1
|
Martins RS, Jesus TT, Cardoso L, Soares P, Vinagre J. Personalized Medicine in Medullary Thyroid Carcinoma: A Broad Review of Emerging Treatments. J Pers Med 2023; 13:1132. [PMID: 37511745 PMCID: PMC10381735 DOI: 10.3390/jpm13071132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/08/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
Medullary thyroid carcinoma (MTC) arises from parafollicular cells in the thyroid gland, and although rare, it represents an aggressive type of thyroid cancer. MTC is recognized for its low mutational burden, with point mutations in RET or RAS genes being the most common oncogenic events. MTC can be resistant to cytotoxic chemotherapy, and multitarget kinase inhibitors (MKIs) have been considered a treatment option. They act by inhibiting the activities of specific tyrosine kinase receptors involved in tumor growth and angiogenesis. Several tyrosine kinase inhibitors are approved in the treatment of advanced MTC, including vandetanib and cabozantinib. However, due to the significant number of adverse events, debatable efficiency and resistance, there is a need for novel RET-specific TKIs. Newer RET-specific TKIs are expected to overcome previous limitations and improve patient outcomes. Herein, we aim to review MTC signaling pathways, the most recent options for treatment and the applications for personalized medicine.
Collapse
Affiliation(s)
- Rui Sousa Martins
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), 4200-135 Porto, Portugal
- Faculdade de Ciências da Universidade do Porto (FCUP), 4169-007 Porto, Portugal
| | - Tito Teles Jesus
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), 4200-135 Porto, Portugal
| | - Luís Cardoso
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), 4200-135 Porto, Portugal
- Departamento de Endocrinologia, Diabetes e Metabolismo do Centro Hospitalar Universitário de Coimbra, 3000-075 Coimbra, Portugal
| | - Paula Soares
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), 4200-135 Porto, Portugal
- Faculdade de Medicina da Universidade do Porto (FMUP), 4200-319 Porto, Portugal
| | - João Vinagre
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), 4200-135 Porto, Portugal
- Faculdade de Medicina da Universidade do Porto (FMUP), 4200-319 Porto, Portugal
| |
Collapse
|
2
|
Zhu YS, Tang K, Lv J. Peptide-drug conjugate-based novel molecular drug delivery system in cancer. Trends Pharmacol Sci 2021; 42:857-869. [PMID: 34334251 DOI: 10.1016/j.tips.2021.07.001] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 06/25/2021] [Accepted: 07/06/2021] [Indexed: 01/18/2023]
Abstract
Drug delivery systems are generally believed to comprise drugs and excipients. A peptide-drug conjugate is a single molecule that can simultaneously play multiple roles in a drug delivery system, such as in vivo drug distribution, targeted release, and bioactivity functions. This molecule can be regarded as an integrated drug delivery system, so it is called a molecular drug delivery system. In the context of cancer therapy, a peptide-drug conjugate comprises a tumor-targeting peptide, a payload, and a linker. Tumor-targeting peptides specifically identify membrane receptors on tumor cells, improve drug-targeted therapeutic effects, and reduce toxic and side effects. Payloads with bioactive functions connect to tumor-targeting peptides through linkers. In this review, we explored ongoing clinical work on peptide-drug conjugates targeting various receptors. We discuss the binding mechanisms of tumor-targeting peptides and related receptors, as well as the limiting factors for peptide-drug conjugate-based molecular drug delivery systems.
Collapse
Affiliation(s)
- Yi-Shen Zhu
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, Jiangsu Province, China.
| | - Kexing Tang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, Jiangsu Province, China
| | - Jiayi Lv
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, Jiangsu Province, China
| |
Collapse
|
3
|
Maiti BK, Govil N, Kundu T, Moura JJ. Designed Metal-ATCUN Derivatives: Redox- and Non-redox-Based Applications Relevant for Chemistry, Biology, and Medicine. iScience 2020; 23:101792. [PMID: 33294799 PMCID: PMC7701195 DOI: 10.1016/j.isci.2020.101792] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023] Open
Abstract
The designed "ATCUN" motif (amino-terminal copper and nickel binding site) is a replica of naturally occurring ATCUN site found in many proteins/peptides, and an attractive platform for multiple applications, which include nucleases, proteases, spectroscopic probes, imaging, and small molecule activation. ATCUN motifs are engineered at periphery by conjugation to recombinant proteins, peptides, fluorophores, or recognition domains through chemically or genetically, fulfilling the needs of various biological relevance and a wide range of practical usages. This chemistry has witnessed significant growth over the last few decades and several interesting ATCUN derivatives have been described. The redox role of the ATCUN moieties is also an important aspect to be considered. The redox potential of designed M-ATCUN derivatives is modulated by judicious choice of amino acid (including stereochemistry, charge, and position) that ultimately leads to the catalytic efficiency. In this context, a wide range of M-ATCUN derivatives have been designed purposefully for various redox- and non-redox-based applications, including spectroscopic probes, target-based catalytic metallodrugs, inhibition of amyloid-β toxicity, and telomere shortening, enzyme inactivation, biomolecules stitching or modification, next-generation antibiotic, and small molecule activation.
Collapse
Affiliation(s)
- Biplab K. Maiti
- National Institute of Technology Sikkim, Ravangla Campus, Barfung Block, Ravangla Sub Division, South Sikkim 737139, India
| | - Nidhi Govil
- National Institute of Technology Sikkim, Ravangla Campus, Barfung Block, Ravangla Sub Division, South Sikkim 737139, India
| | - Taraknath Kundu
- National Institute of Technology Sikkim, Ravangla Campus, Barfung Block, Ravangla Sub Division, South Sikkim 737139, India
| | - José J.G. Moura
- LAQV-REQUIMTE, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus de Caparica, 2829-516 Caparica, Portugal
| |
Collapse
|
4
|
Xiang Q, Li JJ, Li CY, Tian RB, Li XH. Somatostatin Type 2 Receptor Antibody Enhances Mechanical Hyperalgesia in the Dorsal Root Ganglion Neurons after Sciatic Nerve-pinch Injury: Evidence of Behavioral Studies and Bax Protein Expression. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2019; 18:791-797. [PMID: 31686636 DOI: 10.2174/1871527318666191101094412] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 09/16/2019] [Accepted: 10/22/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Our previous study has indicated that somatostatin potently inhibits neuropathic pain through the activation of its type 2 receptor (SSTR2) in mouse dorsal root ganglion and spinal cord. However, the underlying mechanism of this activation has not been elucidated clearly. OBJECTIVE The aim of this study is to perform the pharmacological studies on the basis of sciatic nerve-pinch mice model and explore the underlying mechanism involving SSTR2. METHODS On the basis of a sciatic nerve-pinch injury model, we aimed at comparing the painful behavior and dorsal root ganglion neurons neurochemical changes after the SSTR2 antibody (anti- SSTR2;5μl,1μg/ml) administration in the mouse. RESULTS After pinch nerve injury, we found that the mechanical hyperalgesia and severely painful behavior (autotomy) were detected after the application of SSTR2 antibody (anti-SSTR2; 5μl, 1μg/ml) on the pinch-injured nerve. The up-regulated phosphorylated ERK (p-ERK) expression and the apoptotic marker (i.e., Bax) were significantly decreased in DRGs after anti-SSTR2 treatment. CONCLUSION The current data suggested that inhibitory changes in proteins from the apoptotic pathway in anti-SSTR2-treated groups might be taking place to overcome the protein deficits caused by SSTR2 antibody and supported the new therapeutic intervention with SSTR2 antagonist for neuronal degeneration following nerve injury.
