
- Department of Biology, Section of Genetics, Cell Biology and Development, University of Patras, Patras, GR-26500, Greece
| | - Stavroula Fili
- Department of Biology, Section of Genetics, Cell Biology and Development, University of Patras, Patras, GR-26500, Greece
| | - Christos Kosinas
- Department of Biology, Section of Genetics, Cell Biology and Development, University of Patras, Patras, GR-26500, Greece
| | | | | | - Mickael Morin
- Excelsus Structural Solutions (Swiss) AG, Park Innovaare, Villigen, 5234, Switzerland
| | | | - Fabia Gozzo
- Excelsus Structural Solutions (Swiss) AG, Park Innovaare, Villigen, 5234, Switzerland
| | - Irene Margiolaki
- Department of Biology, Section of Genetics, Cell Biology and Development, University of Patras, Patras, GR-26500, Greece
| |
Collapse
|
23
|
Ionovici N, Carsote M, Terzea DC, Predescu AM, Rauten AM, Popescu M. Somatostatin receptors in normal and acromegalic somatotroph cells: the U-turn of the clinician to immunohistochemistry report - a review. ROMANIAN JOURNAL OF MORPHOLOGY AND EMBRYOLOGY 2021; 61:353-359. [PMID: 33544787 PMCID: PMC7864306 DOI: 10.47162/rjme.61.2.05] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
This is a narrative review of literature introducing somatostatin receptors (SSTRs) as part of understanding the somatotroph cells since they are positive in normal cells but also in tumoral cells as seen in somatotropinoma, a growth hormone (GH)-producing neoplasia, which causes acromegaly. They are five subtypes of SSTRs (1 to 5), which are immunohistochemically positive in different proportions in somatotropinomas. SSTR types 2 and 5 are most frequent in GH-secreting adenomas and they are both targeted by medical therapy with somatostatin analogues (SSTAs) like first generation Octreotide and Lanreotide (mainly targeting SSTR2) and second generation Pasireotide (with highest affinity for SSTR5), thus heterogeneous SSTRs configuration into the tumor explains different pattern of response to treatment and it might predict it once the SSTRs immunostaining is performed. Monoclonal antibodies are used for immunohistochemical detection of SSTRs; currently, a lack of standardization is presented, and scoring systems, such as Volante, H-score or human epidermal growth factor receptor 2 (HER2)-score, are applied. Immunoreactive markers like SSTRs are the U-turn in clinical practice regarding somatotropinomas since the configuration of subtypes 2 and 5 explains the responsiveness to medical therapy like SSTA. Further achievement of disease control is imperiously necessary because acromegaly has an increased rate of morbidity and mortality.
Collapse
Affiliation(s)
- Nina Ionovici
- Department of Endocrinology, Carol Davila University of Medicine and Pharmacy, C. I. Parhon National Institute of Endocrinology, Bucharest, Romania;
| | | | | | | | | | | |
Collapse
|
24
|
Hou ZS, Xin YR, Zeng C, Zhao HK, Tian Y, Li JF, Wen HS. GHRH-SST-GH-IGF axis regulates crosstalk between growth and immunity in rainbow trout (Oncorhynchus mykiss) infected with Vibrio anguillarum. FISH & SHELLFISH IMMUNOLOGY 2020; 106:887-897. [PMID: 32866610 DOI: 10.1016/j.fsi.2020.08.037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 08/15/2020] [Accepted: 08/20/2020] [Indexed: 06/11/2023]
Abstract
An energy trade-off is existed between immunological competence and growth. The axis of growth hormone releasing hormone, somatostatin, growth hormone, insulin-like growth factor (GHRH-SST-GH-IGF axis) regulates growth performances and immune competences in rainbow trout (Oncorhynchus mykiss). The salmonid-specific whole genome duplication event is known to result in duplicated copies of several key genes in GHRH-SST-GH-IGF axis. In this study, we evaluated the physiological functions of GHRH-SST-GH-IGF axis in regulating crosstalk between growth and immunity. Based on principal components analysis (PCA), we observed the overall expression profiles of GHRH-SST-GH-IGF axis were significantly altered by Vibrio anguillarum infection. Trout challenged with Vibrio anguillarum showed down-regulated igf1s subtypes and up-regulated igfbp1a1. The brain sst genes (sst1a, sst1b, sst3b and sst5) and igfpbs genes (igfbp4s and igfbp5b2) were significantly affected by V. anguillarum infection, while the igfbp4s, igfbp5s, igfbp6s and igf2bps genes showed significant changes in peripheral immune tissues in response to V. anguillarum infection. Gene enrichment analyses showed functional and signaling pathways associated with apoptosis (such as p53, HIF-1 or FoxO signaling) were activated. We further proposed a possible model that describes the IGF and IGFBPs-regulated interaction between cell growth and programmed death. Our study provided new insights into the physiological functions and potentially regulatory mechanisms of the GHRH-SST-GH-IGF axis, indicating the pleiotropic effects of GHRH-SST-GH-IGF axis in regulating crosstalk between growth and immunity in trout.
Collapse
Affiliation(s)
- Zhi-Shuai Hou
- Key Laboratory of Mariculture, Ocean University of China, Ministry of Education (KLMME), Qingdao, China.
| | - Yuan-Ru Xin
- Key Laboratory of Mariculture, Ocean University of China, Ministry of Education (KLMME), Qingdao, China
| | - Chu Zeng
- Key Laboratory of Mariculture, Ocean University of China, Ministry of Education (KLMME), Qingdao, China
| | - Hong-Kui Zhao
- Key Laboratory of Mariculture, Ocean University of China, Ministry of Education (KLMME), Qingdao, China
| | - Yuan Tian
- Key Laboratory of Mariculture, Ocean University of China, Ministry of Education (KLMME), Qingdao, China
| | - Ji-Fang Li
- Key Laboratory of Mariculture, Ocean University of China, Ministry of Education (KLMME), Qingdao, China
| | - Hai-Shen Wen
- Key Laboratory of Mariculture, Ocean University of China, Ministry of Education (KLMME), Qingdao, China.
| |
Collapse
|
25
|
Sakata K, Fujimori K, Komaki S, Furuta T, Sugita Y, Ashida K, Nomura M, Morioka M. Pituitary Gangliocytoma Producing TSH and TRH: A Review of "Gangliocytomas of the Sellar Region". J Clin Endocrinol Metab 2020; 105:5876003. [PMID: 32706866 PMCID: PMC7451506 DOI: 10.1210/clinem/dgaa474] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 07/17/2020] [Indexed: 02/06/2023]
Abstract
PURPOSE Pituitary gangliocytomas (GCs) are rare neuronal tumors that present with endocrinological disorders, such as acromegaly, amenorrhea-galactorrhea syndrome, and Cushing's disease. Most pituitary GCs coexist with pituitary adenomas pathologically and are diagnosed as mixed gangliocytoma-adenomas. Herein, we report a case of 45-year-old man who presented with the syndrome of inappropriate secretion of thyroid-stimulating hormone (SITSH) and discuss the pathogenesis of pituitary GCs. METHODS Pituitary magnetic resonance imaging showed an 8-mm homogeneous and poorly enhanced mass inside the pituitary gland. Endoscopic transsphenoidal surgery was performed under a preoperative diagnosis of thyrotroph adenoma. However, the tumor was finally diagnosed as gangliocytoma without an adenomatous component. The tumor was further analyzed via immunohistochemistry and electron microscopy. Additionally, we searched MEDLINE and PubMed for previously published cases of isolated pituitary GCs and analyzed the reported clinicopathological findings. RESULTS The patient showed complete clinical and endocrinological recovery after an operation. The tumor was positive for thyrotropin (TSH), TSH-releasing hormone (TRH), Pit-1, GATA-2, and most neuronal markers. Electron microscopy demonstrated the presence of intracytoplasmic secretory granules and neuronal processes. Co-secreting hypothalamic and pituitary hormone inside the tumor indicated autocrine/paracrine endocrinological stimulation. CONCLUSION Herein, we report a case of SITSH caused by an isolated pituitary gangliocytoma, expressing both TSH and TRH, which, to our best knowledge, is the first reported case of such a condition. The multidirectional differentiation and multihormonal endocrine characteristics of these tumors indicate that they are a member of neuroendocrine neoplasms, further supporting that they are derived from neural crest cells.
