1
|
Vargas-Ortega G, Gonzalez-Virla B, Romero-Gameros CA. Pharmacological Treatment of Non-Functioning Pituitary Adenomas. Arch Med Res 2023; 54:102917. [PMID: 37996269 DOI: 10.1016/j.arcmed.2023.102917] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 10/24/2023] [Accepted: 11/09/2023] [Indexed: 11/25/2023]
Abstract
Treatment strategies for NFPA include surgery, radiotherapy, medical treatment, or follow-up. The treatment of NFPAs with compressive symptoms is surgical. However, in case of post-surgical tumor remnants, there may be treatment strategies that include observation and radiotherapy. Recently, medical treatment with cabergoline (CAB) has been recommended to contain and/or reduce the size of the tumor remnant. Based on the findings that many NFPAs show a dopamine receptor (DR) and somatostatin receptor (SR) expression, medical therapy with dopamine agonists (DAs) and somatostatin receptor ligands (SSRLs) has been tested as an alternative to prevent recurrence after surgery. The DAs have been the most extensively studied, showing some potential in terms of tumor shrinkage. SSRLs and other emerging medical options are much less studied. We will review and critically evaluate the current data on the medical therapy of NFPAs to elucidate their role in the management of this tumor type. In the case of actively growing remnants (more than 10% growth per year) and high-risk pituitary adenomas, treatment with CAB at a dose of 1.5-3.0 mg is indicated for tumor containment and/or reduction. In relation to combined chemotherapy with CAB, there is little information in the literature to support its use. In our experience, CAB treatment can be used after radiotherapy as an adjuvant treatment.
Collapse
Affiliation(s)
- Guadalupe Vargas-Ortega
- Endocrinology Service, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico.
| | - Baldomero Gonzalez-Virla
- Endocrinology Service, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Carlos Alfonso Romero-Gameros
- Otorhinolaryngology Service, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| |
Collapse
|
2
|
Sosa LDV, Picech F, Perez P, Gutierrez S, Leal RB, De Paul A, Torres A, Petiti JP. Regulation of FGF2-induced proliferation by inhibitory GPCR in normal pituitary cells. Front Endocrinol (Lausanne) 2023; 14:1183151. [PMID: 37576961 PMCID: PMC10414184 DOI: 10.3389/fendo.2023.1183151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 07/13/2023] [Indexed: 08/15/2023] Open
Abstract
Introduction Intracellular communication is essential for the maintenance of the anterior pituitary gland plasticity. The aim of this study was to evaluate whether GPCR-Gαi modulates basic fibroblast growth factor (FGF2)-induced proliferative activity in normal pituitary cell populations. Methods Anterior pituitary primary cell cultures from Wistar female rats were treated with FGF2 (10ng/mL) or somatostatin analog (SSTa, 100nM) alone or co-incubated with or without the inhibitors of GPCR-Gαi, pertussis toxin (PTX, 500nM), MEK inhibitor (U0126, 100µM) or PI3K inhibitor (LY 294002, 10 μM). Results FGF2 increased and SSTa decreased the lactotroph and somatotroph BrdU uptak2e (p<0.05) whereas the FGF2-induced S-phase entry was prevented by SSTa co-incubation in both cell types, with these effects being reverted by PTX, U0126 or LY294002 pre-incubation. The inhibition of lactotroph and somatotroph mitosis was associated with a downregulation of c-Jun expression, a decrease of phosphorylated (p) ERK and pAKT. Furthermore, SSTa was observed to inhibit the S-phase entry induced by FGF2, resulting in a further increase in the number of cells in the G1 phase and a concomitant reduction in the number of cells in the S phases (p< 0.05), effects related to a decrease of cyclin D1 expression and an increase in the expression of the cell cycle inhibitors p27 and p21. Discussion In summary, the GPCR-Gαi activated by SSTa blocked the pro-proliferative effect of FGF2 in normal pituitary cells via a MEK-dependent mechanism, which acts as a mediator of both anti and pro-mitogenic signals, that may regulate the principal effectors of the G1 to S-phase transition.
Collapse
Affiliation(s)
- Liliana del V. Sosa
- Universidad Nacional de Córdoba, Facultad de Ciencias Médicas, Centro de Microscopía Electrónica, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas Técnicas (CONICET), Instituto de Investigaciones en Ciencias de la Salud (INICSA), Córdoba, Argentina
| | - Florencia Picech
- Universidad Nacional de Córdoba, Facultad de Ciencias Médicas, Centro de Microscopía Electrónica, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas Técnicas (CONICET), Instituto de Investigaciones en Ciencias de la Salud (INICSA), Córdoba, Argentina
| | - Pablo Perez
- Universidad Nacional de Córdoba, Facultad de Ciencias Médicas, Centro de Microscopía Electrónica, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas Técnicas (CONICET), Instituto de Investigaciones en Ciencias de la Salud (INICSA), Córdoba, Argentina
| | - Silvina Gutierrez
- Universidad Nacional de Córdoba, Facultad de Ciencias Médicas, Centro de Microscopía Electrónica, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas Técnicas (CONICET), Instituto de Investigaciones en Ciencias de la Salud (INICSA), Córdoba, Argentina
| | - Rodrigo Bainy Leal
- Universidade Federal de Santa Catarina, Florianópolis, Departamento de Bioquímica e Programa de Pós-graduação em Bioquímica, Centro de Ciências Biológicas, Santa Catarina, Brazil
| | - Ana De Paul
- Universidad Nacional de Córdoba, Facultad de Ciencias Médicas, Centro de Microscopía Electrónica, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas Técnicas (CONICET), Instituto de Investigaciones en Ciencias de la Salud (INICSA), Córdoba, Argentina
| | - Alicia Torres
- Universidad Nacional de Córdoba, Facultad de Ciencias Médicas, Centro de Microscopía Electrónica, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas Técnicas (CONICET), Instituto de Investigaciones en Ciencias de la Salud (INICSA), Córdoba, Argentina
| | - Juan Pablo Petiti
- Universidad Nacional de Córdoba, Facultad de Ciencias Médicas, Centro de Microscopía Electrónica, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas Técnicas (CONICET), Instituto de Investigaciones en Ciencias de la Salud (INICSA), Córdoba, Argentina
| |
Collapse
|
3
|
Modena D, Moras ML, Sandrone G, Stevenazzi A, Vergani B, Dasgupta P, Kliever A, Gulde S, Marangelo A, Schillmaier M, Luque RM, Bäuerle S, Pellegata NS, Schulz S, Steinkühler C. Identification of a Novel SSTR3 Full Agonist for the Treatment of Nonfunctioning Pituitary Adenomas. Cancers (Basel) 2023; 15:3453. [PMID: 37444563 DOI: 10.3390/cancers15133453] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 07/15/2023] Open
Abstract
Somatostatin receptor (SSTR) agonists have been extensively used for treating neuroendocrine tumors. Synthetic therapeutic agonists showing selectivity for SSTR2 (Octreotide) or for SSTR2 and SSTR5 (Pasireotide) have been approved for the treatment of patients with acromegaly and Cushing's syndrome, as their pituitary tumors highly express SSTR2 or SSTR2/SSTR5, respectively. Nonfunctioning pituitary adenomas (NFPAs), which express high levels of SSTR3 and show only modest response to currently available SSTR agonists, are often invasive and cannot be completely resected, and therefore easily recur. The aim of the present study was the evaluation of ITF2984, a somatostatin analog and full SSTR3 agonist, as a new potential treatment for NFPAs. ITF2984 shows a 10-fold improved affinity for SSTR3 compared to Octreotide or Pasireotide. Molecular modeling and NMR studies indicated that the higher affinity for SSTR3 correlates with a higher stability of a distorted β-I turn in the cyclic peptide backbone. ITF2984 induces receptor internalization and phosphorylation, and triggers G-protein signaling at pharmacologically relevant concentrations. Furthermore, ITF2984 displays antitumor activity that is dependent on SSTR3 expression levels in the MENX (homozygous mutant) NFPA rat model, which closely recapitulates human disease. Therefore, ITF2984 may represent a novel therapeutic option for patients affected by NFPA.