Collapse
Affiliation(s)
- Qiong Xiang
- Institute of Medicine, Medical Research Center, Jishou University, Hunan, China
| | - Jing-Jing Li
- Institute of Medicine, Medical Research Center, Jishou University, Hunan, China
| | - Chun-Yan Li
- Institute of Medicine, Medical Research Center, Jishou University, Hunan, China
| | - Rong-Bo Tian
- Institute of Medicine, Medical Research Center, Jishou University, Hunan, China
| | - Xian-Hui Li
- Institute of Medicine, Medical Research Center, Jishou University, Hunan, China
| |
Collapse
|
5
|
Wang S, Zhang R, Hou X, Jiang F, Wang J, He Z, Jiang F, Hu C, Jia W. Association between serum somatostatin levels and glucose-lipid metabolism in the Jino ethnic minority and Han Chinese population. SCIENCE CHINA-LIFE SCIENCES 2018; 61:1382-1388. [PMID: 29995197 DOI: 10.1007/s11427-017-9289-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 04/08/2018] [Indexed: 10/28/2022]
Abstract
We aim to investigate the relationship between serum somatostatin (SST) levels and glucose-lipid metabolism at various stages of glucose tolerance in the Jino ethnic minority (n=111) and Han population (n=113) of Yunnan Province, southwest China. Anthropometric parameters and biochemical traits were measured. Serum SST and plasma glucagon levels were tested. Participants were divided into three subgroups: isolated fasting hyperglycemia (IFH), isolated post challenge hyperglycemia (IPH) and normal glucose tolerance (NGT). SST levels were found lower while glucagon levels were significantly higher in the Jino ethnic with IPH (P=0.0026 and P=0.0069, respectively). Fasting glucose and high density lipoprotein-cholesterol (HDL-C) levels were higher (P=0.0055 and P=0.0021, respectively) and fasting insulin levels and homeostasis model assessments β-cell function were lower (P=0.0479 and P=0.0007, respectively) in the Jino population. After adjusting for confounding factors, the serum SST level was associated with glucagon (P<0.0001) in both populations. The SST level was correlated with fasting Cpeptide (P=0.0267) in Jino and HDL-C levels in Han (P=0.0079). Our findings suggest that serum SST levels and plasma glucagon levels may vary in subjects with IPH between two ethnics.
Collapse
Affiliation(s)
- Shiyun Wang
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center for Diabetes, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Rong Zhang
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center for Diabetes, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Xuhong Hou
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center for Diabetes, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Fusong Jiang
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center for Diabetes, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Jie Wang
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center for Diabetes, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Zhen He
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center for Diabetes, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Feng Jiang
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center for Diabetes, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Cheng Hu
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center for Diabetes, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China. .,Shanghai Jiao Tong University Affiliated Sixth People's Hospital South Campus, Shanghai, 200233, China.
| | - Weiping Jia
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center for Diabetes, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.
| |
Collapse
|
6
|
Sizdahkhani S, Feldman MJ, Piazza MG, Ksendzovsky A, Edwards NA, Ray-Chaudhury A, Maric D, Merrill MJ, Pacak K, Zhuang Z, Chittiboina P. Somatostatin receptor expression on von Hippel-Lindau-associated hemangioblastomas offers novel therapeutic target. Sci Rep 2017; 7:40822. [PMID: 28094316 PMCID: PMC5240113 DOI: 10.1038/srep40822] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 12/12/2016] [Indexed: 01/03/2023] Open
Abstract
Von Hippel-Lindau (VHL)-associated hemangioblastomas (VHL-HB) arise in the central nervous system (CNS), and are a leading cause of morbidity and mortality in VHL disease. Currently, surgical resection is the most effective way to manage symptomatic VHL-HBs. Surgically unresectable VHL-HBs or those in frail patients are challenging problems. Therapies targeting oncologic and vascular endothelial growth factor (VEGF) pathways have failed to demonstrate tumor control. Our experience and previous reports on VHL-HB avidity to somatostatin analogues suggested somatostatin receptor (SSTR) expression in VHL-HBs, offering an alternative therapeutic strategy. We explored this possibility by demonstrating consistent histologic expression of SSTR1, 2a, 4, and 5 in VHL-HBs. We found that somatostatin analogue octreotide induces apoptosis in VHL-HB stromal cells in a dose-dependent fashion by BAX – caspase-3 pathway unrelated to canonical VHL pathway. When administered to a patient with unresectable symptomatic suprasellar hemangioblastoma, octreotide resulted in tumor volume reduction, symptom stabilization, and tumor cytopenia on repeat 68Ga-DOTA-TATE positron emission tomography (PET) within 6 months, suggesting tumor infarction. We conclude that VHL-HBs harbor multiple SSTR subtypes that offer actionable chemo-therapeutic strategy for management of symptomatic, unresectable tumors by somatostatin analogue therapy.
Collapse
Affiliation(s)
- Saman Sizdahkhani
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Michael J Feldman
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Martin G Piazza
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Alexander Ksendzovsky
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA.,Department of Neurosurgery, University of Virginia Health System, Charlottesville, Virginia USA
| | - Nancy A Edwards
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Abhik Ray-Chaudhury
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Dragan Maric
- Flow Cytometry Core, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Marsha J Merrill
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Karel Pacak
- Section on Medical Neuroendocrinology, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Zhengping Zhuang
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Prashant Chittiboina
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
7
|
van Adrichem RCS, de Herder WW, Kamp K, Brugts MP, de Krijger RR, Sprij-Mooij DM, Lamberts SWJ, van Koetsveld PM, Janssen JAMJL, Hofland LJ. Effects of Somatostatin Analogs and Dopamine Agonists on Insulin-Like Growth Factor 2-Induced Insulin Receptor Isoform A Activation by Gastroenteropancreatic Neuroendocrine Tumor Cells. Neuroendocrinology 2016; 103:815-25. [PMID: 26836610 DOI: 10.1159/000444280] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 01/17/2016] [Indexed: 11/19/2022]
Abstract
BACKGROUND Gastroenteropancreatic neuroendocrine tumors (GEP-NETs) express insulin-like growth factor (IGF)-related factors [IGF1, IGF2; insulin receptor (IR)-A, IR-B; IGF-binding protein (IGFBP) 1-3] as well as somatostatin (SSTRs) and dopamine D2 receptors (D2Rs). OBJECTIVES To (1) compare mRNA expression of IGF-related factors in human pancreatic NET (panNET) cell lines with that in human GEP-NETs to evaluate the usefulness of these cells as a model for studying the IGF system in GEP-NETs, (2) determine whether panNET cells produce growth factors that activate IR-A, and (3) investigate whether somatostatin analogs (SSAs) and/or dopamine agonists (DAs) influence the production of these growth factors. METHODS In panNET cells (BON-1 and QGP-1) and GEP-NETs, mRNA expression of IGF-related factors was measured by quantitative real-time PCR. Effects of the SSAs octreotide and pasireotide (PAS), the DA cabergoline (CAB), and the dopastatin BIM-23A760 (all 100 nM) were evaluated at the IGF2 mRNA and protein level (by ELISA) and regarding IR-A bioactivity (by kinase receptor activation assay) in panNET cells. RESULTS panNET cells and GEP-NETs had comparable expression profiles of IGF-related factors. Especially in BON-1 cells, IGF2 and IR-A were most highly expressed. PAS + CAB inhibited IGF2 (-29.5 ± 4.9%, p < 0.01) and IGFBP3 (-20.0 ± 4.0%, p < 0.01) mRNA expression in BON-1 cells. In BON-1 cells, IGF2 protein secretion was significantly inhibited with BIM-23A760 (-23.7 ± 3.8%). BON-1- but not QGP-1- conditioned medium stimulated IR-A bioactivity. In BON-1 cells, IR-A bioactivity was inhibited by BIM-23A760 and PAS + CAB (-37.8 ± 2.1% and -30.9 ± 4.1%, respectively, p < 0.0001). CONCLUSIONS (1) The BON-1 cell line is a representative model for studying the IGF system in GEP-NETs, (2) BON-1 cells produce growth factors (IGF2) activating IR-A, and (3) combined SSTR and D2R targeting with PAS + CAB and BIM-23A760 suppresses IGF2-induced IR-A activation.