Collapse
Affiliation(s)
- Kiyohiko Sakata
- Department of Neurosurgery, Kurume University, School of Medicine, Fukuoka, Japan
- Correspondence and Reprint Requests: Kiyohiko Sakata, MD, Department of Neurosurgery, Kurume University School of Medicine, 67 Asahimachi, Kurume, Fukuoka 830-0011, Japan. E-mail: .
| | - Kana Fujimori
- Department of Neurosurgery, Kurume University, School of Medicine, Fukuoka, Japan
| | - Satoru Komaki
- Department of Neurosurgery, Kurume University, School of Medicine, Fukuoka, Japan
| | - Takuya Furuta
- Department of Pathology, Kurume University, School of Medicine, Fukuoka, Japan
| | - Yasuo Sugita
- Department of Neuropathology, Neurology Center, St. Mary’s Hospital, Fukuoka, Japan
| | - Kenji Ashida
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kurume University School of Medicine, Fukuoka, Japan
| | - Masatoshi Nomura
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kurume University School of Medicine, Fukuoka, Japan
| | - Motohiro Morioka
- Department of Neurosurgery, Kurume University, School of Medicine, Fukuoka, Japan
| |
Collapse
|
26
|
Akaike T, Qazi J, Anderson A, Behnia FS, Shinohara MM, Akaike G, Hippe DS, Thomas H, Takagishi SR, Lachance K, Park SY, Tarabadkar ES, Iyer JG, Blom A, Parvathaneni U, Vesselle H, Nghiem P, Bhatia S. High somatostatin receptor expression and efficacy of somatostatin analogues in patients with metastatic Merkel cell carcinoma. Br J Dermatol 2020; 184:319-327. [PMID: 32320473 DOI: 10.1111/bjd.19150] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/17/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Merkel cell carcinoma (MCC) is an aggressive, high-grade, cutaneous neuroendocrine tumour (NET). Agents blocking programmed death 1/programmed death ligand 1 have efficacy in metastatic MCC (mMCC), but half of patients do not derive durable benefit. Somatostatin analogues (SSAs) are commonly used to treat low- and moderate-grade NETs that express somatostatin receptors (SSTRs). OBJECTIVES To assess SSTR expression and the efficacy of SSAs in mMCC, a high-grade NET. Methods In this retrospective study of 40 patients with mMCC, SSTR expression was assessed radiologically by somatostatin receptor scintigraphy (SRS; n = 39) and/or immunohistochemically when feasible (n = 9). Nineteen patients (18 had SRS uptake in MCC tumours) were treated with SSA. Disease control was defined as progression-free survival (PFS) of ≥ 120 days after initiation of SSA. RESULTS Thirty-three of 39 patients (85%) had some degree (low 52%, moderate 23%, high 10%) of SRS uptake. Of 19 patients treated with SSA, seven had a response-evaluable target lesion; three of these seven patients (43%) experienced disease control, with a median PFS of 237 days (range 152-358). Twelve of 19 patients did not have a response-evaluable lesion due to antecedent radiation; five of these 12 (42%) experienced disease control (median PFS of 429 days, range 143-1757). The degree of SSTR expression (determined by SRS and/or immunohistochemistry) did not correlate significantly with the efficacy endpoints. CONCLUSIONS In contrast to other high-grade NETs, mMCC tumours appear frequently to express SSTRs. SSAs can lead to clinically meaningful disease control with minimal side-effects. Targeting of SSTRs using SSA or other novel approaches should be explored further for mMCC.
Collapse
Affiliation(s)
- T Akaike
- Division of Dermatology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - J Qazi
- Division of Dermatology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - A Anderson
- Division of Dermatology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - F S Behnia
- Department of Radiology, University of Washington, Seattle, WA, USA
| | - M M Shinohara
- Division of Dermatology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - G Akaike
- Department of Radiology, University of Washington, Seattle, WA, USA
| | - D S Hippe
- Department of Radiology, University of Washington, Seattle, WA, USA
| | - H Thomas
- Division of Dermatology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - S R Takagishi
- Division of Dermatology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - K Lachance
- Division of Dermatology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - S Y Park
- Division of Dermatology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - E S Tarabadkar
- Division of Dermatology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - J G Iyer
- Division of Dermatology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - A Blom
- Division of Dermatology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - U Parvathaneni
- Department of Radiation Oncology, University of Washington, Seattle, WA, USA
| | - H Vesselle
- Department of Radiology, University of Washington, Seattle, WA, USA
| | - P Nghiem
- Division of Dermatology, Department of Medicine, University of Washington, Seattle, WA, USA.,Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - S Bhatia
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA.,Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
27
|
Loos JA, Negro P, Cumino AC. In vitro anti-echinococcal activity of octreotide: Additive effect of metformin linked to autophagy. Acta Trop 2020; 203:105312. [PMID: 31870710 DOI: 10.1016/j.actatropica.2019.105312] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 12/07/2019] [Accepted: 12/12/2019] [Indexed: 01/28/2023]
Abstract
Cystic echinococcosis (CE) is a worldwide zoonosis caused by the Echinococcus granulosus larval stage. The currently available therapy for this disease is based on benzimidazoles, which are rarely curative and cause several adverse effects. Therefore, new treatment options are needed. Octreotide (Oct) is a somatostatin analogue which exhibits anti-proliferative and anti-secretory effects over several cancer cell lines expressing somatostatin receptors. Here, we assessed the in vitro pharmacological effect of Oct against the E. granulosus larval stage. The drug caused a significant dose-dependent decrease in the viability of both protoscoleces and metacestodes. SEM and TEM analysis showed ultrastructural damage in both larval forms under drug treatment. Based on this, we investigated the possible presence of an Oct binding receptor in the parasite. The putative somatostatin/allatostatin-like receptor (Eg-s/ast) conserves the characteristic topology and signature sequences of the prototype somatostatin receptor common to vertebrates and is expressed in both metacestodes and protoscoleces. Moreover, Oct treated-parasites showed the presence of autophagic structures and a significant increase in transcriptional expression of autophagy key genes such as Eg-atg6, Eg-atg8, Eg-atg12 and Eg-atg16. In addition, by in toto immunolocalization assays, an increase in the punctate pattern and Eg-Atg8 protein expression was detected in Oct-treated metacestodes. Subsequently, the combination of Oct and Met had an additive effect on the viability of both larval forms. Our results provide additional evidence for the participation of PI3K/AKT/TOR/autophagy pathway in the Echinococcus survival and suggest the concomitant use of these drugs as potential therapeutic agents in treating of CE.
Collapse
|
28
|
Kim J, Oh JH, Harlem H, Culler MD, Ku CR, Lee EJ. Therapeutic Effect of a Novel Chimeric Molecule Targeting Both Somatostatin and Dopamine Receptors on Growth Hormone-Secreting Pituitary Adenomas. Endocrinol Metab (Seoul) 2020; 35:177-187. [PMID: 32207278 PMCID: PMC7090307 DOI: 10.3803/enm.2020.35.1.177] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 01/12/2020] [Accepted: 01/28/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Acromegaly is a rare disease primarily caused by growth hormone (GH)-secreting pituitary adenomas, and its treatment is costly. Moreover, some patients are unresponsive to treatment. Hence, there are increasing efforts to develop new drugs with improved effectiveness for this disease. BIM23B065 is a novel chimeric molecule that acts on both somatostatin and dopamine receptors. This study aimed to investigate the effects of BIM23B065 compared with those of a somatostatin receptor analog and a dopamine agonist. METHODS The effects of BIM23B065 on the proliferation, GH and insulin-like growth factor-1 (IGF-1) levels, and extracellular signal-regulated kinase (ERK) 1/2 and cyclic AMP response element binding (CREB) phosphorylation of GH3 cells were investigated with MTS assay, enzyme-linked immunosorbent assay, and Western blotting, respectively. The dosage and treatment duration of BIM23B065 were tested in animal models of GH-secreting pituitary adenoma. The effect of BIM23B065 (3 mg/kg/day) on changes in IGF-1 levels before and after treatment was further investigated. RESULTS In vitro, BIM23B065 treatment decreased GH release in the culture media and downregulated ERK 1/2 and CREB phosphorylation to 22% and 26%, respectively. In vivo, IGF-1 expression decreased to 50 % after 4 weeks of treatment with BIM23B065 using an osmotic pump implant. Moreover, magnetic resonance imaging results showed that the tumor size decreased significantly following treatment with BIM23B065 for 4 weeks. CONCLUSION The novel chimeric molecule was effective in decreasing IGF-1 and GH levels and may serve as an effective therapeutic agent for acromegaly.