Collapse
Affiliation(s)
- Daniela Modena
- Preclinical R&D, Italfarmaco Group, 20092 Cinisello Balsamo, Milan, Italy
| | - Maria Luisa Moras
- Preclinical R&D, Italfarmaco Group, 20092 Cinisello Balsamo, Milan, Italy
| | - Giovanni Sandrone
- Preclinical R&D, Italfarmaco Group, 20092 Cinisello Balsamo, Milan, Italy
| | - Andrea Stevenazzi
- Preclinical R&D, Italfarmaco Group, 20092 Cinisello Balsamo, Milan, Italy
| | - Barbara Vergani
- Preclinical R&D, Italfarmaco Group, 20092 Cinisello Balsamo, Milan, Italy
| | - Pooja Dasgupta
- Institute of Pharmacology and Toxicology, Universitätsklinikum Jena, Friedrich-Schiller-Universität, 07747 Jena, Germany
| | - Andrea Kliever
- Institute of Pharmacology and Toxicology, Universitätsklinikum Jena, Friedrich-Schiller-Universität, 07747 Jena, Germany
| | - Sebastian Gulde
- Institute for Diabetes and Cancer, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Alessandro Marangelo
- Institute for Diabetes and Cancer, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Heidelberg University Hospital, 69120 Heidelberg, Germany
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, 27100 Pavia, Italy
| | - Mathias Schillmaier
- Department of Nuclear Medicine, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, 80333 Munich, Germany
- Department of Diagnostic and Interventional Radiology, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, 80333 Munich, Germany
| | - Raul M Luque
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004 Cordoba, Spain
- Reina Sofia University Hospital (HURS), 14004 Cordoba, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004 Cordoba, Spain
| | - Stephen Bäuerle
- Department of Mathematics, Technical University Munich, 85748 Garching, Germany
| | - Natalia S Pellegata
- Institute for Diabetes and Cancer, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Heidelberg University Hospital, 69120 Heidelberg, Germany
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, 27100 Pavia, Italy
| | - Stefan Schulz
- Institute of Pharmacology and Toxicology, Universitätsklinikum Jena, Friedrich-Schiller-Universität, 07747 Jena, Germany
| | | |
Collapse
|
4
|
Molecular basis for the selective G protein signaling of somatostatin receptors. Nat Chem Biol 2023; 19:133-140. [PMID: 36138141 DOI: 10.1038/s41589-022-01130-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 08/01/2022] [Indexed: 02/04/2023]
Abstract
G protein-coupled receptors (GPCRs) modulate every aspect of physiological functions mainly through activating heterotrimeric G proteins. A majority of GPCRs promiscuously couple to multiple G protein subtypes. Here we validate that in addition to the well-known Gi/o pathway, somatostatin receptor 2 and 5 (SSTR2 and SSTR5) couple to the Gq/11 pathway and show that smaller ligands preferentially activate the Gi/o pathway. We further determined cryo-electron microscopy structures of the SSTR2‒Go and SSTR2‒Gq complexes bound to octreotide and SST-14. Structural and functional analysis revealed that G protein selectivity of SSTRs is not only determined by structural elements in the receptor-G protein interface, but also by the conformation of the agonist-binding pocket. Accordingly, smaller ligands fail to stabilize a broader agonist-binding pocket of SSTRs that is required for efficient Gq/11 coupling but not Gi/o coupling. Our studies facilitate the design of drugs with selective G protein signaling to improve therapeutic efficacy.
Collapse
|
5
|
An Overview of Pituitary Neuroendocrine Tumors (PitNET) and Algorithmic Approach to Diagnosis. Adv Anat Pathol 2023; 30:79-83. [PMID: 36069849 DOI: 10.1097/pap.0000000000000367] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The diagnostic algorithm and nomenclature of pituitary neuroendocrine tumors have evolved over the past decade, beginning with simpler categorical schemes focused on histomorphologic features and moving to a more sophisticated lineage-specific categorization. This contemporary overview highlights a multimodal approach to pituitary neuroendocrine tumors with a focus on changes in nomenclature, classification, and subclassification; including, brief comments on treatment, and new guidelines for genetic screening, particularly for young patients with such neoplasms.
Collapse
|
6
|
Bo Q, Yang F, Li Y, Meng X, Zhang H, Zhou Y, Ling S, Sun D, Lv P, Liu L, Shi P, Tian C. Structural insights into the activation of somatostatin receptor 2 by cyclic SST analogues. Cell Discov 2022; 8:47. [PMID: 35595746 PMCID: PMC9122944 DOI: 10.1038/s41421-022-00405-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 03/23/2022] [Indexed: 11/09/2022] Open
Abstract
The endogenous cyclic tetradecapeptide SST14 was reported to stimulate all five somatostatin receptors (SSTR1-5) for hormone release, neurotransmission, cell growth arrest and cancer suppression. Two SST14-derived short cyclic SST analogues (lanreotide or octreotide) with improved stability and longer lifetime were developed as drugs to preferentially activate SSTR2 and treat acromegalia and neuroendocrine tumors. Here, cryo-EM structures of the human SSTR2-Gi complex bound with SST14, octreotide or lanreotide were determined at resolutions of 2.85 Å, 2.97 Å, and 2.87 Å, respectively. Structural and functional analysis revealed that interactions between β-turn residues in SST analogues and transmembrane SSTR2 residues in the ligand-binding pocket are crucial for receptor binding and functional stimulation of the two SST14-derived cyclic octapeptides. Additionally, Q1022.63, N2766.55, and F2947.35 could be responsible for the selectivity of lanreotide or octreotide for SSTR2 over SSTR1 or SSTR4. These results provide valuable insights into further rational development of SST analogue drugs targeting SSTR2.
Collapse
Affiliation(s)
- Qing Bo
- Department of Chemistry and the First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, Joint Center for Biological Analytical Chemistry, Anhui Engineering Laboratory of Peptide Drug, Anhui Laboratory of Advanced Photonic Science and Technology, University of Science and Technology of China, Hefei, Anhui, China
| | - Fan Yang
- Department of Chemistry and the First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, Joint Center for Biological Analytical Chemistry, Anhui Engineering Laboratory of Peptide Drug, Anhui Laboratory of Advanced Photonic Science and Technology, University of Science and Technology of China, Hefei, Anhui, China
| | - Yingge Li
- Department of Chemistry and the First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, Joint Center for Biological Analytical Chemistry, Anhui Engineering Laboratory of Peptide Drug, Anhui Laboratory of Advanced Photonic Science and Technology, University of Science and Technology of China, Hefei, Anhui, China
| | - Xianyu Meng
- Department of Chemistry and the First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, Joint Center for Biological Analytical Chemistry, Anhui Engineering Laboratory of Peptide Drug, Anhui Laboratory of Advanced Photonic Science and Technology, University of Science and Technology of China, Hefei, Anhui, China
| | - Huanhuan Zhang
- Department of Chemistry and the First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, Joint Center for Biological Analytical Chemistry, Anhui Engineering Laboratory of Peptide Drug, Anhui Laboratory of Advanced Photonic Science and Technology, University of Science and Technology of China, Hefei, Anhui, China
| | - Yingxin Zhou
- Department of Chemistry and the First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, Joint Center for Biological Analytical Chemistry, Anhui Engineering Laboratory of Peptide Drug, Anhui Laboratory of Advanced Photonic Science and Technology, University of Science and Technology of China, Hefei, Anhui, China
| | - Shenglong Ling
- Department of Chemistry and the First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, Joint Center for Biological Analytical Chemistry, Anhui Engineering Laboratory of Peptide Drug, Anhui Laboratory of Advanced Photonic Science and Technology, University of Science and Technology of China, Hefei, Anhui, China
| | - Demeng Sun
- Department of Chemistry and the First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, Joint Center for Biological Analytical Chemistry, Anhui Engineering Laboratory of Peptide Drug, Anhui Laboratory of Advanced Photonic Science and Technology, University of Science and Technology of China, Hefei, Anhui, China
| | - Pei Lv
- Department of Chemistry and the First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, Joint Center for Biological Analytical Chemistry, Anhui Engineering Laboratory of Peptide Drug, Anhui Laboratory of Advanced Photonic Science and Technology, University of Science and Technology of China, Hefei, Anhui, China
| | - Lei Liu
- Tsinghua-Peking Joint Center for Life Sciences, Ministry of Education Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Department of Chemistry, Tsinghua University, Beijing, China.
| | - Pan Shi
- Department of Chemistry and the First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, Joint Center for Biological Analytical Chemistry, Anhui Engineering Laboratory of Peptide Drug, Anhui Laboratory of Advanced Photonic Science and Technology, University of Science and Technology of China, Hefei, Anhui, China.
| | - Changlin Tian
- Department of Chemistry and the First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, Joint Center for Biological Analytical Chemistry, Anhui Engineering Laboratory of Peptide Drug, Anhui Laboratory of Advanced Photonic Science and Technology, University of Science and Technology of China, Hefei, Anhui, China.