Collapse
MESH Headings
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Cell Line, Tumor/chemistry
- Culture Media, Conditioned/pharmacology
- Dopamine/analogs & derivatives
- Dopamine/pharmacology
- Dopamine Agonists/pharmacology
- Enzyme-Linked Immunosorbent Assay
- Gene Expression Regulation, Neoplastic/drug effects
- HEK293 Cells
- Humans
- Insulin-Like Growth Factor II/metabolism
- Intestinal Neoplasms/pathology
- Neuroendocrine Tumors/pathology
- Pancreatic Neoplasms/pathology
- RNA, Messenger/metabolism
- Receptor, Insulin/genetics
- Receptor, Insulin/metabolism
- Receptors, Dopamine D2/genetics
- Receptors, Dopamine D2/metabolism
- Receptors, Somatostatin/genetics
- Receptors, Somatostatin/metabolism
- Somatostatin/analogs & derivatives
- Somatostatin/pharmacology
- Stomach Neoplasms/pathology
- Transfection
Collapse
Affiliation(s)
- Roxanne C S van Adrichem
- Division of Endocrinology, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Hernández B, López-Tobar E, Sanchez-Cortes S, Coïc YM, Baron B, Chenal A, Kruglik SG, Pflüger F, Cohen R, Ghomi M. From bulk to plasmonic nanoparticle surfaces: the behavior of two potent therapeutic peptides, octreotide and pasireotide. Phys Chem Chem Phys 2016; 18:24437-50. [DOI: 10.1039/c6cp04421b] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Structural dynamics of two potent somatostatin analogues in an aqueous environment and their binding sites on plasmonic nanoparticles were described.
Collapse
Affiliation(s)
- Belén Hernández
- Sorbonne Paris Cité
- Université Paris 13
- Groupe de Biophysique Moléculaire
- UFR Santé-Médecine-Biologie Humaine
- 93017 Bobigny Cedex
| | | | | | - Yves-Marie Coïc
- Institut Pasteur
- Unité de Chimie des Biomolécules
- UMR 3523
- 75724 Paris Cedex 15
- France
| | - Bruno Baron
- Institut Pasteur
- Plate-Forme de Biophysique de Macromolécules et de leurs Interactions
- 75724 Paris Cedex 15
- France
| | - Alexandre Chenal
- Institut Pasteur
- Unité Biochimie des Interactions Macromoléculaires
- UMR CNRS 3528
- 75724 Paris Cedex 15
- France
| | - Sergei G. Kruglik
- Sorbonne Universités
- UPMC Univ. Paris 06
- UMR 8237
- Laboratoire Jean Perrin
- 75005 Paris
| | - Fernando Pflüger
- Sorbonne Paris Cité
- Université Paris 13
- Groupe de Biophysique Moléculaire
- UFR Santé-Médecine-Biologie Humaine
- 93017 Bobigny Cedex
| | - Régis Cohen
- Service d’Endocrinologie
- Centre Hospitalier de Saint-Denis
- 93200 Saint-Denis
- France
| | - Mahmoud Ghomi
- Sorbonne Paris Cité
- Université Paris 13
- Groupe de Biophysique Moléculaire
- UFR Santé-Médecine-Biologie Humaine
- 93017 Bobigny Cedex
| |
Collapse
|
9
|
Temmerman F, Ho TA, Vanslembrouck R, Coudyzer W, Billen J, Dobbels F, van Pelt J, Bammens B, Pirson Y, Nevens F. Lanreotide Reduces Liver Volume, But Might Not Improve Muscle Wasting or Weight Loss, in Patients With Symptomatic Polycystic Liver Disease. Clin Gastroenterol Hepatol 2015; 13:2353-9.e1. [PMID: 26073493 DOI: 10.1016/j.cgh.2015.05.039] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 05/25/2015] [Accepted: 05/27/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Polycystic liver disease (PCLD) can induce malnutrition owing to extensive hepatomegaly and patients might require liver transplantation. Six months of treatment with the somatostatin analogue lanreotide (120 mg) reduces liver volume. We investigated the efficacy of a lower dose of lanreotide and its effects on nutritional status. METHODS We performed an 18-month prospective study at 2 tertiary medical centers in Belgium from January 2011 through August 2012. Fifty-nine patients with symptomatic PCLD were given lanreotide (90 mg, every 4 weeks) for 6 months. Patients with reductions in liver volume of more than 100 mL (responders, primary end point) continued to receive lanreotide (90 mg) for an additional year (18 months total). Nonresponders were offered increased doses, up to 120 mg lanreotide, until 18 months. Liver volume and body composition were measured by computed tomography at baseline and at months 6 and 18. Patients also were assessed by the PCLD-specific complaint assessment at these time points. RESULTS Fifty-three patients completed the study; 21 patients (40%) were responders. Nineteen of the responders (90%) continued as responders until 18 months. At this time point, they had a mean reduction in absolute liver volume of 430 ± 92 mL. In nonresponders (n = 32), liver volume increased by a mean volume of 120 ± 42 mL at 6 months. However, no further increase was observed after dose escalation in the 24 patients who continued to the 18-month end point. All subjects had decreased scores on all subscales of the PCLD-specific complaint assessment, including better food intake (P = .04). Subjects did not have a mean change in subcutaneous or visceral fat mass, but did have decreases in mean body weight (2 kg) and total muscle mass (1.06 cm(2)/h(2)). Subjects also had a significant mean reduction in their level of insulin-like growth factor 1, from 19% below the age-adjusted normal range level at baseline to 50% at 18 months (P = .002). CONCLUSIONS In a prospective study, we observed that low doses of lanreotide (90 mg every 4 weeks) reduced liver volumes and symptoms in patients with PCLD. However, patients continued to lose weight and muscle mass. The effects of somatostatin analogues on sarcopenia require investigation. Clinicaltrials.gov: NCT01315795.
Collapse
Affiliation(s)
- Frederik Temmerman
- Division and Laboratory of Hepatology, University Hospitals, KU Leuven, Leuven, Belgium.
| | - Thien Ahn Ho
- Division of Nephrology, Université Catholique de Louvain, Brussels, Belgium
| | | | - Walter Coudyzer
- Division of Radiology, University Hospitals, KU Leuven, Leuven, Belgium
| | - Jaak Billen
- Division of Laboratory Medicine, University Hospitals, KU Leuven, Leuven, Belgium
| | - Fabienne Dobbels
- Division of Public Health and Primary Care, University Hospitals, KU Leuven, Leuven, Belgium
| | - Jos van Pelt
- Division and Laboratory of Hepatology, University Hospitals, KU Leuven, Leuven, Belgium
| | - Bert Bammens
- Division of Nephrology, University Hospitals, KU Leuven, Leuven, Belgium
| | - Yves Pirson
- Division of Nephrology, Université Catholique de Louvain, Brussels, Belgium
| | - Frederik Nevens
- Division and Laboratory of Hepatology, University Hospitals, KU Leuven, Leuven, Belgium
| |
Collapse
|
10
|
Lelle M, Kaloyanova S, Freidel C, Theodoropoulou M, Musheev M, Niehrs C, Stalla G, Peneva K. Octreotide-Mediated Tumor-Targeted Drug Delivery via a Cleavable Doxorubicin-Peptide Conjugate. Mol Pharm 2015; 12:4290-300. [PMID: 26524088 DOI: 10.1021/acs.molpharmaceut.5b00487] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Although recent methods for targeted drug delivery have addressed many of the existing problems of cancer therapy associated with undesirable side effects, significant challenges remain that have to be met before they find significant clinical relevance. One such area is the delicate chemical bond that is applied to connect a cytotoxic drug with targeting moieties like antibodies or peptides. Here we describe a novel platform that can be utilized for the preparation of drug-carrier conjugates in a site-specific manner, which provides excellent versatility and enables triggered release inside cancer cells. Its key feature is a cleavable doxorubicin-octreotide bioconjugate that targets overexpressed somatostatin receptors on tumor cells, where the coupling between the two components was achieved through the first cleavable disulfide-intercalating linker. The tumor targeting ability and suppression of adrenocorticotropic hormone secretion in AtT-20 cells by both octreotide and the doxorubicin hybrid were determined via a specific radioimmunoassay. Both substances reduced the hormone secretion to a similar extent, which demonstrated that the tumor homing peptide is able to interact with the relevant cell surface receptors after the attachment of the drug. Effective drug release was quickly accomplished in the presence of the physiological reducing agent glutathione. We also demonstrate the relevance of this scaffold in biological context in cytotoxicity assays with pituitary, pancreatic, and breast cancer cell lines.