Collapse
Affiliation(s)
- Jean Kim
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Korea
- Endocrinology, Institute of Endocrine Research, Yonsei University College of Medicine, Seoul, Korea
| | - Ju Hun Oh
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Korea
- Endocrinology, Institute of Endocrine Research, Yonsei University College of Medicine, Seoul, Korea
| | - Heather Harlem
- Endocrinology Research, Ipsen Bioscience Inc., Cambridge, MA, USA
| | - Michael D Culler
- Endocrinology Research, Ipsen Bioscience Inc., Cambridge, MA, USA
| | - Cheol Ryong Ku
- Endocrinology, Institute of Endocrine Research, Yonsei University College of Medicine, Seoul, Korea.
| | - Eun Jig Lee
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Korea
- Endocrinology, Institute of Endocrine Research, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
29
|
Howe JR, Merchant NB, Conrad C, Keutgen XM, Hallet J, Drebin JA, Minter RM, Lairmore TC, Tseng JF, Zeh HJ, Libutti SK, Singh G, Lee JE, Hope TA, Kim MK, Menda Y, Halfdanarson TR, Chan JA, Pommier RF. The North American Neuroendocrine Tumor Society Consensus Paper on the Surgical Management of Pancreatic Neuroendocrine Tumors. Pancreas 2020; 49:1-33. [PMID: 31856076 PMCID: PMC7029300 DOI: 10.1097/mpa.0000000000001454] [Citation(s) in RCA: 220] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
This manuscript is the result of the North American Neuroendocrine Tumor Society consensus conference on the surgical management of pancreatic neuroendocrine tumors from July 19 to 20, 2018. The group reviewed a series of questions of specific interest to surgeons taking care of patients with pancreatic neuroendocrine tumors, and for each, the available literature was reviewed. What follows are these reviews for each question followed by recommendations of the panel.
Collapse
Affiliation(s)
- James R. Howe
- Department of Surgery, University of Iowa Carver College of Medicine, Iowa City, IA
| | | | - Claudius Conrad
- Department of Surgery, St. Elizabeth Medical Center, Tufts University School of Medicine, Boston, MA
| | | | - Julie Hallet
- Department of Surgery, University of Toronto, Sunnybrook Health Sciences Centre, Toronto, Canada
| | - Jeffrey A. Drebin
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Rebecca M. Minter
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | | | | | - Herbert J. Zeh
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX
| | - Steven K. Libutti
- §§ Department of Surgery, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ
| | - Gagandeep Singh
- Department of Surgery, City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Jeffrey E. Lee
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Thomas A. Hope
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA
| | - Michelle K. Kim
- Department of Medicine, Mt. Sinai Medical Center, New York, NY
| | - Yusuf Menda
- Department of Radiology, University of Iowa Carver College of Medicine, Iowa City, IA
| | | | - Jennifer A. Chan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Rodney F. Pommier
- Department of Surgery, Oregon Health & Sciences University, Portland, OR
| |
Collapse
|
30
|
Reynaert H, Colle I. Treatment of Advanced Hepatocellular Carcinoma with Somatostatin Analogues: A Review of the Literature. Int J Mol Sci 2019; 20:ijms20194811. [PMID: 31569719 PMCID: PMC6801667 DOI: 10.3390/ijms20194811] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 09/20/2019] [Accepted: 09/25/2019] [Indexed: 12/11/2022] Open
Abstract
Hepatocellular carcinoma, one of the most dreaded complications of cirrhosis, is a frequent cancer with high mortality. Early primary liver cancer can be treated by surgery or ablation techniques, but advanced hepatocellular carcinoma remains a challenge for clinicians. Most of these patients have underlying cirrhosis, which complicates or even precludes treatment. Therefore, efficacious treatments without major side effects are welcomed. Initial results of treatment of advanced hepatocellular carcinoma with somatostatin analogues were promising, but subsequent trials have resulted in conflicting outcomes. This might be explained by different patient populations, differences in dosage and type of treatment and differences in somatostatin receptor expression in the tumor or surrounding tissue. It has been shown that the expression of somatostatin receptors in the tumor might be of importance to select patients who could benefit from treatment with somatostatin analogues. Moreover, somatostatin receptor expression in hepatocellular carcinoma has been shown to correlate with recurrence, prognosis, and survival. In this review, we will summarize the available data on treatment of primary liver cancer with somatostatin analogues and analyze the current knowledge of somatostatin receptor expression in hepatocellular carcinoma and its possible clinical impact.
Collapse
Affiliation(s)
- Hendrik Reynaert
- Department of Gastroenterology-hepatology UZBrussel, Laarbeeklaan 101, 1090 Brussels, Belgium.
- Liver Cell Biology Lab, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium.
| | - Isabelle Colle
- Department of Gastroenterology-hepatology, ASZ Aalst, Merestraat 80, 9300 Aalst, Belgium.
- Department of Gastroenterology-hepatology, Ghent University, De Pintelaan, 9000 Ghent, Belgium.
| |
Collapse
|
31
|
Peptides derived from the extracellular domain of the somatostatin receptor splicing variant SST5TMD4 increase malignancy in multiple cancer cell types. Transl Res 2019; 211:147-160. [PMID: 30904441 DOI: 10.1016/j.trsl.2019.02.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 02/06/2019] [Accepted: 02/26/2019] [Indexed: 12/19/2022]
Abstract
Extracellular fragments derived from plasma membrane receptors can play relevant roles in the development/progression of tumor pathologies, thereby offering novel diagnostic or therapeutic opportunities. The truncated variant of somatostatin receptor subtype-5, SST5TMD4, is an aberrantly spliced receptor with 4 transmembrane domains, highly overexpressed in several tumor types, whose C-terminal tail is exposed towards the extracellular matrix, and could therefore be the substrate for proteolytic enzymes. In silico analysis implemented herein predicted 2 possible cleavage sites for metalloproteases MMP2, 9, 14, and 16 in its sequence, which could generate 3 releasable peptides. Of note, expression of those MMPs was directly correlated with SST5TMD4 in several cancer-derived cell lines (ie neuroendocrine tumors and prostate, breast, and liver cancers). Moreover, incubation with SST5TMD4-derived peptides enhanced malignancy features in all cancer cell types tested (ie proliferation, migration, etc.) and blunted the antiproliferative response to somatostatin in QGP-1 cells, acting probably through PI3K/AKT and/or MEK/ERK signaling pathways and the modulation of key cancer-associated genes (eg MMPs, MKI67, ACTR2/3, CD24/44). These results suggest that SST5TMD4-derived peptides could contribute to the strong oncogenic role of SST5TMD4 observed in multiple tumor pathologies, and, therefore, represent potential candidates to identify novel diagnostic, prognostic, or therapeutic targets in cancer.
Collapse
|
32
|
Fili S, Valmas A, Spiliopoulou M, Kontou P, Fitch A, Beckers D, Degen T, Barlos K, Barlos KK, Karavassili F, Margiolaki I. Revisiting the structure of a synthetic somatostatin analogue for peptide drug design. ACTA CRYSTALLOGRAPHICA SECTION B-STRUCTURAL SCIENCE CRYSTAL ENGINEERING AND MATERIALS 2019; 75:611-620. [DOI: 10.1107/s2052520619006012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 04/30/2019] [Indexed: 11/10/2022]
Abstract
Natural or artificially manufactured peptides attract scientific interest worldwide owing to their wide array of pharmaceutical and biological activities. X-ray structural studies are used to provide a precise extraction of information, which can be used to enable a better understanding of the function and physicochemical characteristics of peptides. Although it is vulnerable to disassociation, one of the most vital human peptide hormones, somatostatin, plays a regulatory role in the endocrine system as well as in the release of numerous secondary hormones. This study reports the successful crystallization and complete structural model of octreotide, a stable octapeptide analogue of somatostatin. Common obstacles in crystallographic studies arising from the intrinsic difficulties of obtaining a suitable single-crystal specimen were efficiently overcome as polycrystalline material was employed for synchrotron and laboratory X-ray powder diffraction (XPD) measurements. Data collection and preliminary analysis led to the identification of unit-cell symmetry [orthorhombic, P212121, a = 18.5453 (15), b = 30.1766 (25), c = 39.798 (4) Å], a process which was later followed by complete structure characterization and refinement, underlying the efficacy of the suggested (XPD) approach.