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Hefei, Anhui, China.
| |
Collapse
|
7
|
Iida H, Komagata T, Tanaka H, Nagasawa R, Nishio T, Shono T, Kitagawa J, Ogawara KI, Shinozaki K, Seki A, Bruce M, Ohno T. Novel Platform for Predicting Drug Effects in Patients with Acromegaly: Translational Exposure-Response Evaluation of Growth Hormone-Inhibitory Effect of Octreotide after Growth Hormone-Releasing Hormone Stimulation. J Pharmacol Exp Ther 2021; 379:400-408. [PMID: 34599040 DOI: 10.1124/jpet.121.000769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 09/29/2021] [Indexed: 11/22/2022] Open
Abstract
Acromegaly is a chronic systemic disease characterized by facial and peripheral changes caused by soft tissue overgrowth and is associated with multiple comorbidities. Despite available surgical and medical therapies, suitable treatments for acromegaly are still lacking. Efficient drug development requires an understanding of the exposure-response (E-R) relationship based on nonclinical and early clinical studies. We aimed to establish a platform to facilitate the development of novel drugs to treat acromegaly. We evaluated the E-R relationship of the growth hormone (GH)-inhibitory effect of the somatostatin analog octreotide under growth hormone-releasing hormone + arginine stimulation in healthy participants and compared the results with historical data for patients with acromegaly. This randomized five-way crossover study included two placebo and three active-treatment periods with different doses of octreotide acetate. GH secretion in the two placebo periods was comparable, which confirmed the reproducibility of the response with no carryover effect. GH secretion was inhibited by low-, medium-, and high-dose octreotide acetate in a dose-dependent manner. We also examined the E-R relationship in monkeys as a preclinical drug evaluation study and in rats as a more convenient and simple system for screening candidate drugs. The E-R relationships and EC50 values were similar among animals, healthy participants, and patients with acromegaly, which suggests that GH stimulation studies in early research and development allowed simulation of the drug response in patients with acromegaly. SIGNIFICANCE STATEMENT: This study demonstrated similar exposure-response relationships in terms of the growth hormone-inhibitory effect of octreotide after growth hormone-releasing hormone stimulation among healthy participants, monkeys, and rats. The research methods and analyses utilized in this study will be useful for simulating the dosages and therapeutic effects of drugs for acromegaly and will facilitate the research and development of novel therapeutic agents with similar modes of action.
Collapse
Affiliation(s)
- Hiroyuki Iida
- Ono Pharmaceutical Co., Ltd., Osaka, Japan (H.I., T.K., H.T., R.N., T.N., T.S., J.K., K.S., A.S., T.O.); Laboratory of Pharmaceutics, Kobe Pharmaceutical University, Higashinada-ku, Kobe, Japan (H.I., K.O.); and Ono Pharma UK Ltd., London, United Kingdom (M.B.)
| | - Tatsuya Komagata
- Ono Pharmaceutical Co., Ltd., Osaka, Japan (H.I., T.K., H.T., R.N., T.N., T.S., J.K., K.S., A.S., T.O.); Laboratory of Pharmaceutics, Kobe Pharmaceutical University, Higashinada-ku, Kobe, Japan (H.I., K.O.); and Ono Pharma UK Ltd., London, United Kingdom (M.B.)
| | - Hirotaka Tanaka
- Ono Pharmaceutical Co., Ltd., Osaka, Japan (H.I., T.K., H.T., R.N., T.N., T.S., J.K., K.S., A.S., T.O.); Laboratory of Pharmaceutics, Kobe Pharmaceutical University, Higashinada-ku, Kobe, Japan (H.I., K.O.); and Ono Pharma UK Ltd., London, United Kingdom (M.B.)
| | - Ryusuke Nagasawa
- Ono Pharmaceutical Co., Ltd., Osaka, Japan (H.I., T.K., H.T., R.N., T.N., T.S., J.K., K.S., A.S., T.O.); Laboratory of Pharmaceutics, Kobe Pharmaceutical University, Higashinada-ku, Kobe, Japan (H.I., K.O.); and Ono Pharma UK Ltd., London, United Kingdom (M.B.)
| | - Takuya Nishio
- Ono Pharmaceutical Co., Ltd., Osaka, Japan (H.I., T.K., H.T., R.N., T.N., T.S., J.K., K.S., A.S., T.O.); Laboratory of Pharmaceutics, Kobe Pharmaceutical University, Higashinada-ku, Kobe, Japan (H.I., K.O.); and Ono Pharma UK Ltd., London, United Kingdom (M.B.)
| | - Tomoyuki Shono
- Ono Pharmaceutical Co., Ltd., Osaka, Japan (H.I., T.K., H.T., R.N., T.N., T.S., J.K., K.S., A.S., T.O.); Laboratory of Pharmaceutics, Kobe Pharmaceutical University, Higashinada-ku, Kobe, Japan (H.I., K.O.); and Ono Pharma UK Ltd., London, United Kingdom (M.B.)
| | - Junsaku Kitagawa
- Ono Pharmaceutical Co., Ltd., Osaka, Japan (H.I., T.K., H.T., R.N., T.N., T.S., J.K., K.S., A.S., T.O.); Laboratory of Pharmaceutics, Kobe Pharmaceutical University, Higashinada-ku, Kobe, Japan (H.I., K.O.); and Ono Pharma UK Ltd., London, United Kingdom (M.B.)
| | - Ken-Ichi Ogawara
- Ono Pharmaceutical Co., Ltd., Osaka, Japan (H.I., T.K., H.T., R.N., T.N., T.S., J.K., K.S., A.S., T.O.); Laboratory of Pharmaceutics, Kobe Pharmaceutical University, Higashinada-ku, Kobe, Japan (H.I., K.O.); and Ono Pharma UK Ltd., London, United Kingdom (M.B.)
| | - Koji Shinozaki
- Ono Pharmaceutical Co., Ltd., Osaka, Japan (H.I., T.K., H.T., R.N., T.N., T.S., J.K., K.S., A.S., T.O.); Laboratory of Pharmaceutics, Kobe Pharmaceutical University, Higashinada-ku, Kobe, Japan (H.I., K.O.); and Ono Pharma UK Ltd., London, United Kingdom (M.B.)
| | - Akiteru Seki
- Ono Pharmaceutical Co., Ltd., Osaka, Japan (H.I., T.K., H.T., R.N., T.N., T.S., J.K., K.S., A.S., T.O.); Laboratory of Pharmaceutics, Kobe Pharmaceutical University, Higashinada-ku, Kobe, Japan (H.I., K.O.); and Ono Pharma UK Ltd., London, United Kingdom (M.B.)
| | - Mark Bruce
- Ono Pharmaceutical Co., Ltd., Osaka, Japan (H.I., T.K., H.T., R.N., T.N., T.S., J.K., K.S., A.S., T.O.); Laboratory of Pharmaceutics, Kobe Pharmaceutical University, Higashinada-ku, Kobe, Japan (H.I., K.O.); and Ono Pharma UK Ltd., London, United Kingdom (M.B.)
| | - Tomoya Ohno
- Ono Pharmaceutical Co., Ltd., Osaka, Japan (H.I., T.K., H.T., R.N., T.N., T.S., J.K., K.S., A.S., T.O.); Laboratory of Pharmaceutics, Kobe Pharmaceutical University, Higashinada-ku, Kobe, Japan (H.I., K.O.); and Ono Pharma UK Ltd., London, United Kingdom (M.B.)
| |
Collapse
|
8
|
Manosso KZB, Sampaio CL, Kasuki L, Antunes X, Gadelha MR, Boguszewski CL. GH and IGF-I levels and tumor shrinkage in response to first generation somatostatin receptor ligands in acromegaly: a comparative study between two reference centers for pituitary diseases in Brazil. Endocrine 2021; 74:146-154. [PMID: 34041695 DOI: 10.1007/s12020-021-02766-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 05/15/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE To compare biochemical and tumor response rates between two reference centers for pituitary diseases in Brazil after primary and adjuvant therapy with somatostatin receptor ligands (SRL) in acromegaly. METHODS Patients were classified as non-responders (NR), partial responders (PR), and full responders (FR) to 12-month SRL therapy according to: [criteria A] normal IGF-I and random GH (rGH) < 1 ng/mL (FR); ≥ 50% decrease of IGF-I and/or rGH (PR); < 50% decrease of IGF-I and rGH (NR); [criteria B] normal IGF-I (FR); ≥ 50% decrease of IGF-I (PR); < 50% decrease of IGF-I (NR). Tumor shrinkage <20% defined poor responders (tPR) and ≥ 20% good responders (tGR). RESULTS We studied 219 acromegaly patients (59% women, age 43.1 ± 13.9 years; 73 from Center I and 146 from Center II). After SRL therapy, the proportion of FR, PR, and NR by criteria A and B was 30.2 vs 49.1%, 52.8 vs 21.2% and 17 vs 29.7%, respectively (p < 0.001). Considering criteria A or B separately, there was no difference in the proportion of FR, PR and NR between two centers. However, when comparing criteria A and B, the Center I showed a difference of 30.9% in classification of FR in relation to 13.2% observed in Center II (p = 0.006). tGR were 51.4% of patients, with no differences between the centers. CONCLUSIONS IGF-I alone significantly increased positive response rates to SRLs, whereas the inclusion of rGH levels into therapeutic decision might lead to a significant increment on the costs of acromegaly management.