Collapse
Affiliation(s)
- Marco Lelle
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Stefka Kaloyanova
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Christoph Freidel
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | | | - Michael Musheev
- Institute of Molecular Biology, Ackermannweg 4, 55128 Mainz, Germany
| | - Christof Niehrs
- Institute of Molecular Biology, Ackermannweg 4, 55128 Mainz, Germany.,Division of Molecular Embryology, DKFZ-ZMBH Alliance , 69120 Heidelberg, Germany
| | - Günter Stalla
- Max Planck Institute for Psychiatry, Kraepelinstraße 2-10, 80804 Munich, Germany
| | - Kalina Peneva
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.,Institute of Organic Chemistry and Macromolecular Chemistry, Friedrich Schiller University Jena , Lessingstrasse 8, 07743 Jena, Germany
| |
Collapse
|
11
|
Extended release microparticle-in-gel formulation of octreotide: Effect of polymer type on acylation of peptide during in vitro release. Int J Pharm 2015; 496:676-88. [PMID: 26561725 DOI: 10.1016/j.ijpharm.2015.11.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2015] [Revised: 10/25/2015] [Accepted: 11/02/2015] [Indexed: 12/31/2022]
Abstract
Polymeric microparticles (MPs)-in-gel formulations for extended delivery of octreotide were developed. We investigated influence of polymer composition on acylation of octreotide and kinetics of release during in vitro release from biodegradable polymeric formulations. Polycaprolactone (PCL), polylactic acid (PLA), polyglycolic acid (PGA) and polyethylene glycol (PEG) based triblock (TB≈PCL10k-PEG2k-PCL10k) and pentablock (PBA≈PLA3k-PCL7k-PEG2k-PCL7k-PLA3k and PBB≈PGA3k-PCL7k-PEG2k-PCL7k-PGA3k) polymers were investigated. Octreotide was encapsulated in MPs using methanol-oil/water emulsion solvent evaporation method. The particles were characterized for size, morphology, encapsulation efficiency, drug loading and in vitro release. Release samples were subjected to HPLC analysis for quantitation and HPLC-MS analysis for identification of native and chemically modified octreotide adducts. Entrapment efficiency of methanol-oil/water method with TB, PBA and PBB polymers were 45%, 60%, and 82%, respectively. A significant fraction of released octreotide was acylated from lactide and glycolide based PBA (53%) and PBB (92%) polymers. Substantial amount of peptide was not released from PBB polymers after 330 days of incubation. Complete release of octreotide was achieved from TB polymer over a period of 3 months with minimal acylation of peptide (13%). PCL based polymers resulted in minimal acylation of peptide and hence may be suitable for extended peptide and protein delivery. Conversely, polymers having PLA and PGA blocks may not be appropriate for peptide delivery due to acylation and incomplete release.
Collapse
|
12
|
Reversible hydrophobic ion-paring complex strategy to minimize acylation of octreotide during long-term delivery from PLGA microparticles. Int J Pharm 2015; 489:237-45. [PMID: 25940041 DOI: 10.1016/j.ijpharm.2015.04.075] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Revised: 04/22/2015] [Accepted: 04/28/2015] [Indexed: 11/22/2022]
Abstract
Acylation of peptide has been reported for a number of peptides and proteins during release from polymers comprising of lactide and glycolide. We hypothesize that reversible hydrophobic ion-pairing (HIP) complex may minimize octreotide acylation during release. Sodium dodecyl sulfate (SDS), dextran sulfate A (DSA, Mw 9-20 kDa) and dextran sulfate B (DSB, Mw 36-50 kDa) were selected as ion-pairing agents to prepare reversible HIP complex with octreotide. Complexation efficiency was optimized with respect to the mole ratio of ion-pairing agent to octreotide to achieve 100% complexation of octreotide. Dissociation studies suggested that DSA-octreotide and DSB-octreotide complexes dissociate completely at physiological pH in presence of counter ions unlike SDS-octreotide complex. DSA-octreotide and DSB-octreotide complex encapsulated PLGA microparticles (DSAMPs and DSBMPs) were prepared using the S/O/W emulsion method. Entrapment efficiencies for DSAMPs and DSBMPs were 74.7±8.4% and 81.7±6.3%, respectively. In vitro release of octreotide was performed by suspending MPs in gel. A large fraction of peptide was released in chemically intact form and <7% was acylated from DSAMPs and DSBMPs in gel over 55 days. Therefore, HIP complexation could be a viable strategy to minimize acylation of peptides and proteins during extended release from lactide and glycolide based polymers.
Collapse
|
13
|
Pivonello C, De Martino MC, Negri M, Cuomo G, Cariati F, Izzo F, Colao A, Pivonello R. The GH-IGF-SST system in hepatocellular carcinoma: biological and molecular pathogenetic mechanisms and therapeutic targets. Infect Agent Cancer 2014; 9:27. [PMID: 25225571 PMCID: PMC4164328 DOI: 10.1186/1750-9378-9-27] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Accepted: 06/23/2014] [Indexed: 12/15/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the sixth most common malignancy worldwide. Different signalling pathways have been identified to be implicated in the pathogenesis of HCC; among these, GH, IGF and somatostatin (SST) pathways have emerged as some of the major pathways implicated in the development of HCC. Physiologically, GH-IGF-SST system plays a crucial role in liver growth and development since GH induces IGF1 and IGF2 secretion and the expression of their receptors, involved in hepatocytes cell proliferation, differentiation and metabolism. On the other hand, somatostatin receptors (SSTRs) are exclusively present on the biliary tract. Importantly, the GH-IGF-SST system components have been indicated as regulators of hepatocarcinogenesis. Reduction of GH binding affinity to GH receptor, decreased serum IGF1 and increased serum IGF2 production, overexpression of IGF1 receptor, loss of function of IGF2 receptor and appearance of SSTRs are frequently observed in human HCC. In particular, recently, many studies have evaluated the correlation between increased levels of IGF1 receptors and liver diseases and the oncogenic role of IGF2 and its involvement in angiogenesis, migration and, consequently, in tumour progression. SST directly or indirectly influences tumour growth and development through the inhibition of cell proliferation and secretion and induction of apoptosis, even though SST role in hepatocarcinogenesis is still opened to argument. This review addresses the present evidences suggesting a role of the GH-IGF-SST system in the development and progression of HCC, and describes the therapeutic perspectives, based on the targeting of GH-IGF-SST system, which have been hypothesised and experimented in HCC.