Collapse
|
33
|
Muhammad A, Coopmans EC, Gatto F, Franck SE, Janssen JAMJL, van der Lely AJ, Hofland LJ, Neggers SJCMM. Pasireotide Responsiveness in Acromegaly Is Mainly Driven by Somatostatin Receptor Subtype 2 Expression. J Clin Endocrinol Metab 2019; 104:915-924. [PMID: 30346538 DOI: 10.1210/jc.2018-01524] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 10/16/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND The response to first-generation somatostatin receptor ligands (SRLs) treatment in acromegaly correlates with expression of somatostatin receptor subtype 2 (SSTR2). However, pasireotide shows the highest binding affinity for SSTR subtype 5 (SSTR5). It has been suggested that in acromegaly, SSTR5 expression is better at predicting the response to pasireotide long-acting release (PAS-LAR) treatment than SSTR2 expression. AIM To investigate in patients with active acromegaly whether response to SRL treatment correlates to PAS-LAR treatment and to what extent SSTR2 and SSTR5 expression are correlated to the response to PAS-LAR treatment. METHODS We included 52 patients from a cohort that initially received SRL treatment, followed by SRL and pegvisomant combination treatment, and finally PAS-LAR treatment. The long-term response to PAS-LAR was evaluated using a PAS-LAR score. In 14 out of 52 patients, somatotroph adenoma tissue samples were available to evaluate SSTR2 and SSTR5 expression using a previously validated immunoreactivity score (IRS). RESULTS The percentage IGF-I (times the upper limit of normal) reduction, which was observed after SRL treatment, correlated with PAS-LAR response score during follow-up (r = 0.40; P = 0.003; n = 52). After exclusion of SRL-pretreated patients, SSTR2 IRS was positively correlated to PAS-LAR score (r = 0.58; P = 0.039; n = 9), whereas SSTR5 IRS showed no relation (r = 0.35; P = 0.36; n = 9). CONCLUSIONS In a cohort of patients partially responsive to SRLs, the IGF-I-lowering effects of PAS-LAR treatment correlated with the effect of SRL treatment and seemed to be mainly driven by SSTR2 expression instead of SSTR5.
Collapse
Affiliation(s)
- Ammar Muhammad
- Department of Medicine, Endocrinology Section, Pituitary Center Rotterdam, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Eva C Coopmans
- Department of Medicine, Endocrinology Section, Pituitary Center Rotterdam, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Federico Gatto
- Endocrinology Unit, Department of Internal Medicine and Medical Specialties, Ospedale Policlinico San Martino, Genova, Italy
| | - Sanne E Franck
- Department of Medicine, Endocrinology Section, Pituitary Center Rotterdam, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Joseph A M J L Janssen
- Department of Medicine, Endocrinology Section, Pituitary Center Rotterdam, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Aart Jan van der Lely
- Department of Medicine, Endocrinology Section, Pituitary Center Rotterdam, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Leo J Hofland
- Department of Medicine, Endocrinology Section, Pituitary Center Rotterdam, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Sebastian J C M M Neggers
- Department of Medicine, Endocrinology Section, Pituitary Center Rotterdam, Erasmus University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
34
|
Paramonov VM, Desai D, Kettiger H, Mamaeva V, Rosenholm JM, Sahlgren C, Rivero-Müller A. Targeting Somatostatin Receptors By Functionalized Mesoporous Silica Nanoparticles - Are We Striking Home? Nanotheranostics 2018; 2:320-346. [PMID: 30148051 PMCID: PMC6107779 DOI: 10.7150/ntno.23826] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 06/25/2018] [Indexed: 12/02/2022] Open
Abstract
The concept of delivering nanoformulations to desired tissues by means of targeting membrane receptors of high local abundance by ligands anchored to the nanocarrier has gained a lot of attention over the last decade. Currently, there is no unanimous opinion on whether surface functionalization of nanocarriers by targeting ligands translates into any real benefit in terms of pharmacokinetics or treatment outcomes. Having examined the published nanocarriers designed to engage with somatostatin receptors, we realized that in the majority of cases targetability claims were not supported by solid evidence of targeting ligand-targeted receptor coupling, which is the very crux of a targetability concept. Here, we present an approach to characterize targetability of mesoporous silica-based nanocarriers functionalized with ligands of somatostatin receptors. The targetability proof in our case comes from a functional assay based on a genetically-encoded cAMP probe, which allows for real-time capture of receptor activation in living cells, triggered by targeting ligands on nanoparticles. We elaborate on the development and validation of the assay, highlighting the power of proper functional tests in the characterization pipeline of targeted nanoformulations.
Collapse
Affiliation(s)
- Valeriy M Paramonov
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Finland.,Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Finland.,Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Finland
| | - Diti Desai
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Finland
| | - Helene Kettiger
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Finland
| | - Veronika Mamaeva
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Finland.,Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Finland
| | - Jessica M Rosenholm
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Finland
| | - Cecilia Sahlgren
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Finland.,Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Finland.,Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Adolfo Rivero-Müller
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Finland.,Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Finland.,Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Finland.,Department of Biochemistry and Molecular Biology, Medical University of Lublin, Poland
| |
Collapse
|
35
|
Rodriguez M, Frost JA, Schonbrunn A. Real-Time Signaling Assays Demonstrate Somatostatin Agonist Bias for Ion Channel Regulation in Somatotroph Tumor Cells. J Endocr Soc 2018; 2:779-793. [PMID: 30151433 PMCID: PMC6106105 DOI: 10.1210/js.2018-00115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 06/11/2018] [Indexed: 11/19/2022] Open
Abstract
Acromegaly is a neuroendocrine disorder caused by excess secretion of GH by somatotroph tumor cells. It is often treated with somatostatin receptor (SSTR) 2 agonists, which suppress GH secretion. SOM230 is a somatostatin analogue that targets multiple SSTRs and was recently approved for patients with treatment-resistant acromegaly. Previous reports indicate that SOM230 may function as a biased agonist, suggesting that its ability to selectively activate SSTR-dependent signaling events may contribute to its therapeutic efficacy. To better understand how SOM230 modulates Sstr2A function, which is the most commonly expressed SSTR in somatotrophs, we used real-time assays to study SOM230-dependent signaling in rat pituitary tumor cells. We observed that SOM230 suppressed cAMP production in a Gαi-dependent manner, similar to conventional Sstr2A agonists. However, it did not cause receptor internalization as would be expected for an Sstr2A agonist. Surprisingly, SOM230 did not cause membrane hyperpolarization, which is an important mechanism by which Sstr2a activation suppresses intracellular calcium (Ca2+) accumulation and GH secretion. In fact, SOM230 inhibited the ability of conventional somatostatin analogues to control membrane potential. However, SOM230 still inhibited intracellular Ca2+ accumulation in a novel, Gβγ-dependent manner. These studies show that SOM230 exhibits strong agonist bias in regulating signaling pathways downstream of Sstr2A that control GH secretion.