Collapse
Affiliation(s)
- Karina Zanlorenzi Basso Manosso
- SEMPR, Serviço de Endocrinologia e Metabologia do Hospital de Clínicas, Departamento de Clínica Médica, Universidade Federal do Paraná, Curitiba, Brazil
| | - Carolina Labigalini Sampaio
- SEMPR, Serviço de Endocrinologia e Metabologia do Hospital de Clínicas, Departamento de Clínica Médica, Universidade Federal do Paraná, Curitiba, Brazil
| | - Leandro Kasuki
- Centro de Pesquisa em Neuroendocrinologia, Divisão de Endocrinologia, Faculdade de Medicina e Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
- Serviço de Neuroendocrinologia, Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria Estadual de Saúde, Rio de Janeiro, RJ, Brazil
- Serviço de Endocrinologia, Hospital Federal de Bonsucesso, Rio de Janeiro, RJ, Brazil
| | - Ximene Antunes
- Centro de Pesquisa em Neuroendocrinologia, Divisão de Endocrinologia, Faculdade de Medicina e Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
- Serviço de Neuroendocrinologia, Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria Estadual de Saúde, Rio de Janeiro, RJ, Brazil
| | - Monica R Gadelha
- Centro de Pesquisa em Neuroendocrinologia, Divisão de Endocrinologia, Faculdade de Medicina e Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
- Serviço de Neuroendocrinologia, Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria Estadual de Saúde, Rio de Janeiro, RJ, Brazil
| | - Cesar Luiz Boguszewski
- SEMPR, Serviço de Endocrinologia e Metabologia do Hospital de Clínicas, Departamento de Clínica Médica, Universidade Federal do Paraná, Curitiba, Brazil.
| |
Collapse
|
9
|
Octreotide and Pasireotide Combination Treatment in Somatotroph Tumor Cells: Predominant Role of SST 2 in Mediating Ligand Effects. Cancers (Basel) 2021; 13:cancers13081816. [PMID: 33920241 PMCID: PMC8069349 DOI: 10.3390/cancers13081816] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 04/08/2021] [Indexed: 12/16/2022] Open
Abstract
Simple Summary First-generation somatostatin receptor ligands, such as octreotide, are the first-line medical therapy in acromegaly. Octreotide shows preferential binding for somatostatin receptor subtype 2 (SST2), while the second-generation ligand, pasireotide, has high affinity for multiple SSTs. We aimed to elucidate whether pasireotide acts via other receptors than SST2 in somatotroph tumors, and to investigate the potential role of the combination therapy octreotide plus pasireotide. We found that octreotide and pasireotide are superimposable in reducing GH secretion in cultured somatotroph tumor cells, as well as in inhibiting cell proliferation and intracellular pathway activity in rat GH4C1 cells (a model of somatotroph tumors). We did not find any additive/synergistic effect for the combination treatment. Furthermore, we observed that co-incubation with a SST2-selective antagonist reversed the inhibitory effect of both compounds. Therefore, the two drugs act mainly via SST2 in somatotroph tumor cells, and their combination is not superior to single agent treatment. Abstract First-generation somatostatin receptor ligands (fg-SRLs), such as octreotide (OCT), represent the first-line medical therapy in acromegaly. Fg-SRLs show a preferential binding affinity for somatostatin receptor subtype-2 (SST2), while the second-generation ligand, pasireotide (PAS), has high affinity for multiple SSTs (SST5 > SST2 > SST3 > SST1). Whether PAS acts via SST2 in somatotroph tumors, or through other SSTs (e.g., SST5), is a matter of debate. In this light, the combined treatment OCT+PAS could result in additive/synergistic effects. We evaluated the efficacy of OCT and PAS (alone and in combination) on growth hormone (GH) secretion in primary cultures from human somatotroph tumors, as well as on cell proliferation, intracellular signaling and receptor trafficking in the rat GH4C1 cell line. The results confirmed the superimposable efficacy of OCT and PAS in reducing GH secretion (primary cultures), cell proliferation, cAMP accumulation and intracellular [Ca2+] increase (GH4C1 cells), without any additive effect observed for OCT+PAS. In GH4C1 cells, co-incubation with a SST2-selective antagonist reversed the inhibitory effect of OCT and PAS on cell proliferation and cAMP accumulation, while both compounds resulted in a robust internalization of SST2 (but not SST5). In conclusion, OCT and PAS seem to act mainly through SST2 in somatotroph tumor cells in vitro, without inducing any additive/synergistic effect when tested in combination.
Collapse
|
10
|
Leonart LP, Riveros BS, Krahn MD, Pontarolo R. Pharmacological Acromegaly Treatment: Cost-Utility and Value of Information Analysis. Neuroendocrinology 2021; 111:388-402. [PMID: 32299084 DOI: 10.1159/000507890] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 04/14/2020] [Indexed: 11/19/2022]
Abstract
OBJECTIVES To conduct a cost-utility analysis comparing drug strategies involving octreotide, lanreotide, pasireotide, and pegvisomant for the treatment of patients with acromegaly who have failed surgery, from a Brazilian public payer perspective. METHODS A probabilistic cohort Markov model was developed. One-year cycles were employed. The patients started at 45 years of age and were followed lifelong. Costs, efficacy, and quality of life parameters were retrieved from the literature. A discount rate (5%) was applied to both costs and efficacy. The results were reported as costs per quality-adjusted life year (QALY), and incremental cost-effectiveness ratios (ICERs) were calculated when applicable. Scenario analyses considered alternative dosages, discount rate, tax exemption, and continued use of treatment despite lack of response. Value of information (VOI) analysis was conducted to explore uncertainty and to estimate the costs to be spent in future research. RESULTS Only lanreotide showed an ICER reasonable for having its use considered in clinical practice (R$ 112,138/US$ 28,389 per QALY compared to no treatment). Scenario analyses corroborated the base-case result. VOI analysis showed that much uncertainty surrounds the parameters, and future clinical research should cost less than R$ 43,230,000/US$ 10,944,304 per year. VOI also showed that almost all uncertainty that precludes an optimal strategy choice involves quality of life. CONCLUSIONS With current information, the only strategy that can be considered cost-effective in Brazil is lanreotide treatment. No second-line treatment is recommended. Significant uncertainty of parameters impairs optimal decision-making, and this conclusion can be generalized to other countries. Future research should focus on acquiring utility data.
Collapse
Affiliation(s)
- Leticia P Leonart
- Graduate Program in Pharmaceutical Sciences, Universidade Federal do Paraná, Curitiba, Brazil
| | - Bruno S Riveros
- Graduate Program in Pharmaceutical Sciences, Universidade Federal do Paraná, Curitiba, Brazil
| | - Murray D Krahn
- Toronto Health Economics and Technology Assessment Collaborative (THETA), Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, Toronto, Ontario, Canada
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Roberto Pontarolo
- Graduate Program in Pharmaceutical Sciences, Universidade Federal do Paraná, Curitiba, Brazil,
| |
Collapse
|
11
|
Gatto F, Arvigo M, Ferone D. Somatostatin receptor expression and patients' response to targeted medical treatment in pituitary tumors: evidences and controversies. J Endocrinol Invest 2020; 43:1543-1553. [PMID: 32557353 DOI: 10.1007/s40618-020-01335-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 06/11/2020] [Indexed: 12/23/2022]
Abstract
BACKGROUND Somatostatin receptors (SSTs) are widely co-expressed in pituitary tumors. SST2 and SST5 are the most represented SST subtypes. First-generation somatostatin receptor ligands (SRLs) mainly target SST2, while pasireotide, a multi-receptor ligand, shows high binding affinity for both SST5 and SST2. Therefore, SRLs are routinely used as medical treatment for GH-, TSH-, and ACTH-secreting pituitary tumors. METHODS Critical revision of literature data correlating SST expression with patients' response to SRLs. RESULTS SST2 expression in somatroph tumors directly correlates with GH and IGF-1 decrease after first-generation SRL treatment. SST2 immunohistochemistry represents a valuable tool to predict biochemical response to first-generation SRLs in acromegalic patients. Pasireotide seems to exert its biological effects via SST2 in unselected patients. However, in those subjects resistant to first-generation SRLs, harbouring tumors with negligible SST2 expression, pasireotide can act throughout SST5. More than somatotroph tumors, TSH-omas represent the paradigm of tumors showing a satisfactory response to SRLs. This is probably due to the high SST2 expression observed in nearly 100% of cases, as well as to the balanced amount of SST5. In corticotroph tumors, pasireotide mainly act via SST5, although there is a need for translational studies correlating its efficacy with SST expression in this peculiar tumor histotype. CONCLUSIONS The assumption "more target receptor, more drug efficacy" is not straightforward for SRLs. The complex pathophysiology of SSTs, and the technical challenges faced to translate research findings into clinical practice, still need our full commitment to make receptor evaluation a worthwhile procedure for individualizing treatment decisions.
Collapse
Affiliation(s)
- F Gatto
- Endocrinology Unit, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi, 10, 16132, Genoa, Italy.