Collapse
Affiliation(s)
- Claudia Pivonello
- Dipartimento di Medicina Clinica e Chirurgia, Università Federico II di Napoli, Via Sergio Pansini, 5, Naples 80131, Italy
| | - Maria Cristina De Martino
- Dipartimento di Medicina Clinica e Chirurgia, Università Federico II di Napoli, Via Sergio Pansini, 5, Naples 80131, Italy
| | - Mariarosaria Negri
- Dipartimento di Medicina Clinica e Chirurgia, Università Federico II di Napoli, Via Sergio Pansini, 5, Naples 80131, Italy
| | | | - Federica Cariati
- Dipartimento di Medicina Clinica e Chirurgia, Università Federico II di Napoli, Via Sergio Pansini, 5, Naples 80131, Italy
| | - Francesco Izzo
- National Cancer Institute G Pascale Foundation, Naples, Italy
| | - Annamaria Colao
- Dipartimento di Medicina Clinica e Chirurgia, Università Federico II di Napoli, Via Sergio Pansini, 5, Naples 80131, Italy
| | - Rosario Pivonello
- Dipartimento di Medicina Clinica e Chirurgia, Università Federico II di Napoli, Via Sergio Pansini, 5, Naples 80131, Italy
| |
Collapse
|
14
|
Celik N, Cinaz P, Emeksiz HC, Hussain K, Çamurdan O, Bideci A, Döğer E, Yüce Ö, Türkyılmaz Z, Oğuz AD. Octreotide-induced long QT syndrome in a child with congenital hyperinsulinemia and a novel missense mutation (p.Met115Val) in the ABCC8 gene. Horm Res Paediatr 2014; 80:299-303. [PMID: 24080777 DOI: 10.1159/000354666] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Accepted: 07/24/2013] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS Congenital hyperinsulinism (CHI) denotes an inappropriate secretion of insulin from pancreatic β-cells in the presence of a low blood glucose level due to various genetic causes. Diazoxide is the first-line medical treatment for CHI. In case of failure, a somatostatin analogue called octreotide is used. A prolonged QT interval is an unusual side effect of octreotide which can be lethal if unrecognized. CASE PRESENTATION We report on a 35-day-old infant who was diagnosed with CHI on the 3rd day of his life and underwent pancreatectomy due to failure of medical treatment at 8 months. His genetic analysis revealed a compound heterozygosity for a novel missense mutation (p.Met115Val) and a nonsense mutation (p.Trp1339X) in the ABCC8 gene. Furthermore, at the 6th month of follow-up, a long QT (0.49 s) was determined by ECG examination, which was normalized following discontinuation of octreotide treatment after pancreatectomy. Thus, the long QT was considered to be secondary to octreotide medication. CONCLUSION We recommend ECG monitoring before and during octreotide treatment in order to recognize a prolonged QT interval and to prevent related complications in cases with congenital hyperinsulinemia.
Collapse
Affiliation(s)
- Nurullah Celik
- Department of Pediatric Endocrinology, Gazi University, Ankara, Turkey
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Trahair LG, Horowitz M, Jones KL. Postprandial hypotension: a systematic review. J Am Med Dir Assoc 2014; 15:394-409. [PMID: 24630686 DOI: 10.1016/j.jamda.2014.01.011] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 01/24/2014] [Indexed: 02/07/2023]
Abstract
BACKGROUND Postprandial hypotension (PPH) is an important clinical problem, which has received inappropriately little attention. METHODS A systematic search of the databases PubMed, Embase, Cochrane Library, and Web of Knowledge, from their inception to the present time, was conducted to identify studies relevant to the epidemiology, pathophysiology, and/or management of PPH. RESULTS A total of 417 full-text papers were retrieved from database searching and, following screening, 248 were retained. Of these, 167 papers were considered eligible for inclusion. CONCLUSIONS PPH occurs commonly in older people and represents a major cause of morbidity. Although the pathophysiology of PPH remains poorly defined, diverse factors, including impairments in sympathetic and baroreflex function, release of vasodilatory peptides, the rate of small intestinal nutrient delivery, gastric distension, and splanchnic blood pooling, appear important. Current pharmacologic and nonpharmacologic management is suboptimal. Research into the pathophysiology of PPH represents a priority so that management can be targeted more effectively.
Collapse
Affiliation(s)
- Laurence G Trahair
- Discipline of Medicine, University of Adelaide, Adelaide, Australia; NHMRC Center of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, Australia
| | - Michael Horowitz
- Discipline of Medicine, University of Adelaide, Adelaide, Australia; NHMRC Center of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, Australia
| | - Karen L Jones
- Discipline of Medicine, University of Adelaide, Adelaide, Australia; NHMRC Center of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, Australia.
| |
Collapse
|
16
|
Abstract
This paper summarizes the current understanding of the biology of somatostatin receptor (sst), role of immunotherapy in neuroendocrine tumor (NET), new agents for PPRT, and methods to assess response and clinical benefit in NET. One of the most interesting aspects of sst biology is the recent discovery of truncated variants of the sst5 receptor subtype with unique tissue distribution and response to somatostatin (SST). These truncated receptors are associated with bad patient prognosis, decreased response to SST analogs, and may be new targets for diagnoses and treatment. IFN remains a cost-effective agent, particularly in classic mid gut carcinoids, and there is interest to continue examining immunotherapy's in this disease. PRRT remains a key strategy for treatment and imaging. In addition to the classic agents, there are a series of new agents targeting other receptors such as the incretin receptors (GLP-1R; GIPR) and other G-protein coupled receptors with great potential. With regards to therapy monitoring, the most commonly used criteria are Response Criteria Evaluation in Solid Tumors (RECIST). However, for different reasons, these criteria are not very useful in NET. Incorporation of other criteria such as Choi as well as functional imaging assessment with PET would be of great interest in this area.
Collapse
|
17
|
Harvey M, Cave G. Octreotide may attenuate absorption and ameliorate toxicity following enteric drug overdose. Med Hypotheses 2013; 81:424-5. [DOI: 10.1016/j.mehy.2013.05.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Accepted: 05/27/2013] [Indexed: 11/28/2022]
|
18
|
Veenstra MJ, de Herder WW, Feelders RA, Hofland LJ. Targeting the somatostatin receptor in pituitary and neuroendocrine tumors. Expert Opin Ther Targets 2013; 17:1329-43. [DOI: 10.1517/14728222.2013.830711] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
19
|
Temmerman F, Gevers T, Ho TA, Vanslembrouck R, Coudyzer W, van Pelt J, Bammens B, Pirson Y, Drenth JP, Nevens F. Safety and efficacy of different lanreotide doses in the treatment of polycystic liver disease: pooled analysis of individual patient data. Aliment Pharmacol Ther 2013; 38:397-406. [PMID: 23799922 DOI: 10.1111/apt.12384] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2013] [Revised: 04/01/2013] [Accepted: 06/04/2013] [Indexed: 01/02/2023]
Abstract
BACKGROUND Long-acting lanreotide (LAN) 120 mg every 4 weeks reduces liver volume (LV) in patients with polycystic liver diseases (PCLD). Animal studies demonstrated that the inhibition of hepatic and renal cystogenesis is dose dependent. AIM To investigate the safety and efficacy of two different LAN doses in PCLD patients. METHODS The 6-month results of the LOCKCYST I trial, its extension study and the LOCKCYST II trial were pooled. LV at baseline and month 6 was measured by CT-scan and blindly re-analysed by two independent radiologists. RESULTS The study population [132 treatment periods, age 49 years (IQR: 45-55), 114 women] consisted of three groups. Each received treatment every 4 weeks during 6 months: placebo (n = 26); LAN 90 mg (n = 55) or LAN 120 mg (n = 51). The inter-observer variability and agreement in the calculation of LV were excellent. Severe side effects occurred with placebo, LAN 90 mg and LAN 120 mg in respectively 0%, 7% and 16%. Change in LV's after 6 months in these three groups were respectively: increase of +36 mL [(-45)-(+138)]; decrease of -82 mL [(-285)-(+92)] and decrease of -123 mL [(-312)-(+4)] (Kruskal-Wallis One Way anova on Ranks; P = 0.002). Based on ROC analysis, a reduction of ≥120 mL in LV has a positive predictive value of 64% for improving symptoms (ROC analysis AUC: 0.729; sensitivity 73%, specificity 69%, P < 0.0001). CONCLUSIONS Both LAN 90 mg and LAN 120 mg reduce liver volume. LAN 90 mg has less side effects. This suggests that in case of intolerance to LAN 120 mg, a dose reduction to LAN 90 mg is meaningful.