Collapse
Affiliation(s)
- Melissa Rodriguez
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas
| | - Jeffrey A Frost
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas
| | - Agnes Schonbrunn
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas
| |
Collapse
|
36
|
Importance of Immunohistochemical Detection of Somatostatin Receptors. Pathol Oncol Res 2018; 25:521-525. [DOI: 10.1007/s12253-018-0426-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 05/28/2018] [Indexed: 01/10/2023]
|
37
|
Pedraza-Arévalo S, Hormaechea-Agulla D, Gómez-Gómez E, Requena MJ, Selth LA, Gahete MD, Castaño JP, Luque RM. Somatostatin receptor subtype 1 as a potential diagnostic marker and therapeutic target in prostate cancer. Prostate 2017; 77:1499-1511. [PMID: 28905400 DOI: 10.1002/pros.23426] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 08/23/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND Prostate cancer (PCa) is a highly prevalent neoplasia that is strongly influenced by the endocrine system. Somatostatin (SST) and its five receptors (sst1-5 encoded by SSTR1-5 genes) comprise a pleiotropic system present in most endocrine-related cancers, some of which are successfully treated with SST analogs. Interestingly, it has been reported that SSTR1 is overexpressed in PCa, but its regulation, functional role, and clinical implications are still poorly known. METHODS PCa specimens (n = 52) from biopsies and control prostates from cystoprostatectomies (n = 12), as well as in silico databases were used to evaluate SSTR1 and miRNAs expression. In vitro studies in 22Rv1 PCa cells were implemented to explore the regulation of SSTR1/sst1 by different miRNAs, and to evaluate the consequences of SSTR1/sst1 overexpression, silencing and/or activation [with the specific BIM-23926 sst1 agonist (IPSEN)] on cell-proliferation, migration, signaling-pathways, and androgen-signaling. RESULTS We found that SSTR1 is overexpressed in multiple cohorts of PCa samples, as compared with normal prostate tissues, wherein it correlates with androgen receptor (AR) expression, and appears to be associated with aggressiveness (metastasis). Furthermore, our data revealed that SSTR1/sst1 expression might be regulated by specific miRNAs in PCa, including miR-24, which is downregulated in PCa samples and correlates inversely with SSTR1 expression. In vitro studies indicated that treatment with the BIM-23926 sst1 agonist, as well as SSTR1 overexpression, decreased, whereas SSTR1 silencing increased, cell-proliferation in 22Rv1 cells, likely through the regulation of PI3K/AKT-CCND3 signaling-pathway. Importantly, sst1 action was also able to modulate androgen/AR activity, and reduced PSA secretion from PCa cell lines. CONCLUSIONS Altogether, our results indicate that SSTR1 is overexpressed in PCa, where it can exert a relevant pathophysiological role by decreasing cell-proliferation and PSA secretion. Therefore, sst1, possibly in combination with miR-24, could be used as a novel tool to explore therapeutic targets in PCa.
Collapse
MESH Headings
- Aged
- Aged, 80 and over
- Biomarkers, Tumor/biosynthesis
- Biomarkers, Tumor/genetics
- Cell Line, Tumor
- Humans
- Male
- Middle Aged
- Molecular Targeted Therapy
- Prostatic Neoplasms, Castration-Resistant/diagnosis
- Prostatic Neoplasms, Castration-Resistant/genetics
- Prostatic Neoplasms, Castration-Resistant/metabolism
- Prostatic Neoplasms, Castration-Resistant/therapy
- Receptors, Somatostatin/biosynthesis
- Receptors, Somatostatin/genetics
Collapse
Affiliation(s)
- Sergio Pedraza-Arévalo
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, Universidad de Córdoba, Córdoba, Spain
- Reina Sofia University Hospital, Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain
| | - Daniel Hormaechea-Agulla
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, Universidad de Córdoba, Córdoba, Spain
- Reina Sofia University Hospital, Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain
| | - Enrique Gómez-Gómez
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, Universidad de Córdoba, Córdoba, Spain
- Reina Sofia University Hospital, Córdoba, Spain
- Urology Service, Reina Sofia University Hospital, Córdoba, Spain
| | - María J Requena
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, Universidad de Córdoba, Córdoba, Spain
- Reina Sofia University Hospital, Córdoba, Spain
- Urology Service, Reina Sofia University Hospital, Córdoba, Spain
| | - Luke A Selth
- Dame Roma Mitchell Cancer Research Laboratories and Freemasons Foundation Centre for Men's Health, Adelaide Medical School, The University of Adelaide, SA, 5005, Australia
| | - Manuel D Gahete
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, Universidad de Córdoba, Córdoba, Spain
- Reina Sofia University Hospital, Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain
| | - Justo P Castaño
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, Universidad de Córdoba, Córdoba, Spain
- Reina Sofia University Hospital, Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain
| | - Raul M Luque
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, Universidad de Córdoba, Córdoba, Spain
- Reina Sofia University Hospital, Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain
| |
Collapse
|
38
|
Ugrinova I, Petrova M, Chalabi-Dchar M, Bouvet P. Multifaceted Nucleolin Protein and Its Molecular Partners in Oncogenesis. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2017; 111:133-164. [PMID: 29459030 DOI: 10.1016/bs.apcsb.2017.08.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Discovered in 1973, nucleolin is one of the most abundant phosphoproteins of the nucleolus. The ability of nucleolin to be involved in many cellular processes is probably related to its structural organization and its capability to form many different interactions with other proteins. Many functions of nucleolin affect cellular processes involved in oncogenesis-for instance: in ribosome biogenesis; in DNA repair, remodeling, and genome stability; in cell division and cell survival; in chemokine and growth factor signaling pathways; in angiogenesis and lymphangiogenesis; in epithelial-mesenchymal transition; and in stemness. In this review, we will describe the different functions of nucleolin in oncogenesis through its interaction with other proteins.
Collapse
Affiliation(s)
- Iva Ugrinova
- "Roumen Tsanev" Institute of Molecular Biology, Bulgarian Academy of Sciences, Sofia, Bulgaria.
| | - Maria Petrova
- "Roumen Tsanev" Institute of Molecular Biology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Mounira Chalabi-Dchar
- Université de Lyon, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Lyon, France
| | - Philippe Bouvet
- Université de Lyon, Ecole Normale Supérieure de Lyon, Lyon, France
| |
Collapse
|
39
|
Progress in the formulation and delivery of somatostatin analogs for acromegaly. Ther Deliv 2017; 8:867-878. [DOI: 10.4155/tde-2017-0064] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
A 14 amino acid cystin bridge containing neuropeptide was discovered in 1973 and designated as growth hormone-inhibiting hormone, in other words, somatostatin. Its discovery led to the synthesis of three analogs which were licensed for the treatment of acromegaly: octreotide, lanreotide and pasireotide. Somatostatin analogs are currently approved only as either subcutaneous or intramuscular long-acting injections. We examine the challenges that must be overcome to create oral formulations of somatostatin analogs and examine selected clinical trial data. While octreotide has low intestinal permeability, similar to almost all other peptides, it has an advantage of being more stable against intestinal peptidases. The development of new oral formulation strategies may eventually allow for the successful oral administration of potent somatostatin analogs with high therapeutic indices.
Collapse
|
40
|
Graillon T, Romano D, Defilles C, Lisbonis C, Saveanu A, Figarella-Branger D, Roche PH, Fuentes S, Chinot O, Dufour H, Barlier A. Pasireotide is more effective than octreotide, alone or combined with everolimus on human meningioma in vitro. Oncotarget 2017; 8:55361-55373. [PMID: 28903425 PMCID: PMC5589664 DOI: 10.18632/oncotarget.19517] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 07/12/2017] [Indexed: 12/18/2022] Open
Abstract
Pasireotide is a somatostatin analog (SSA) that targets somatostatin receptor subtype 1 (SST1), SST2, SST3, and SST5 with a high affinity. Pasireotide has a better antisecretory effect in acromegaly, Cushing's disease, and neuroendocrine tumors than octreotide. In this study, we compared the effects of pasireotide to those of octreotide in vitro on meningioma primary cell cultures, both alone and in combination with the mTOR inhibitor everolimus. Significant mRNA expression levels of SST1, SST2, and SST5 were observed in 40.5%, 100%, and 35% of meningioma samples, respectively. Pasireotide had a significantly stronger inhibitory effect on cell proliferation than octreotide. The effect of pasireotide, but not of octreotide, was significantly stronger in the group expressing the highest level of SST1 mRNA. Combined treatment with pasireotide and everolimus induced a higher reduction in cell viability than that with octreotide plus everolimus. Moreover, pasireotide decreased Akt phosphorylation and reversed everolimus-induced Akt hyperphosphorylation to a higher degree than octreotide. Using 4E-BP1 siRNA (si4E-BP), we demonstrated that 4E-BP1 protein silencing significantly reversed the response to everolimus, both alone and in combination with SSAs. Moreover, si4E-BP completely reversed the inhibition of cyclin D1 expression level and the increase in p27kip1 induced by SSAs, both alone and in combination with everolimus. Our results strongly support the need for further studies on the combination of pasireotide and everolimus in medical therapy for meningiomas.