- Endocrinology Unit, Department of Internal Medicine and Medical Specialties (DIMI), Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy.
| | - M Arvigo
- Endocrinology Unit, Department of Internal Medicine and Medical Specialties (DIMI), Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| | - D Ferone
- Endocrinology Unit, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi, 10, 16132, Genoa, Italy
- Endocrinology Unit, Department of Internal Medicine and Medical Specialties (DIMI), Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| |
Collapse
|
12
|
Singh S, Somvanshi RK, Panda V, Kumar U. Comparative distribution of somatostatin and somatostatin receptors in PTU-induced hypothyroidism. Endocrine 2020; 70:92-106. [PMID: 32335798 DOI: 10.1007/s12020-020-02309-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 04/06/2020] [Indexed: 12/21/2022]
Abstract
PURPOSE Propylthiouracil (PTU)-induced hypothyroidism is a well-established model for assessing hormonal and morphological changes in thyroid as well as other central and peripheral tissues. Somatostatin (SST) is known to regulate hormonal secretion and synthesis in endocrine tissues; however, nothing is currently known about the distribution of SST and its receptor in hypothyroidism. METHOD In the present study, the comparative immunohistochemical distribution of SST and somatostatin receptors (SSTRs) were analyzed in PTU-induced hypothyroid rats. Rats were treated with PTU for 15 days followed by a co-administration of levothyroxine (LVT) for 15 days. After PTU and LVT treatments (day 30), rats were further administered LVT alone for 15 more days (day 45). The subcellular distribution of SST and SSTR subtypes was determined by peroxidase immunohistochemistry in the thyroid gland collected from control and treated rats. RESULTS SST and SSTR subtypes were found to be moderately expressed in control thyroid tissues. SST and SSTR subtypes like immunoreactivity increased significantly in follicular and parafollicular epithelial cells in the thyroid of PTU-treated rats. The PTU-induced changes in the expression of SST and SSTR subtypes were suppressed by the administration of the LVT. In addition to thyroid tissues, SST and SSTRs expression was also changed in non-follicular tissues including blood vessels, smooth muscle cells, and connective tissue following treatments. CONCLUSION The present study revealed a distinct subcellular distribution of SST and SSTR subtypes in the thyroid and provides a new insight for the role of SST and SSTR subtypes in hypothyroidism in addition to its well-established role in negative regulation of hormonal secretion.
Collapse
Affiliation(s)
- Sneha Singh
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Rishi K Somvanshi
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Vandana Panda
- Department of Pharmacology & Toxicology, Principal K. M. Kundnani College of Pharmacy, Colaba, Mumbai, India
| | - Ujendra Kumar
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
13
|
Biological and Biochemical Basis of the Differential Efficacy of First and Second Generation Somatostatin Receptor Ligands in Neuroendocrine Neoplasms. Int J Mol Sci 2019; 20:ijms20163940. [PMID: 31412614 PMCID: PMC6720449 DOI: 10.3390/ijms20163940] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/1970] [Revised: 08/05/2019] [Accepted: 08/08/2019] [Indexed: 02/07/2023] Open
Abstract
Endogenous somatostatin shows anti-secretory effects in both physiological and pathological settings, as well as inhibitory activity on cell growth. Since somatostatin is not suitable for clinical practice, researchers developed synthetic somatostatin receptor ligands (SRLs) to overcome this limitation. Currently, SRLs represent pivotal tools in the treatment algorithm of neuroendocrine tumors (NETs). Octreotide and lanreotide are the first-generation SRLs developed and show a preferential binding affinity to somatostatin receptor (SST) subtype 2, while pasireotide, which is a second-generation SRL, has high affinity for multiple SSTs (SST5 > SST2 > SST3 > SST1). A number of studies demonstrated that first-generation and second-generation SRLs show distinct functional properties, besides the mere receptor affinity. Therefore, the aim of the present review is to critically review the current evidence on the biological effects of SRLs in pituitary adenomas and neuroendocrine tumors, by mainly focusing on the differences between first-generation and second-generation ligands.
Collapse
|
14
|
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: 1.2] [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
|
15
|
Tampourlou M, Karapanou O, Vassiliadi DA, Tsagarakis S. Medical therapy for non-functioning pituitary tumors-a critical approach. Hormones (Athens) 2019; 18:117-126. [PMID: 30368687 DOI: 10.1007/s42000-018-0070-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Accepted: 10/01/2018] [Indexed: 01/12/2023]
Abstract
Non-functioning pituitary adenomas (NFPAs) are the second most common variant of pituitary tumors. When symptomatic, primary therapy is surgery. Recurrence rates are high. Since many NFPAs express dopamine and somatostatin receptors, medical therapy has been used after surgery in order to prevent recurrence. So far, dopamine agonists have been more widely tested with some promise when introduced immediately after surgery but with less efficacy when introduced later upon tumor regrowth. Currently, the role of medical therapy to prevent tumor regrowth in NFPAs is limited by imprecisions as to final outcome and uncertainties concerning on patient selection, dosing, duration, and side effects.
Collapse
Affiliation(s)
- Metaxia Tampourlou
- Department of Endocrinology, Diabetes and Metabolism, Evangelismos Hospital, Athens, Greece.
| | - Olga Karapanou
- Department of Endocrinology, Diabetes and Metabolism, Evangelismos Hospital, Athens, Greece
| | - Dimitra A Vassiliadi
- Department of Endocrinology, Diabetes and Metabolism, Evangelismos Hospital, Athens, Greece
| | - Stylianos Tsagarakis
- Department of Endocrinology, Diabetes and Metabolism, Evangelismos Hospital, Athens, Greece
| |
Collapse
|
16
|
Dicuonzo F, Purciariello S, De Marco A, Guastamacchia E, Triggiani V. Inoperable Giant Growth Hormone-secreting Pituitary Adenoma: Radiological Aspects, Clinical Management and Pregnancy Outcome. Endocr Metab Immune Disord Drug Targets 2019; 19:214-220. [DOI: 10.2174/1871530318666180807160712] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 06/25/2018] [Accepted: 06/26/2018] [Indexed: 01/15/2023]
Abstract
Background and Objective: Giant pituitary adenomas (GPAs) are benign tumours with a
diameter ≥ 4 cm [1]. They can cause symptoms and signs due to the possible hyper-secretion of one or
more pituitary hormones, and involvement of the surrounding structures whereas the compression of
the pituitary itself can lead to hypopituitarism.
Methods:
We report on a young woman with acromegaly due to an inoperable giant GH-secreting
pituitary adenoma extending to right cavernous sinus, right orbital cavity, ethmoid, right maxillary
sinus, sphenoid sinus, clivus and right temporal fossa, in which medical treatment with Octreotide-
LAR was able to promptly relieve headache and bilateral hemianopsia due to optic chiasm involvement,
improve acromegaly symptoms and, over the time, control tumor expansion, improving fertility
and therefore allowing the patient to become pregnant.
Results:
Octreotide-LAR therapy was withdrawn during pregnancy and the patient did not experience
complications and gave birth to a healthy son. On magnetic resonance, the size of the tumor at the end
of pregnancy and in the subsequent follow up was not increased.
Conclusion:
The history we report, therefore, confirms previous experiences reporting a possible
favourable outcome of pregnancy in patients affected by acromegaly and adds further information
about the behaviour of giant pituitary tumors in patients underwent pregnancy.
Collapse
Affiliation(s)
- Franca Dicuonzo
- Department of Neuroradiology, School of Medicine, Deptartment of Basic Medical Sciences Neuroscience & Sense Organs, University of Bari , Italy
| | - Stefano Purciariello
- Department of Neuroradiology, School of Medicine, Deptartment of Basic Medical Sciences Neuroscience & Sense Organs, University of Bari , Italy
| | - Aurora De Marco
- Department of Neuroradiology, School of Medicine, Deptartment of Basic Medical Sciences Neuroscience & Sense Organs, University of Bari , Italy
| | - Edoardo Guastamacchia
- Interdisciplinary Department of Medicine, Section of Internal Medicine, Geriatrics, Endocrinology & Rare Diseases, University of Bari , Italy
| | - Vicenzo Triggiani
- Interdisciplinary Department of Medicine, Section of Internal Medicine, Geriatrics, Endocrinology & Rare Diseases, University of Bari , Italy
| |
Collapse
|
17
|
Gadelha MR, Kasuki L, Lim DST, Fleseriu M. Systemic Complications of Acromegaly and the Impact of the Current Treatment Landscape: An Update. Endocr Rev 2019; 40:268-332. [PMID: 30184064 DOI: 10.1210/er.2018-00115] [Citation(s) in RCA: 206] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 07/26/2018] [Indexed: 12/19/2022]
Abstract
Acromegaly is a chronic systemic disease with many complications and is associated with increased mortality when not adequately treated. Substantial advances in acromegaly treatment, as well as in the treatment of many of its complications, mainly diabetes mellitus, heart failure, and arterial hypertension, were achieved in the last decades. These developments allowed change in both prevalence and severity of some acromegaly complications and furthermore resulted in a reduction of mortality. Currently, mortality seems to be similar to the general population in adequately treated patients with acromegaly. In this review, we update the knowledge in complications of acromegaly and detail the effects of different acromegaly treatment options on these complications. Incidence of mortality, its correlation with GH (cumulative exposure vs last value), and IGF-I levels and the shift in the main cause of mortality in patients with acromegaly are also addressed.