Collapse
Affiliation(s)
- F Temmerman
- Department and Laboratory of Hepatology, University Hospitals KULeuven, Leuven, Belgium.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
|
21
|
Oberg K, Casanovas O, Castaño JP, Chung D, Delle Fave G, Denèfle P, Harris P, Khan MS, Kulke MH, Scarpa A, Tang LH, Wiedenmann B. Molecular pathogenesis of neuroendocrine tumors: implications for current and future therapeutic approaches. Clin Cancer Res 2013; 19:2842-9. [PMID: 23459719 DOI: 10.1158/1078-0432.ccr-12-3458] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The treatment landscape and biologic understanding of neuroendocrine tumors (NET) has shifted dramatically in recent years. Recent studies have shown that somatostatin analogues have the potential not only to control symptoms of hormone hypersecretion but also have the ability to slow tumor growth in patients with advanced carcinoid. The results of clinical trials have further shown that the VEGF pathway inhibitor sunitinib and the mTOR inhibitor everolimus have efficacy in patients with advanced pancreatic NETs. The efficacy of these targeted therapies in NET suggests that the molecular characterization of NETs may provide an avenue to predict both which patients may benefit most from the treatment and to overcome potential drug resistance. Recent genomic studies of NETs have further suggested that pathways regulating chromatin remodeling and epigenetic modification may play a key role in regulating NET growth. These observations offer the potential for new therapeutic and diagnostic advances for patients with NET.
Collapse
Affiliation(s)
- Kjell Oberg
- Department of Endocrine Oncology, University Hospital, Uppsala, Sweden.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Lupp A, Nagel F, Schulz S. Reevaluation of sst₁ somatostatin receptor expression in human normal and neoplastic tissues using the novel rabbit monoclonal antibody UMB-7. ACTA ACUST UNITED AC 2013; 183:1-6. [PMID: 23466804 DOI: 10.1016/j.regpep.2013.02.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Revised: 12/20/2012] [Accepted: 02/13/2013] [Indexed: 11/17/2022]
Abstract
BACKGROUND The somatostatin receptor 1 (sst1) is widely distributed throughout the body and is also present in neoplastic tissues. However, little is known about its precise tissue distribution, regulation and function, which may in part be due to the lack of specific monoclonal anti-sst1 antibodies. METHODS We have characterized the novel rabbit monoclonal anti-human sst1 antibody UMB-7 using sst1-expressing cells and human pituitary samples. The antibody was then used for immunohistochemical staining of a large panel of formalin-fixed, paraffin-embedded human tissues. RESULTS Western blot analyses of BON-1 cells and human pituitary revealed a broad band migrating at a molecular weight of 45,000-60,000. After enzymatic deglycosylation the size of this band decreased to a molecular weight of 45,000. UMB-7 yielded an efficient immunostaining of distinct cell populations in the human tissue samples with a predominance of plasma membrane staining, which was completely abolished by preadsorption of UMB-7 with its immunizing peptide. The sst1 receptor was detected in anterior pituitary, pancreatic islets, distal tubules, enteric ganglion cells and nerve fibers, chief cells of the gastric mucosa, macrophages and mast cells. In addition, sst1 was observed in pituitary adenomas, gastrointestinal neuroendocrine tumors and pheochromocytoma as well as in pancreatic adenocarcinomas, gastric carcinomas, urinary bladder carcinomas and sarcomas. CONCLUSIONS UMB-7 may prove of great value in the identification of sst1-expressing tumors during routine histopathological examinations. This may open up new routes for diagnostic and therapeutic intervention.
Collapse
Affiliation(s)
- Amelie Lupp
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Germany
| | - Falko Nagel
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Germany
| | - Stefan Schulz
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Germany.
| |
Collapse
|
23
|
Fusco A, Giampietro A, Bianchi A, Cimino V, Lugli F, Piacentini S, Lorusso M, Tofani A, Perotti G, Lauriola L, Anile C, Maira G, Pontecorvi A, De Marinis L. Treatment with octreotide LAR in clinically non-functioning pituitary adenoma: results from a case-control study. Pituitary 2012; 15:571-8. [PMID: 22207350 DOI: 10.1007/s11102-011-0370-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
Surgical cure cannot be achieved in most patients with invasive non-functioning pituitary macroadenoma (NFPA). Short-term residual tumor treatment with somatostatin analogs has produced disappointing results. This prospective case-control study assessed the efficacy of chronic treatment with long acting octreotide (octreotide LAR) on tumor volume in patients harboring post-surgical NFPA residue. The study population comprised 39 patients with NFPAs not cured by surgery. All patients underwent somatostatin receptor scintigraphy at least 6 months after the last surgery. Patients with a positive pituitary level octreoscan at (n = 26) received octreotide LAR (20 mg every 28 days) for ≥ 12 months (mean follow-up 37 ± 18 months) (Treated group). Moreover, a fragment of tumor tissue from patients in the treated group was retrospectively collected to assess the immunohistochemical expression of somatostatin receptor subtypes (SSTRs). The patients with a negative octreoscan (n = 13) formed the control group (mean follow-up 37 ± 16 months). Hormonal, radiological and visual field parameters were periodically assessed. In the treated group, all tumors expressed at least one SSTR subtype. The SSTR5 subtype was the most abundant, followed by SSTR3. The tumor residue increased in five of 26 patients (19%) in the treated group and in seven of 13 controls (53%). Visual field and pituitary function did not change in any patient. This study indicates that SSTR5 and SSTR3 are the most frequently expressed SSTR subtypes in NFPAs and supports a potential role of SSTR subtypes in stabilization of tumor remnant from NFPAs.
Collapse
Affiliation(s)
- Alessandra Fusco
- Division of Endocrinology, School of Medicine, Catholic University, Largo A. Gemelli 8, 00168 Rome, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Li SC, Martijn C, Cui T, Essaghir A, Luque RM, Demoulin JB, Castaño JP, Öberg K, Giandomenico V. The somatostatin analogue octreotide inhibits growth of small intestine neuroendocrine tumour cells. PLoS One 2012; 7:e48411. [PMID: 23119007 PMCID: PMC3485222 DOI: 10.1371/journal.pone.0048411] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Accepted: 10/01/2012] [Indexed: 01/02/2023] Open
Abstract
Octreotide is a widely used synthetic somatostatin analogue that significantly improves the management of neuroendocrine tumours (NETs). Octreotide acts through somatostatin receptors (SSTRs). However, the molecular mechanisms leading to successful disease control or symptom management, especially when SSTRs levels are low, are largely unknown. We provide novel insights into how octreotide controls NET cells. CNDT2.5 cells were treated from 1 day up to 16 months with octreotide and then were profiled using Affymetrix microarray analysis. Quantitative real-time PCR and western blot analyses were used to validate microarray profiling in silico data. WST-1 cell proliferation assay was applied to evaluate cell growth of CNDT2.5 cells in the presence or absence of 1 µM octreotide at different time points. Moreover, laser capture microdissected tumour cells and paraffin embedded tissue slides from SI-NETs at different stages of disease were used to identify transcriptional and translational expression. Microarrays analyses did not reveal relevant changes in SSTR expression levels. Unexpectedly, six novel genes were found to be upregulated by octreotide: annexin A1 (ANXA1), rho GTPase-activating protein 18 (ARHGAP18), epithelial membrane protein 1 (EMP1), growth/differentiation factor 15 (GDF15), TGF-beta type II receptor (TGFBR2) and tumour necrosis factor (ligand) superfamily member 15 (TNFSF15). Furthermore, these novel genes were expressed in tumour tissues at transcript and protein levels. We suggest that octreotide may use a potential novel framework to exert its beneficial effect as a drug and to convey its action on neuroendocrine cells. Thus, six novel genes may regulate cell growth and differentiation in normal and tumour neuroendocrine cells and have a role in a novel octreotide mechanism system.