Collapse
Affiliation(s)
- Thomas Graillon
- Aix Marseille Univ, CNRS CRN2M UMR7286, Marseille, France.,APHM, La Timone Hospital, Department of Neurosurgery, Marseille, France
| | - David Romano
- Aix Marseille Univ, CNRS CRN2M UMR7286, Marseille, France
| | | | | | - Alexandru Saveanu
- Aix Marseille Univ, CNRS CRN2M UMR7286, Marseille, France.,APHM, La Conception Hospital, Molecular Biology Laboratory, Marseille, France
| | - Dominique Figarella-Branger
- APHM, La Timone Hospital, Department of Anatomopathology and Neuropathology, Marseille, France.,Aix Marseille Univ, INSERM, CRO2 UMR911, Marseille, France
| | | | - Stéphane Fuentes
- APHM, La Timone Hospital, Department of Neurosurgery, Marseille, France
| | - Olivier Chinot
- Aix Marseille Univ, INSERM, CRO2 UMR911, Marseille, France.,APHM, La Timone Hospital, Department of Neuro-oncology, Marseille, France
| | - Henry Dufour
- Aix Marseille Univ, CNRS CRN2M UMR7286, Marseille, France.,APHM, La Timone Hospital, Department of Neurosurgery, Marseille, France
| | - Anne Barlier
- Aix Marseille Univ, CNRS CRN2M UMR7286, Marseille, France.,APHM, La Conception Hospital, Molecular Biology Laboratory, Marseille, France
| |
Collapse
|
41
|
Mende KC, Matschke J, Burkhardt T, Saeger W, Buslei R, Buchfelder M, Fahlbusch R, Westphal M, Flitsch J. Pituicytoma-An outlook on possible targeted therapies. CNS Neurosci Ther 2017; 23:620-626. [PMID: 28556544 DOI: 10.1111/cns.12709] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 04/30/2017] [Accepted: 05/03/2017] [Indexed: 12/01/2022] Open
Abstract
INTRODUCTION Pituicytoma is a rare neoplasm of the sella region. Tumor resection is the primary treatment option, but remains subtotal due to excessive bleeding in many cases. The search for alternative or additional treatment regimens is necessary. AIMS We aimed to determine the receptor expression of pituicytoma to find alternatives or supplements to surgical therapy in the use of targeted therapies. METHODS Pituicytoma samples were collected from three institutions between 2006 and 2015 and were stained for vascular endothelial growth factors (VEGF), thyroid transcription factor (TTF1), and somatostatin receptors (SSTR 2/3/5). The stains were classified from 0=no staining to +++=strong staining. A complementary retrospective analysis of the patient charts regarding sex, age, and primary symptoms, pituitary function, and perioperative complications was performed. RESULTS Ten samples were analyzed; mean patient age was 57.8 years SD 16.3 years. Seven samples were acquired from male patients (one relapse) and three from female. All tumors stained strongly positive (+++) for VEGF-R. Six samples stained positive for TTF1. As for somatostatin receptors, three samples were slightly positive for SSTR 2; seven were negative. SSTR 3 was + in one, three were ++, three were +++, and three were 0. SSTR 5 stained +++ in 1, ++ in 5, + in 1, and 0 in three patients. CONCLUSIONS Pituicytomas were generally positive for VEGFR and showed regular expression of SSTR 3 and 5 indicating a possible treatment option through targeted therapies in cases where resection remains insufficient. Further research is necessary as to whether tumor growth can be inhibited using these pathways.
Collapse
Affiliation(s)
| | - Jakob Matschke
- Institute of Neuropathology, Hamburg University Medical Center, Hamburg, Germany
| | - Till Burkhardt
- Department of Neurosurgery, Hamburg University Medical Center, Hamburg, Germany
| | - Wolfgang Saeger
- Institute of Neuropathology, Hamburg University Medical Center, Hamburg, Germany
| | - Rolf Buslei
- Department of Neuropathology, University of Erlangen, Erlangen, Germany
| | | | - Rudolf Fahlbusch
- International Neuroscience Institute Hannover GmbH, Hannover, Germany
| | - Manfred Westphal
- Department of Neurosurgery, Hamburg University Medical Center, Hamburg, Germany
| | - Jörg Flitsch
- Department of Neurosurgery, Hamburg University Medical Center, Hamburg, Germany
| |
Collapse
|
42
|
Lequoy M, Desbois-Mouthon C, Wendum D, Gupta V, Blachon JL, Scatton O, Dumont S, Bonnemaire M, Schmidlin F, Rosmorduc O, Fartoux L. Somatostatin receptors in resected hepatocellular carcinoma: status and correlation with markers of poor prognosis. Histopathology 2016; 70:492-498. [DOI: 10.1111/his.13034] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 07/06/2016] [Indexed: 12/16/2022]
Affiliation(s)
- Marie Lequoy
- Department of Hepatology; AP-HP; Saint-Antoine Hospital; Paris France
- Saint-Antoine Research Centre; Sorbonne Universités; UPMC Univ. Paris 06, UMR_S 938; Paris France
| | | | - Dominique Wendum
- Saint-Antoine Research Centre; Sorbonne Universités; UPMC Univ. Paris 06, UMR_S 938; Paris France
- Department of Pathology; AP-HP; Saint-Antoine Hospital; Paris France
| | - Vandana Gupta
- Oncology and Biomarkers; Ipsen Biosci. Inc.; Cambridge MA USA
| | | | - Olivier Scatton
- Saint-Antoine Research Centre; Sorbonne Universités; UPMC Univ. Paris 06, UMR_S 938; Paris France
- Department of Hepatobiliary Surgery; AP-HP; Pitié Salpêtrière Hospital; Paris France
| | - Sylvie Dumont
- Saint-Antoine Research Centre; Sorbonne Universités; UPMC Univ. Paris 06, UMR_S 938; Paris France
- Department of Pathology; AP-HP; Saint-Antoine Hospital; Paris France
| | | | | | - Olivier Rosmorduc
- Saint-Antoine Research Centre; Sorbonne Universités; UPMC Univ. Paris 06, UMR_S 938; Paris France
- Department of Hepatology; AP-HP; Pitié Salpêtrière Hospital; Paris France
| | - Laetitia Fartoux
- Saint-Antoine Research Centre; Sorbonne Universités; UPMC Univ. Paris 06, UMR_S 938; Paris France
- Department of Hepatology; AP-HP; Pitié Salpêtrière Hospital; Paris France
| |
Collapse
|
43
|
Ibáñez-Costa A, Rivero-Cortés E, Vázquez-Borrego MC, Gahete MD, Jiménez-Reina L, Venegas-Moreno E, de la Riva A, Arráez MÁ, González-Molero I, Schmid HA, Maraver-Selfa S, Gavilán-Villarejo I, García-Arnés JA, Japón MA, Soto-Moreno A, Gálvez MA, Luque RM, Castaño JP. Octreotide and pasireotide (dis)similarly inhibit pituitary tumor cells in vitro. J Endocrinol 2016; 231:135-145. [PMID: 27587848 DOI: 10.1530/joe-16-0332] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 09/01/2016] [Indexed: 02/01/2023]
Abstract
Somatostatin analogs (SSA) are the mainstay of pharmacological treatment for pituitary adenomas. However, some patients escape from therapy with octreotide, a somatostatin receptor 2 (sst2)-preferring SSA, and pasireotide, a novel multi-sst-preferring SSA, may help to overcome this problem. It has been proposed that correspondence between sst1-sst5 expression pattern and SSA-binding profile could predict patient's response. To explore the cellular/molecular features associated with octreotide/pasireotide response, we performed a parallel comparison of their in vitro effects, evaluating sst1-sst5 expression, intracellular Ca2+ signaling ([Ca2+]i), hormone secretion and cell viability, in a series of 85 pituitary samples. Somatotropinomas expressed sst5>sst2, yet octreotide reduced [Ca2+]i more efficiently than pasireotide, while both SSA similarly decreased growth hormone release/expression and viability. Corticotropinomas predominantly expressed sst5, but displayed limited response to pasireotide, while octreotide reduced functional endpoints. Non-functioning adenomas preferentially expressed sst3 but, surprisingly, both SSA increased cell viability. Prolactinomas mainly expressed sst1 but were virtually unresponsive to SSA. Finally, both SSA decreased [Ca2+]i in normal pituitaries. In conclusion, both SSA act in vitro on pituitary adenomas exerting both similar and distinct effects; however, no evident correspondence was found with the sst1-sst5 profile. Thus, it seems plausible that additional factors, besides the simple abundance of a given sst, critically influence the SSA response.