Collapse
Affiliation(s)
- Mônica R Gadelha
- Neuroendocrinology Research Center/Endocrine Section and Medical School, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Neuroendocrine Section, Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria Estadual de Saúde do Rio de Janeiro, Rio de Janeiro, Brazil.,Neuropathology and Molecular Genetics Laboratory, Instituto Estadual do Cérebro Paulo Niemeyer, Rio de Janeiro, Brazil
| | - Leandro Kasuki
- Neuroendocrinology Research Center/Endocrine Section and Medical School, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Neuroendocrine Section, Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria Estadual de Saúde do Rio de Janeiro, Rio de Janeiro, Brazil.,Endocrine Unit, Hospital Federal de Bonsucesso, Rio de Janeiro, Brazil
| | - Dawn S T Lim
- Department of Endocrinology, Singapore General Hospital, Singapore, Singapore
| | - Maria Fleseriu
- Department of Endocrinology, Diabetes and Metabolism, Oregon Health and Science University, Portland, Oregon.,Department of Neurological Surgery, Oregon Health and Science University, Portland, Oregon.,Northwest Pituitary Center, Oregon Health and Science University, Portland, Oregon
| |
Collapse
|
18
|
Gatto F, Arvigo M, Amarù J, Campana C, Cocchiara F, Graziani G, Bruzzone E, Giusti M, Boschetti M, Ferone D. Cell specific interaction of pasireotide: review of preclinical studies in somatotroph and corticotroph pituitary cells. Pituitary 2019; 22:89-99. [PMID: 30483918 DOI: 10.1007/s11102-018-0926-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Pasireotide is a second-generation somatostatin (SRIF) receptor ligand (SRL), approved for medical treatment of acromegaly and Cushing's disease (CD). The molecule is a stable cyclohexapeptide synthetized based on SRIF structure. Differently from first-generation SRLs (e.g. octreotide), preferentially binding somatostatin receptor (SST) subtype 2 (SST2), pasireotide has high affinity for multiple SSTs (SST5 > SST2 > SST3 > SST1). Interestingly, early preclinical studies demonstrated that pasireotide shows distinct functional properties compared to SRIF and first-generation SRLs when binding SSTs. METHODS We aimed to highlight the differential receptor-targeted action of pasireotide in the treatment of somatotroph and corticotroph adenomas, throughout the critical revision of preclinical studies carried out on acromegaly and CD models. RESULTS Different authors demonstrated that the antisecretory effect of pasireotide in somatotroph adenoma cell cultures is comparable to that of the SST2-preferential agonist octreotide. Some reports even show a direct correlation between SST2 mRNA expression and GH reduction after pasireotide treatment, thus laying for a predominant role of SST2 in driving pasireotide efficacy in somatotropinomas in vitro. On the other hand, the inhibitory effect of pasireotide on ACTH secretion in corticotropinoma cells seems to be mainly mediated by SST5. Indeed, most reports show a higher potency and efficacy of pasireotide compared to SST2 preferential agonists, while functional studies confirm the pivotal role of SST5 targeting in corticotroph cells. CONCLUSIONS The analysis of preclinical studies carried out in somatotroph and corticoph adenomas points out that pasireotide shows a cell-specific activity, exerting its biological effects via different SSTs in the different adenoma histotypes.
Collapse
Affiliation(s)
- Federico Gatto
- Endocrinology Unit, Department of Internal Medicine, Policlinico San Martino, 16132, Genoa, Italy.
| | | | | | | | | | | | | | - Massimo Giusti
- Endocrinology Unit, Department of Internal Medicine, Policlinico San Martino, 16132, Genoa, Italy
- University of Genoa, Genoa, Italy
| | - Mara Boschetti
- Endocrinology Unit, Department of Internal Medicine, Policlinico San Martino, 16132, Genoa, Italy
- University of Genoa, Genoa, Italy
| | - Diego Ferone
- Endocrinology Unit, Department of Internal Medicine, Policlinico San Martino, 16132, Genoa, Italy
- University of Genoa, Genoa, Italy
| |
Collapse
|
19
|
Godara A, Siddiqui NS, Byrne MM, Saif MW. The safety of lanreotide for neuroendocrine tumor. Expert Opin Drug Saf 2018; 18:1-10. [PMID: 30582380 DOI: 10.1080/14740338.2019.1559294] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Lanreotide autogel is a synthetic somatostatin analogue which has been FDA and EMA approved for unresectable, well to moderately differentiated, locally advanced or metastatic gastroenteropancreatic neuroendocrine tumor. Its action is mediated by its affinity to somatostatin receptors, especially sst2 and sst5 receptors. Its longer half-life offers the convenience of 4-week dosing over the need for frequent injections of short-acting somatostatin analogues. Areas covered: Lanreotide ATG offers progression-free survival benefit in locally advanced or metastatic neuroendocrine tumor (NET) compared to placebo, reflecting a strong antiproliferative signal. As lanreotide is commonly used for management of NET, it is imperative to recognize and appropriately manage any drug-related toxicities. In this review, we will provide an overview of the toxicity with lanreotide and its management. Expert opinion: Lanreotide is highly effective in managing carcinoid symptoms and has a robust anti-tumor effect in NET. Overall, it is well tolerated with low rates of treatment discontinuation due to toxicity. It's toxicity profile is mostly predictable, and patients should be informed of the transient nature of some of the upfront toxicities.
Collapse
Affiliation(s)
- Amandeep Godara
- a Gastrointestinal Oncology Program and Experimental therapeutics, Division of Hematology/Oncology , Tufts Medical Center - Tufts University School of Medicine , Boston , MA , USA
| | - Nauman S Siddiqui
- a Gastrointestinal Oncology Program and Experimental therapeutics, Division of Hematology/Oncology , Tufts Medical Center - Tufts University School of Medicine , Boston , MA , USA
| | - Margaret M Byrne
- a Gastrointestinal Oncology Program and Experimental therapeutics, Division of Hematology/Oncology , Tufts Medical Center - Tufts University School of Medicine , Boston , MA , USA
| | - Muhammad Wasif Saif
- a Gastrointestinal Oncology Program and Experimental therapeutics, Division of Hematology/Oncology , Tufts Medical Center - Tufts University School of Medicine , Boston , MA , USA
| |
Collapse
|
20
|
Protein expression of somatostatin receptor 2, somatostatin receptor 5 and dopamine D2 receptor in normal pituitary gland and ACTH-secreting pituitary adenoma in dogs. Res Vet Sci 2018; 119:61-66. [PMID: 29864631 DOI: 10.1016/j.rvsc.2018.04.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 04/27/2018] [Accepted: 04/30/2018] [Indexed: 11/20/2022]
Abstract
PURPOSE To evaluate the protein expression of somatostatin receptor (SSTR) 2, SSTR5 and dopamine D2 receptor (DA2R)-targets of somatostatin analogs and dopamine agonists-in normal canine pituitary and canine adrenocorticotropic hormone (ACTH)-secreting pituitary adenomas. METHODS Six normal canine pituitary glands and 14 canine ACTH-secreting pituitary adenoma cases were included in this study. The protein expression of SSTR2, SSTR5 and DA2R was determined by double-label immunofluorescence staining of these receptors and ACTH. RESULTS SSTR2, SSTR5, and DA2R proteins were expressed in the anterior and intermediate lobes of normal canine pituitary glands. In the anterior pituitary lobes, the percentages of SSTR2-, SSTR5-, and DA2R-positive cells among the ACTH-positive population were 27.0 ± 8.6%, 27.9 ± 5.9%, and 34.0 ± 9.4%, respectively. In contrast, the corresponding percentages in the intermediate pituitary lobes were 97.8 ± 1.5%, 94.1 ± 4.4%, and 96.1 ± 6.6%, respectively. Of the 14 ACTH-secreting pituitary adenoma cases, 11, 12, and 6 cases expressed SSTR2, SSTR5, and DA2R, respectively. Additionally, four cases showed strong positive staining for both SSTR2 and SSTR5. Two of these were ACTH-secreting pituitary adenomas likely derived from the intermediate pituitary lobe, because these are α-Melanocyte-stimulation hormone (α-MSH)-positive stains. CONCLUSION Immunohistological detection and characterization of SSTR2, SSTR5 and DA2R may provide useful additional information for determining treatment options when an ACTH-secreting pituitary adenoma cannot be completely resected, or in the case of recurrence.
Collapse
|
21
|
Ciresi A, Radellini S, Guarnotta V, Giordano C. Efficacy of combined treatment with pasireotide, pegvisomant and cabergoline in an acromegalic patient resistant to other treatments: a case report. BMC Endocr Disord 2018; 18:2. [PMID: 29361932 PMCID: PMC5781307 DOI: 10.1186/s12902-018-0231-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 01/18/2018] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND The approach to acromegalic patients with persistent acromegaly after surgery and inadequate response to first-generation somatostatin receptor ligands (SRLs) should be strictly tailored. Current options include new pituitary surgery and/or radiosurgery, or alternative medical treatment with SRLs high dose regimens, pegvisomant (PEG) as monotherapy, or combined therapy with the addition of PEG or cabergoline to SRLs. A new pharmacological approach includes pasireotide, a second-generation SRL approved for patients who do not adequately respond to surgery and/or for whom surgery is not an option. No reports on efficacy and safety of combined therapy with pasireotide and pegvisomant (PEG) in acromegaly are available. CASE PRESENTATION Here we report the case of a 41-year-old acromegalic man with a mixed GH/PRL pituitary adenoma post-surgical resistant to first-generation SRLs both alone and in combination with cabergoline and PEG who achieved biochemical and tumor control with the combined triple treatment with pasireotide, PEG and cabergoline without adverse events and with a good compliance to treatment. CONCLUSIONS Twelve months of therapy with pasireotide, PEG and cabergoline proved to be safe and effective in this particular patient and the clinical improvement of disease resulted in an improved compliance to treatment.