Collapse
Affiliation(s)
- Su-Chen Li
- Department of Medical Sciences, Endocrine Oncology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Cécile Martijn
- Department of Surgical Sciences, Anaesthesiology & Intensive Care, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Tao Cui
- Department of Medical Sciences, Endocrine Oncology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Ahmed Essaghir
- Université Catholique de Louvain, de Duve Institute, Brussels, Belgium
| | - Raúl M. Luque
- Department of Cell Biology, Physiology, and Immunology, Instituto Maimónides de Investigación Biomédica (IMIBIC), Hospital Universitario Reina Sofia, University of Cordoba, and CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Cordoba, Spain
| | | | - Justo P. Castaño
- Department of Cell Biology, Physiology, and Immunology, Instituto Maimónides de Investigación Biomédica (IMIBIC), Hospital Universitario Reina Sofia, University of Cordoba, and CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Cordoba, Spain
| | - Kjell Öberg
- Department of Medical Sciences, Endocrine Oncology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
- Centre of Excellence for Endocrine Tumours, Uppsala University Hospital, Uppsala, Sweden
| | - Valeria Giandomenico
- Department of Medical Sciences, Endocrine Oncology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
- * E-mail:
| |
Collapse
|
25
|
Shpakov AO. Somatostatin receptors and signaling cascades coupled to them. J EVOL BIOCHEM PHYS+ 2012. [DOI: 10.1134/s0022093012040020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
26
|
Zhou G, Liu SH, Shahi KM, Wang H, Duan X, Lin X, Feng XH, Li M, Fisher WE, Demayo FJ, Dawson D, Brunicardi FC. Negative regulation of pancreatic and duodenal homeobox-1 by somatostatin receptor subtype 5. Mol Endocrinol 2012; 26:1225-34. [PMID: 22669743 DOI: 10.1210/me.2012-1095] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Somatostatin receptor subtype 5 (SSTR5) mediates the inhibitory effect of somatostatin and its analogs on insulin expression/secretion and islet cell proliferation. We provide biochemical and genetic evidence that SSTR5 exerted its physiological actions via down-regulating pancreatic and duodenal homeobox-1 (PDX-1), a β-cell-specific homeodomain-containing transcription factor. Cotransfection of SSTR5 with PDX-1 resulted in dose-dependent inhibition of PDX-1 expression in human embryonic kidney 293 cells. SSTR5 agonist RPL-1980 inhibited PDX-1 expression and abolished glucagon-like peptide 1-stimulated PDX-1 expression in mouse insulinoma β-TC-6 cells. SSTR5 knockdown by short hairpin RNA led to increased PDX-1 expression that was accompanied by enhanced insulin secretion stimulated by high glucose in β-TC6 cells and alternated expressions of cell cycle proteins that favor cell proliferation in mouse insulinoma MIN6 cells. Quantitative RT-PCR analysis showed that cotransfected SSTR5 inhibited PDX-1 mRNA expression, whereas knockdown of SSTR5 increased PDX-1 mRNA expression. In addition, we found that cotransfected wild-type SSTR5 increased PDX-1 ubiquitination in human embryonic kidney 293 cells, whereas SSTR5 P335L, a hypofunctional single nucleotide polymorphism of SSTR5, inhibited PDX-1 ubiquitination. SSTR5 knockout resulted in increased expression of PDX-1, insulin, and proliferating cell nuclear antigen in the islets of sstr(-/-) mice. Immunohistochemistry analysis showed that SSTR5 P335L was associated with elevated expression of PDX-1 in human pancreatic neuroendocrine tumor. Taken together, our studies demonstrated that SSTR5 is a negative regulator for PDX-1 expression and that SSTR5 may mediate the inhibitory effects of somatostatin and its analogs on insulin expression/secretion and cell proliferation via down-regulating PDX-1 at both transcriptional and posttranslational levels.
Collapse
Affiliation(s)
- Guisheng Zhou
- Department of Surgery, David Geffen School of Medicine at University of California, Los Angeles, California 90095, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Lupp A, Nagel F, Doll C, Röcken C, Evert M, Mawrin C, Saeger W, Schulz S. Reassessment of sst3 somatostatin receptor expression in human normal and neoplastic tissues using the novel rabbit monoclonal antibody UMB-5. Neuroendocrinology 2012; 96:301-10. [PMID: 22414742 DOI: 10.1159/000337659] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2011] [Accepted: 02/28/2012] [Indexed: 11/19/2022]
Abstract
BACKGROUND Among the five somatostatin receptors (sst(1)-sst(5)), the sst(3) receptor displays a distinct pharmacological profile. Like sst(2), the sst(3) receptor efficiently internalizes radiolabeled somatostatin analogs. Unlike sst(2), however, internalized sst(3) receptors are rapidly transferred to lysosomes for degradation. Apart from this, very little is known about the clinical relevance of the sst(3) receptor, which may in part be due to the lack of specific monoclonal sst(3) antibodies. METHODS Here, we have extensively characterized the novel rabbit monoclonal anti-human sst(3) antibody UMB-5 using transfected cells and receptor-expressing tissues. UMB-5 was then subjected to immunohistochemical staining of a series of 190 formalin-fixed, paraffin-embedded normal and neoplastic human tissues. RESULTS Specificity of UMB-5 was demonstrated by detection of a broad band migrating at a molecular weight of 70,000-85,000 in immunoblots from human pituitary. After enzymatic deglycosylation, the size of this band decreased to a molecular weight of 45,000. Tissue immunostaining was completely abolished by pre-adsorption of UMB-5 with its immunizing peptide. In addition, UMB-5 detected distinct cell populations in human tissues like pancreatic islands, anterior pituitary, adrenal cortex, adrenal medulla, and enteric ganglia, similar to that seen with a rabbit polyclonal antibody generated against a different carboxyl-terminal epitope of the sst(3) receptor. In a comparative immunohistochemical study, UMB-5 yielded predominant plasma membrane staining in the majority of pituitary adenomas, pheochromocytomas, and a subset of neuroendocrine tumors. The sst(3) receptor was also present in many glioblastomas, pancreatic, breast, cervix, and ovarian carcinomas. CONCLUSION The rabbit monoclonal antibody UMB-5 may prove of great value in the identification of sst(3)-expressing tumors during routine histopathological examinations. Given its unique trafficking properties, these tumors may be potential candidates for sst(3)-directed receptor radiotherapy.
Collapse
Affiliation(s)
- Amelie Lupp
- Department of Pharmacology and Toxicology, University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Pfisterer A, Eisele K, Chen X, Wagner M, Müllen K, Weil T. Bioactive unnatural somatostatin analogues through bioorthogonal iodo- and ethynyl-disulfide intercalators. Chemistry 2011; 17:9697-707. [PMID: 21748812 DOI: 10.1002/chem.201100287] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2011] [Revised: 04/26/2011] [Indexed: 11/09/2022]
Abstract
Iodo- and ethynyl-containing bisalkylating bioconjugation agents 5 and 8 were achieved and allow the introduction of reactive unnatural substituents into proteins and peptides whilst the bioactive 3D structure is retained. Derivatives of the peptide hormone somatostatin bearing a single iodo or ethynyl group were prepared through intercalation into the disulfide bridge. For the first time, the exact reaction mechanism of the intercalation was elucidated by applying 2D NMR experiments and it was shown that, during the reaction, somatostatin diastereomers were formed. Site-directed modification of the ethynyl-modified peptide with a coumarin chromophore was achieved through a [1,3] dipolar Huisgen cycloaddition reaction; this suggests that such a derivative could serve as an attractive platform to prepare artificial somatostatin compound libraries. The biological activity and specificity of a representative modified somatostatin derivative was demonstrated and efficient receptor-mediated cell uptake occurred in a dose-dependent manner into receptor positive cells only. The iodo and ethynyl bioconjugation reagents presented herein could be applied for introducing such substituents into alternative peptides and proteins and, in principle, could facilitate the efficient design of a broad variety of artificial protein and peptide analogues with previously unknown bioactivities.