Collapse
Affiliation(s)
- Alejandro Ibáñez-Costa
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC)Córdoba, Spain
- Department of Cell BiologyPhysiology and Immunology, Universidad de Córdoba, Córdoba, Spain
- Hospital Universitario Reina SofíaCórdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn)Córdoba, Spain
| | - Esther Rivero-Cortés
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC)Córdoba, Spain
- Department of Cell BiologyPhysiology and Immunology, Universidad de Córdoba, Córdoba, Spain
- Hospital Universitario Reina SofíaCórdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn)Córdoba, Spain
| | - Mari C Vázquez-Borrego
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC)Córdoba, Spain
- Department of Cell BiologyPhysiology and Immunology, Universidad de Córdoba, Córdoba, Spain
- Hospital Universitario Reina SofíaCórdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn)Córdoba, Spain
| | - Manuel D Gahete
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC)Córdoba, Spain
- Department of Cell BiologyPhysiology and Immunology, Universidad de Córdoba, Córdoba, Spain
- Hospital Universitario Reina SofíaCórdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn)Córdoba, Spain
| | - Luis Jiménez-Reina
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC)Córdoba, Spain
- Department of Morphological SciencesUniversidad de Córdoba, Córdoba, Spain
| | - Eva Venegas-Moreno
- Metabolism and Nutrition UnitHospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS), Seville, Spain
| | - Andrés de la Riva
- Service of NeurosurgeryHospital Universitario Reina Sofía, Córdoba, Spain
| | | | | | - Herbert A Schmid
- Novartis Pharma AGNovartis Institutes for Biomedical Research, Oncology, CH-4057 Basel, Switzerland
| | - Silvia Maraver-Selfa
- Service of Endocrinology and NutritionHospital Clínico Universitario Virgen de la Victoria, Málaga, Spain
| | | | | | - Miguel A Japón
- Department of PathologyHospital Universitario Virgen del Rocío, Seville, Spain
| | - Alfonso Soto-Moreno
- Metabolism and Nutrition UnitHospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS), Seville, Spain
| | - María A Gálvez
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC)Córdoba, Spain
- Service of Endocrinology and NutritionHospital Universitario Reina Sofía, Córdoba, Spain
| | - Raúl M Luque
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC)Córdoba, Spain
- Department of Cell BiologyPhysiology and Immunology, Universidad de Córdoba, Córdoba, Spain
- Hospital Universitario Reina SofíaCórdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn)Córdoba, Spain
| | - Justo P Castaño
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC)Córdoba, Spain
- Department of Cell BiologyPhysiology and Immunology, Universidad de Córdoba, Córdoba, Spain
- Hospital Universitario Reina SofíaCórdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn)Córdoba, Spain
| |
Collapse
|
44
|
Wang M, Shan F, Zou Y, Sun X, Zhang ZR, Fu Y, Gong T. Pharmacokinetic and pharmacodynamic study of a phospholipid-based phase separation gel for once a month administration of octreotide. J Control Release 2016; 230:45-56. [DOI: 10.1016/j.jconrel.2016.03.036] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2015] [Revised: 03/03/2016] [Accepted: 03/29/2016] [Indexed: 01/28/2023]
|
45
|
Duluc C, Moatassim-Billah S, Chalabi-Dchar M, Perraud A, Samain R, Breibach F, Gayral M, Cordelier P, Delisle MB, Bousquet-Dubouch MP, Tomasini R, Schmid H, Mathonnet M, Pyronnet S, Martineau Y, Bousquet C. Pharmacological targeting of the protein synthesis mTOR/4E-BP1 pathway in cancer-associated fibroblasts abrogates pancreatic tumour chemoresistance. EMBO Mol Med 2016; 7:735-53. [PMID: 25834145 PMCID: PMC4459815 DOI: 10.15252/emmm.201404346] [Citation(s) in RCA: 159] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is extremely stroma-rich. Cancer-associated fibroblasts (CAFs) secrete proteins that activate survival and promote chemoresistance of cancer cells. Our results demonstrate that CAF secretome-triggered chemoresistance is abolished upon inhibition of the protein synthesis mTOR/4E-BP1 regulatory pathway which we found highly activated in primary cultures of α-SMA-positive CAFs, isolated from human PDAC resections. CAFs selectively express the sst1 somatostatin receptor. The SOM230 analogue (Pasireotide) activates the sst1 receptor and inhibits the mTOR/4E-BP1 pathway and the resultant synthesis of secreted proteins including IL-6. Consequently, tumour growth and chemoresistance in nude mice xenografted with pancreatic cancer cells and CAFs, or with pieces of resected human PDACs, are reduced when chemotherapy (gemcitabine) is combined with SOM230 treatment. While gemcitabine alone has marginal effects, SOM230 is permissive to gemcitabine-induced cancer cell apoptosis and acts as an antifibrotic agent. We propose that selective inhibition of CAF protein synthesis with sst1-directed pharmacological compounds represents an anti-stromal-targeted therapy with promising chemosensitization potential.