Collapse
Affiliation(s)
- A. Ciresi
- Section of Endocrinology, Diabetology and Metabolic Diseases, Biomedical Department of Internal and Specialist Medicine (DIBIMIS), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy
| | - S. Radellini
- Section of Endocrinology, Diabetology and Metabolic Diseases, Biomedical Department of Internal and Specialist Medicine (DIBIMIS), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy
| | - V. Guarnotta
- Section of Endocrinology, Diabetology and Metabolic Diseases, Biomedical Department of Internal and Specialist Medicine (DIBIMIS), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy
| | - C. Giordano
- Section of Endocrinology, Diabetology and Metabolic Diseases, Biomedical Department of Internal and Specialist Medicine (DIBIMIS), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy
| |
Collapse
|
22
|
Paragliola RM, Salvatori R. Novel Somatostatin Receptor Ligands Therapies for Acromegaly. Front Endocrinol (Lausanne) 2018; 9:78. [PMID: 29563895 PMCID: PMC5845985 DOI: 10.3389/fendo.2018.00078] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 02/20/2018] [Indexed: 12/21/2022] Open
Abstract
Surgery is considered the treatment of choice in acromegaly, but patients with persistent disease after surgery or in whom surgery cannot be considered require medical therapy. Somatostatin receptor ligands (SRLs) octreotide (OCT), lanreotide, and the more recently approved pasireotide, characterized by a broader receptor ligand binding profile, are considered the mainstay in the medical management of acromegaly. However, in the attempt to offer a more efficacious and better tolerated medical approach, recent research has been aimed to override some limitations related to the use of currently approved drugs and novel SRLs therapies, with potential attractive features, have been proposed. These include both new formulation of older molecules and new molecules. Novel OCT formulations are aimed in particular to improve patients' compliance and to reduce injection discomfort. They include an investigational ready-to-use subcutaneous depot OCT formulation (CAM2029), delivered via prefilled syringes and oral OCT that uses a "transient permeability enhancer" technology, which allows for OCT oral absorption. Another new delivery system is a long-lasting OCT implant (VP-003), which provide stable doses of OCT throughout a period of several months. Finally, a new SRL DG3173 (somatoprim) seems to be more selective for GH secretion, suggesting possible advantages in the presence of hyperglycemia or diabetes. How much these innovations will actually be beneficial to acromegaly patients in real clinical practice remains to be seen.
Collapse
Affiliation(s)
| | - Roberto Salvatori
- Department of Medicine, Division of Endocrinology, Metabolism and Diabetes, Pituitary Center Johns Hopkins University School of Medicine, Baltimore, MD, United States
- *Correspondence: Roberto Salvatori,
| |
Collapse
|
23
|
Henninot A, Collins JC, Nuss JM. The Current State of Peptide Drug Discovery: Back to the Future? J Med Chem 2017; 61:1382-1414. [PMID: 28737935 DOI: 10.1021/acs.jmedchem.7b00318] [Citation(s) in RCA: 703] [Impact Index Per Article: 87.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Over the past decade, peptide drug discovery has experienced a revival of interest and scientific momentum, as the pharmaceutical industry has come to appreciate the role that peptide therapeutics can play in addressing unmet medical needs and how this class of compounds can be an excellent complement or even preferable alternative to small molecule and biological therapeutics. In this Perspective, we give a concise description of the recent progress in peptide drug discovery in a holistic manner, highlighting enabling technological advances affecting nearly every aspect of this field: from lead discovery, to synthesis and optimization, to peptide drug delivery. An emphasis is placed on describing research efforts to overcome the inherent weaknesses of peptide drugs, in particular their poor pharmacokinetic properties, and how these efforts have been critical to the discovery, design, and subsequent development of novel therapeutics.
Collapse
Affiliation(s)
- Antoine Henninot
- Ferring Research Institute , 4245 Sorrento Valley Boulevard, San Diego, California 92121, United States
| | - James C Collins
- Ferring Research Institute , 4245 Sorrento Valley Boulevard, San Diego, California 92121, United States
| | - John M Nuss
- Ferring Research Institute , 4245 Sorrento Valley Boulevard, San Diego, California 92121, United States
| |
Collapse
|
24
|
de Fátima Borges M, Lara BHJ, Tomé JM, de Araújo LP, Bugiga FCL, Sousa JC, Soares JMF, Dezena RA, Ferreira BP. Treatment of acromegaly patients at the Federal University of Triângulo Mineiro (UFTM): Experience Report. Clinics (Sao Paulo) 2017; 72:218-223. [PMID: 28492721 PMCID: PMC5401613 DOI: 10.6061/clinics/2017(04)05] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 12/20/2016] [Accepted: 01/12/2017] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE: To evaluate the effectiveness of the treatment of acromegaly patients at the Federal University of Triangulo Mineiro. METHODS: Cross-sectional and retrospective study of thirty cases treated over a period of two decades. RESULTS: 17 men (56.7%) aged 14-67 years and 13 women aged 14-86 years were analyzed. Twenty-one patients underwent transphenoidal surgery, whichwas associated with somatostatin receptor ligands in 11 patients (39.3%), somatostatin receptor ligands + radiotherapyin 5 patients (17.8%), radiotherapy in 3 patients (10.7%), and radiotherapy + somatostatin receptorligands + cabergoline in 1 patient (3.6%). Additionally, 2 patients underwent radiotherapy and surgeryalone. Six patients received somatostatin receptor ligands before surgery, and 2 were not treated due to refusal and death. Nine patients have died, and 20 are being followed; 13 (65%) have growth hormonelevels o1 ng/mL, and 11 have normal insulin-like growth factor 1 levels. CONCLUSION: The current treatment options enable patients seen in regional reference centers to achieve strict control parameters, which allows them to be treated close to their homes.
Collapse
Affiliation(s)
- Maria de Fátima Borges
- Divisão de Endocrinologia e Metabolismo, Universidade Federal do Triângulo Mineiro, Uberaba, MG, BR
| | | | - Janaíne Machado Tomé
- Divisão de Endocrinologia e Metabolismo, Universidade Federal do Triângulo Mineiro, Uberaba, MG, BR
| | | | | | - Júlio Cláudio Sousa
- Divisão de Otorrinolaringologia, Universidade Federal do Triângulo Mineiro, Uberaba, MG, BR
| | | | | | - Beatriz Pires Ferreira
- Divisão de Endocrinologia e Metabolismo, Universidade Federal do Triângulo Mineiro, Uberaba, MG, BR
| |
Collapse
|
25
|
Abstract
First-generation somatostatin receptors ligands (SRL) are the mainstay in the medical treatment of acromegaly, however the percentage of patients controlled with these drugs significantly varies in the different studies. Many factors are involved in the resistance to SRL. In this review, we update the physiology of somatostatin and its receptors (sst), the use of SRL in the treatment of acromegaly and the factors involved in the response to these drugs. The SRL act through interaction with the sst, which up to now have been characterized as five subtypes. The first-generation SRL, octreotide and lanreotide, are considered sst2 specific and have biochemical response rates varying from 20 to 70%. Tumor volume reduction can be found in 36-75% of patients. Several factors may determine the response to these drugs, such as sst, AIP, E-cadherin, ZAC1, filamin A and β-arrestin expression in the somatotropinomas. In patients resistant to first-generation SRL, alternative medical treatment options include: SRL high dose regimens, SRL in combination with cabergoline or pegvisomant, or the use of pasireotide. Pasireotide is a next-generation SRL with a broader pattern of interaction with sst. In the light of the recent increase of treatment options in acromegaly and the deeper knowledge of the determinants of response to the current first-line therapy, a shift from a trial-and-error treatment to a personalized one could be possible.
Collapse
Affiliation(s)
- Monica R Gadelha
- Neuroendocrinology Research Center/Endocrinology Section, Medical School and Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rua Prof. Rodolpho Paulo Rocco, 9th floor, Ilha do Fundão, Rio de Janeiro, 21941-913, Brazil.