Collapse
Affiliation(s)
- Anne Pfisterer
- Max Planck Institute for Polymer Research, Mainz, Germany
| | | | | | | | | | | |
Collapse
|
29
|
Ghassemi AH, van Steenbergen MJ, Barendregt A, Talsma H, Kok RJ, van Nostrum CF, Crommelin DJA, Hennink WE. Controlled release of octreotide and assessment of peptide acylation from poly(D,L-lactide-co-hydroxymethyl glycolide) compared to PLGA microspheres. Pharm Res 2011; 29:110-20. [PMID: 21744173 PMCID: PMC3246586 DOI: 10.1007/s11095-011-0517-3] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Accepted: 06/15/2011] [Indexed: 11/24/2022]
Abstract
Purpose To investigate the in vitro release of octreotide acetate, a somatostatin agonist, from microspheres based on a hydrophilic polyester, poly(D,L-lactide-co-hydroxymethyl glycolide) (PLHMGA). Methods Spherical and non-porous octreotide-loaded PLHMGA microspheres (12 to 16 μm) and loading efficiency of 60–70% were prepared by a solvent evaporation. Octreotide release profiles were compared with commercial PLGA formulation (Sandostatin LAR®); possible peptide modification with lactic, glycolic and hydroxymethyl glycolic acid units was monitored. Results PLHMGA microspheres showed burst release (~20%) followed by sustained release for 20–60 days, depending on the hydrophilicity of the polymer. Percentage of released loaded peptide was high (70–90%); > 60% of released peptide was native octreotide. PLGA microspheres did not show peptide release for the first 10 days, after which it was released in a sustained manner over the next 90 days; > 75% of released peptides were acylated adducts. Conclusions PLHMGA microspheres are promising controlled systems for peptides with excellent control over release kinetics. Moreover, substantially less peptide modification occurred in PLHMGA than in PLGA microspheres. Electronic Supplementary Material The online version of this article (doi:10.1007/s11095-011-0517-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Amir H Ghassemi
- Department of Pharmaceutics Utrecht Institute for Pharmaceutical Sciences, Utrecht University, P.O. Box 80082, 3508 TB Utrecht, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Kao YJ, Ghosh M, Schonbrunn A. Ligand-dependent mechanisms of sst2A receptor trafficking: role of site-specific phosphorylation and receptor activation in the actions of biased somatostatin agonists. Mol Endocrinol 2011; 25:1040-54. [PMID: 21493671 DOI: 10.1210/me.2010-0398] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The somatostatin receptor subtype 2A (sst2A) mediates many of somatostatin's neuroendocrine actions and is the primary therapeutic target for the stable somatostatin analogs used to inhibit hormone secretion by pituitary and gastroenteropancreatic tumors. Two new multireceptor targeting somatostatin analogs currently under clinical investigation, the multisomatostatin receptor agonist cyclo-[diaminoethylcarbamoyl-HydroxyPro-Phenylglycine-D-Trp-Lys-(4-O-benzyl)Tyr-Phe] (SOM230) (Pasireotide) and pan-somatostatin receptor agonist Tyr-cyclo-[D-diaminobutyric acid-Arg-Phe-Phe-D-Trp-Lys-Thr-Phe] (KE108), behave as functionally selective ligands at the sst2A receptor, mimicking some of somatostatin's actions but antagonizing others. Further, SOM230 and KE108 are less able to induce receptor internalization than somatostatin, indicating that they exhibit functional selectivity for receptor regulation as well as signaling. Here, we identify agonist-specific differences in the molecular events regulating sst2A receptor endocytosis. SOM230 and KE108 were less potent and less effective than somatostatin at stimulating sst2A receptor phosphorylation at two pairs of residues, Ser341/343 and Thr353/354. Only the pattern of Thr353/354 phosphorylation correlated with receptor internalization, consistent with the known importance of Thr phosphorylation for sst2A receptor endocytosis. As expected, arrestin recruitment to membrane receptors was reduced with SOM230 and KE108. In addition, both receptor dephosphorylation and receptor recycling occurred more rapidly with SOM230 and KE108 than with somatostatin. Surprisingly, however, SOM230 and KE108 also altered sst2A internalization in a phosphorylation-independent manner, because these analogs were less effective than somatostatin at stimulating the endocytosis of a phosphorylation-negative receptor mutant. These results show that the decreased receptor internalization produced by SOM230 and KE108 compared with somatostatin result from phosphorylation-independent effects as well as reduced site-specific receptor phosphorylation and receptor-arrestin association.
Collapse
Affiliation(s)
- Yachu J Kao
- Department of Integrative Biology and Pharmacology, University of Texas, Health Science Center-Houston, Houston, Texas 77030, USA
| | | | | |
Collapse
|
31
|
Lupp A, Hunder A, Petrich A, Nagel F, Doll C, Schulz S. Reassessment of sst(5) somatostatin receptor expression in normal and neoplastic human tissues using the novel rabbit monoclonal antibody UMB-4. Neuroendocrinology 2011; 94:255-64. [PMID: 21952553 DOI: 10.1159/000329876] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2011] [Accepted: 05/31/2011] [Indexed: 11/19/2022]
Abstract
OBJECTIVE The frequent overexpression of somatostatin receptors (sst) in neuroendocrine tumors provides the molecular basis for the diagnostic and therapeutic application of stable somatostatin analogs. Whereas octreotide acts mainly via the sst(2) receptor, the novel pan-somatostatin analog pasireotide exhibits particular high affinity for the sst(5) receptor. To determine whether a patient is a candidate for octreotide or pasireotide therapy, it is important to evaluate the somatostatin receptor status. However, so far highly specific rabbit monoclonal antibodies have been developed for the sst(2) receptor only (clone UMB-1). METHODS Here, we have extensively characterized a novel rabbit monoclonal antibody for the human sst(5) receptor (clone UMB-4). In a comparative immunohistochemical study, the expression of sst(5) and sst(2) receptors was assessed using UMB-4 and UMB-1, respectively. RESULTS Western blot experiments unequivocally demonstrated that UMB-4 selectively detected its cognate sst(5) receptor and did not cross-react with other proteins present in crude tissue homogenates. UMB-4 yielded a highly effective immunostaining of distinct cell populations in formalin-fixed, paraffin-embedded human tissues with a predominance of plasma membrane staining. In the pituitary, sst(5) was present on all growth hormone (GH)- and adrenocorticotropin hormone (ACTH)-producing cells whereas sst(2) was only observed on a subpopulation of GH-positive cells. Consequently, sst(5) was detectable on the majority of GH and ACTH adenomas. In contrast, sst(2) was only seen on GH but not on ACTH adenomas. CONCLUSIONS The rabbit monoclonal antibodies UMB-4 and UMB-1 will facilitate the assessment of the somatostatin receptor status of human tumors during routine histopathological examinations.
Collapse
Affiliation(s)
- Amelie Lupp
- Department of Pharmacology and Toxicology, University Hospital, Friedrich Schiller University, Jena, Germany
| | | | | | | | | | | |
Collapse
|