Collapse
Affiliation(s)
- Camille Duluc
- INSERM UMR-1037, Cancer Research Center of Toulouse (CRCT), Equipe labellisée Ligue Contre le Cancer and Laboratoire d'Excellence Toulouse Cancer (TOUCAN), Université de Toulouse, Toulouse, France
| | - Siham Moatassim-Billah
- INSERM UMR-1037, Cancer Research Center of Toulouse (CRCT), Equipe labellisée Ligue Contre le Cancer and Laboratoire d'Excellence Toulouse Cancer (TOUCAN), Université de Toulouse, Toulouse, France Biochemistry-Immunology Laboratory, Faculty of Sciences Rabat, University Mohammed V - Agdal, Agdal, Morocco
| | - Mounira Chalabi-Dchar
- INSERM UMR-1037, Cancer Research Center of Toulouse (CRCT), Equipe labellisée Ligue Contre le Cancer and Laboratoire d'Excellence Toulouse Cancer (TOUCAN), Université de Toulouse, Toulouse, France
| | - Aurélie Perraud
- EA 3842 Laboratory, Medicine and Pharmacy Faculties, Limoges University, Limoges, France
| | - Rémi Samain
- INSERM UMR-1037, Cancer Research Center of Toulouse (CRCT), Equipe labellisée Ligue Contre le Cancer and Laboratoire d'Excellence Toulouse Cancer (TOUCAN), Université de Toulouse, Toulouse, France
| | | | - Marion Gayral
- INSERM UMR-1037, Cancer Research Center of Toulouse (CRCT), Equipe labellisée Ligue Contre le Cancer and Laboratoire d'Excellence Toulouse Cancer (TOUCAN), Université de Toulouse, Toulouse, France
| | - Pierre Cordelier
- INSERM UMR-1037, Cancer Research Center of Toulouse (CRCT), Equipe labellisée Ligue Contre le Cancer and Laboratoire d'Excellence Toulouse Cancer (TOUCAN), Université de Toulouse, Toulouse, France
| | | | - Marie-Pierre Bousquet-Dubouch
- CNRS UMR-5089, Institut de Pharmacologie et de Biologie structurale (IPBS), Université de Toulouse, Toulouse, France
| | - Richard Tomasini
- CRCM, INSERM, U1068; Paoli-Calmettes Institute; Aix-Marseille University, UM105; CNRS, UMR7258, Marseille, France
| | | | - Muriel Mathonnet
- EA 3842 Laboratory, Medicine and Pharmacy Faculties, Limoges University, Limoges, France
| | - Stéphane Pyronnet
- INSERM UMR-1037, Cancer Research Center of Toulouse (CRCT), Equipe labellisée Ligue Contre le Cancer and Laboratoire d'Excellence Toulouse Cancer (TOUCAN), Université de Toulouse, Toulouse, France
| | - Yvan Martineau
- INSERM UMR-1037, Cancer Research Center of Toulouse (CRCT), Equipe labellisée Ligue Contre le Cancer and Laboratoire d'Excellence Toulouse Cancer (TOUCAN), Université de Toulouse, Toulouse, France
| | - Corinne Bousquet
- INSERM UMR-1037, Cancer Research Center of Toulouse (CRCT), Equipe labellisée Ligue Contre le Cancer and Laboratoire d'Excellence Toulouse Cancer (TOUCAN), Université de Toulouse, Toulouse, France
| |
Collapse
|
46
|
Gradiz R, Silva HC, Carvalho L, Botelho MF, Mota-Pinto A. MIA PaCa-2 and PANC-1 - pancreas ductal adenocarcinoma cell lines with neuroendocrine differentiation and somatostatin receptors. Sci Rep 2016; 6:21648. [PMID: 26884312 PMCID: PMC4756684 DOI: 10.1038/srep21648] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 01/28/2016] [Indexed: 02/06/2023] Open
Abstract
Studies using cell lines should always characterize these cells to ensure that the results are not distorted by unexpected morphological or genetic changes possibly due to culture time or passage number. Thus, the aim of this study was to describe those MIA PaCa-2 and PANC-1 cell line phenotype and genotype characteristics that may play a crucial role in pancreatic cancer therapeutic assays, namely neuroendocrine chemotherapy and peptide receptor radionuclide therapy. Epithelial, mesenchymal, endocrine and stem cell marker characterization was performed by immunohistochemistry and flow cytometry, and genotyping by PCR, gene sequencing and capillary electrophoresis. MIA PaCa-2 (polymorphism) expresses CK5.6, AE1/AE3, E-cadherin, vimentin, chromogranin A, synaptophysin, SSTR2 and NTR1 but not CD56. PANC-1 (pleomorphism) expresses CK5.6, MNF-116, vimentin, chromogranin A, CD56 and SSTR2 but not E-cadherin, synaptophysin or NTR1. MIA PaCA-1 is CD24−, CD44+/++, CD326−/+ and CD133/1−, while PANC-1 is CD24−/+, CD44+, CD326−/+ and CD133/1−. Both cell lines have KRAS and TP53 mutations and homozygous deletions including the first 3 exons of CDKN2A/p16INK4A, but no SMAD4/DPC4 mutations or microsatellite instability. Both have neuroendocrine differentiation and SSTR2 receptors, precisely the features making them suitable for the therapies we propose to assay in future studies.
Collapse
Affiliation(s)
- Rui Gradiz
- General Pathology Laboratory, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,CIMAGO - Research Center for Environment, Genetics and Oncobiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Henriqueta C Silva
- Medical Genetics' Unit, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,CIMAGO - Research Center for Environment, Genetics and Oncobiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Lina Carvalho
- Anatomical and Molecular Pathology Department, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,CIMAGO - Research Center for Environment, Genetics and Oncobiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Maria Filomena Botelho
- Biophysics' Unit, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,CIMAGO - Research Center for Environment, Genetics and Oncobiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,CNC.IBILI, University of Coimbra, Portugal
| | - Anabela Mota-Pinto
- General Pathology Laboratory, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,CIMAGO - Research Center for Environment, Genetics and Oncobiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,CNC.IBILI, University of Coimbra, Portugal
| |
Collapse
|
47
|
Chauhan A, Ramirez RA. Diffuse Idiopathic Pulmonary Neuroendocrine Cell Hyperplasia (DIPNECH) and the Role of Somatostatin analogs: A Case Series. Lung 2015; 193:653-7. [DOI: 10.1007/s00408-015-9754-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 06/16/2015] [Indexed: 11/30/2022]
|
48
|
Therapeutic uses of somatostatin and its analogues: Current view and potential applications. Pharmacol Ther 2015; 152:98-110. [PMID: 25956467 DOI: 10.1016/j.pharmthera.2015.05.007] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 04/28/2015] [Indexed: 01/22/2023]
Abstract
Somatostatin is an endogeneous cyclic tetradecapeptide hormone that exerts multiple biological activities via five ubiquitously distributed receptor subtypes. Classified as a broad inhibitory neuropeptide, somatostatin has anti-secretory, anti-proliferative and anti-angiogenic effects. The clinical use of native somatostatin is limited by a very short half-life (1 to 3min) and the broad spectrum of biological responses. Thus stable, receptor-selective agonists have been developed. The majority of these somatostatin therapeutic agonists bind strongly to two of the five receptor subtypes, although recently an agonist of wider affinity has been introduced. Somatostatin agonists are established in the treatment of acromegaly with recently approved indications in the therapy of neuroendocrine tumours. Potential therapeutic uses for somatostatin analogues include diabetic complications like retinopathy, nephropathy and obesity, due to inhibition of IGF-1, VEGF together with insulin secretion and effects upon the renin-angiotensin-aldosterone system. Wider uses in anti-neoplastic therapy may also be considered and recent studies have further revealed anti-inflammatory and anti-nociceptive effects. This review provides a comprehensive, current view of the biological functions of somatostatin and potential therapeutic uses, informed by the wide range of pharmacological advances reported since the last published review in 2004 by P. Dasgupta. The pharmacology of somatostatin receptors is explained, the current uses of somatostatin agonists are discussed, and the potential future of therapeutic applications is explored.
Collapse
|
49
|
Nelson CA, Azure MT, Adams CT, Zinn KR. The somatostatin analog 188Re-P2045 inhibits the growth of AR42J pancreatic tumor xenografts. J Nucl Med 2014; 55:2020-5. [PMID: 25359879 DOI: 10.2967/jnumed.114.140780] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
UNLABELLED P2045 is a peptide analog of somatostatin with picomolar affinity for the somatostatin receptor subtype 2 (SSTR2) upregulated in some pancreatic tumors. Studies were conducted in rat AR42J pancreatic tumor xenograft mice to determine whether (188)Re-P2045 could inhibit the growth of pancreatic cancer in an animal model. METHODS (188)Re-P2045 was intravenously administered every 3 d for 16 d to nude mice with AR42J tumor xenografts that were approximately 20 mm(3) at study initiation. Tumor volumes were recorded throughout the dosing period. At necropsy, all tissues were assessed for levels of radioactivity and evaluated for histologic abnormalities. Clinical chemistry and hematology parameters were determined from terminal blood samples. The affinity of nonradioactive (185/187)Re-P2045 for somatostatin receptors was compared in human NCI-H69 and rat AR42J tumor cell membranes expressing predominantly SSTR2. RESULTS In the 1.85- and 5.55-MBq groups, tumor growth was inhibited in a dose-dependent fashion. In the 11.1-MBq group, tumor growth was completely inhibited throughout the dosing period and for 12 d after the last administered dose. The radioactivity level in tumors 4 h after injection was 10 percentage injected dose per gram, which was 2-fold higher than in the kidneys. (188)Re-P2045 was well tolerated in all dose groups, with no adverse clinical, histologic, or hematologic findings. The nonradioactive (185/187)Re-P2045 bound more avidly (0.2 nM) to SSTR2 in human than rat tumor membranes, suggesting that these studies are relevant to human studies. CONCLUSION (188)Re-P2045 is a promising therapeutic candidate for patients with somatostatin receptor-positive cancer.
Collapse
Affiliation(s)
- Carol A Nelson
- Translational Medicine Consulting, Westford, Massachusetts Department of Research and Development, Andarix Pharmaceuticals, Watertown, Massachusetts; and
| | - Michael T Azure
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Christopher T Adams
- Department of Research and Development, Andarix Pharmaceuticals, Watertown, Massachusetts; and
| | - Kurt R Zinn
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|