- Neuroendocrinology Section and Molecular Genetics Laboratory, Secretaria Estadual de Saúde do Rio de Janeiro, Instituto Estadual do Cérebro Paulo Niemeyer, Rio de Janeiro, Brazil.
| | - Luiz Eduardo Wildemberg
- Neuroendocrinology Research Center/Endocrinology Section, Medical School and Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rua Prof. Rodolpho Paulo Rocco, 9th floor, Ilha do Fundão, Rio de Janeiro, 21941-913, Brazil
- Neuroendocrinology Section and Molecular Genetics Laboratory, Secretaria Estadual de Saúde do Rio de Janeiro, Instituto Estadual do Cérebro Paulo Niemeyer, Rio de Janeiro, Brazil
| | - Marcello D Bronstein
- Neuroendocrine Unit, Division of Endocrinology and Metabolism, Hospital das Clinicas, University of Sao Paulo Medical School, São Paulo, Brazil
| | - Federico Gatto
- Endocrinology Unit, Department of Internal Medicine and Medical Specialties and Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Diego Ferone
- Endocrinology Unit, Department of Internal Medicine and Medical Specialties and Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
- IRCCS AOU San Martino-IST Genoa, Genoa, Italy
| |
Collapse
|
26
|
Calderón MDR, Delgado E, García Campos F. Acromegaly and associated tumours: what should gastroenterologists know? GASTROENTEROLOGIA Y HEPATOLOGIA 2017; 40:41-47. [PMID: 26966026 DOI: 10.1016/j.gastrohep.2015.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 11/21/2015] [Accepted: 12/04/2015] [Indexed: 06/05/2023]
Abstract
Acromegaly is a clinical syndrome caused by the excessive production of growth hormone. It is associated with high morbidity and significantly increased mortality, mainly due to cardiovascular and respiratory complications, and cancer. Mortality is reduced to that of the general population following successful treatment, in other words, when insulin-like growth factor (IGF-I) and growth hormone values return to normal levels. Not all tumours associated with this syndrome benefit from cost-effective early diagnosis programmes. An in-depth knowledge on the part of clinicians of the morbidity and mortality associated with acromegaly, allowing them in many cases to anticipate the expected clinical course of the disease, is the best therapeutic and follow-up strategy in these patients.
Collapse
|
27
|
Liu W, Xie L, He M, Shen M, Zhu J, Yang Y, Wang M, Hu J, Ye H, Li Y, Zhao Y, Zhang Z. Expression of Somatostatin Receptor 2 in Somatotropinoma Correlated with the Short-Term Efficacy of Somatostatin Analogues. Int J Endocrinol 2017; 2017:9606985. [PMID: 28396686 PMCID: PMC5370518 DOI: 10.1155/2017/9606985] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 01/23/2017] [Indexed: 11/17/2022] Open
Abstract
The expression of somatostatin receptor subtypes (SSTRs) in pituitary growth hormone- (GH-) secreting adenomas may predict the response to somatostatin analogues (SSA). Our aim was to evaluate the value of the immunohistochemical (IHC) scores of 2 subtypes, SSTR2 and SSTR5, in predicting the short-term efficacy of SSA therapy in patients with active acromegaly. Ninety-three newly diagnosed acromegalic patients were included in our study. These patients were categorized into either a SSA-pretreated group (SA, n = 63) or a direct-surgery group (DS, n = 30), depending on whether or not presurgical SSA treatment was received. IHC analysis, using a 12-grade scoring system, with rabbit monoclonal antibodies against SSTR2 and SSTR5, was performed on all adenoma tissues. The reduction of GH, IGF-1, and tumor size after treatment with SSA for 3 months was measured. Compared with that in the DS group, SSTR2 expression was lower in the SA group. Additionally, in the SA group, SSTR2 expression was positively correlated with the reduction of IGF-1 and tumor volume. However, there was no correlation between the SSTR5 score and the efficacy of SSA. In conclusion, the protein expression of SSTR2, but not of SSTR5, is a valuable indicator in predicting biochemical and tumor size response to short-term SSA treatment in acromegalic patients.
Collapse
Affiliation(s)
- Wenjuan Liu
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai 200040, China
- Institute of Endocrinology and Diabetology, Fudan University, Shanghai 200040, China
| | - Lina Xie
- Department of Endocrinology, Kunshan Rehabilitation Hospital, Suzhou, Jiangsu 215314, China
| | - Min He
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai 200040, China
- Institute of Endocrinology and Diabetology, Fudan University, Shanghai 200040, China
| | - Ming Shen
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China
- Shanghai Pituitary Tumor Center, Shanghai 200040, China
| | - Jingjing Zhu
- Department of Pathology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yeping Yang
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai 200040, China
- Institute of Endocrinology and Diabetology, Fudan University, Shanghai 200040, China
| | - Meng Wang
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai 200040, China
- Institute of Endocrinology and Diabetology, Fudan University, Shanghai 200040, China
| | - Ji Hu
- Department of Endocrinology, The Second Affiliated Hospital, Soochow University, Suzhou, Jiangsu 215004, China
| | - Hongying Ye
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai 200040, China
- Shanghai Pituitary Tumor Center, Shanghai 200040, China
| | - Yiming Li
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai 200040, China
- Institute of Endocrinology and Diabetology, Fudan University, Shanghai 200040, China
- Shanghai Pituitary Tumor Center, Shanghai 200040, China
| | - Yao Zhao
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China
- Shanghai Pituitary Tumor Center, Shanghai 200040, China
| | - Zhaoyun Zhang
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai 200040, China
- Institute of Endocrinology and Diabetology, Fudan University, Shanghai 200040, China
- Shanghai Pituitary Tumor Center, Shanghai 200040, China
- *Zhaoyun Zhang:
| |
Collapse
|
28
|
Paragliola RM, Prete A, Papi G, Torino F, Corsello A, Pontecorvi A, Corsello SM. Clinical utility of lanreotide Autogel ® in gastroenteropancreatic neuroendocrine tumors. DRUG DESIGN DEVELOPMENT AND THERAPY 2016; 10:3459-3470. [PMID: 27822010 PMCID: PMC5087808 DOI: 10.2147/dddt.s76732] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Somatostatin analogs (SSAs), which were initially used to control hormonal syndromes associated with neuroendocrine neoplasms (NENs), have been successfully proposed as antiproliferative agents, able to control tumor growth in patients affected by gastroenteropancreatic (GEP)-NENs. The development of long-acting formulations of SSAs which require only weekly or monthly injections can improve patient compliance. In particular, lanreotide (LAN) Autogel®, which is a viscous aqueous formulation supplied in ready-to-use prefilled syringes, can be administered every 28–56 days. Since its introduction in the clinical practice, several studies evaluated the clinical utility of LAN Autogel in the medical treatment of GEP-NENs. Although there is no evidence of an overall survival benefit, these studies confirm the efficacy of LAN Autogel in terms of benefit in progression-free survival, and in more than half of cases, a reduction of tumor markers can be observed during treatment with this drug. Moreover, LAN Autogel is widely recognized to be effective in controlling tumor-related symptoms in the majority of patients affected by GEP tumors, especially in patients affected by carcinoid syndrome, improving considerably patients’ quality of life.
Collapse
Affiliation(s)
| | - Alessandro Prete
- Department of Medicine, Unit of Endocrinology, Università Cattolica del Sacro Cuore
| | - Giampaolo Papi
- Department of Medicine, Unit of Endocrinology, Università Cattolica del Sacro Cuore
| | | | - Andrea Corsello
- Department of General Medicine and Endocrine Tumor Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Alfredo Pontecorvi
- Department of Medicine, Unit of Endocrinology, Università Cattolica del Sacro Cuore
| | | |
Collapse
|
29
|
Fleseriu M, Hoffman AR, Katznelson L. AMERICAN ASSOCIATION OF CLINICAL ENDOCRINOLOGISTS AND AMERICAN COLLEGE OF ENDOCRINOLOGY DISEASE STATE CLINICAL REVIEW: MANAGEMENT OF ACROMEGALY PATIENTS: WHAT IS THE ROLE OF PRE-OPERATIVE MEDICAL THERAPY? Endocr Pract 2016; 21:668-73. [PMID: 26135961 DOI: 10.4158/ep14575.dscr] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVE Acromegaly is a complex disease characterized by growth hormone (GH) excess originating in most cases from a pituitary tumor. The goals of treatment include removing the tumor or reducing tumor burden, normalizing GH secretion and insulin-like growth factor 1 levels, and preserving normal pituitary function if possible. Surgery by an experienced neurosurgeon is still considered first-line therapy, especially in cases with small tumors. In the last few decades, significant progress in the development of selective pharmacologic agents has greatly facilitated the management of active acromegaly, with agents such as somatostatin-receptor ligands (SRLs), GH-receptor antagonists, and dopamine agonists. In addition to adjuvant treatment, pre-operative medical therapy and primary therapy in de novo patients are increasingly employed. METHODS A United States National Library of Medicine PubMed search (through July 2014) was conducted for the following terms: acromegaly, pre-operative medical therapy, somatostatin-receptor ligands, and somatostatin analogs. Articles not in English and those not in peer-reviewed journals were excluded. In reviewing pertinent articles, focus was placed on biochemical and other postoperative outcomes of medical therapy. RESULTS An analysis of the full effect of pre-operative use of SRLs on surgical outcomes (remission rates and peri-operative complications) is limited by heterogeneity of methodology, low overall surgical cure rates, and different study designs. The assumption that SRL use prior to surgery reduces peri-operative surgical risk has yet to be proven. A variable degree of tumor shrinkage with preoperative SRLs is observed. Likewise, SRL treatment 3 months before surgery may improve surgical remission rates in the short term; however, positive results do not persist in the long term. CONCLUSION We consider that medical therapy before surgery could play a role in carefully selected patients, but treatment should be individualized. Primary medical therapy with a SRL may be considered in patients with macroadenomas without local mass effects on the optic chiasm, as SRLs have been shown to reduce tumor size and control GH hypersecretion. However, the data are insufficient to support general use of a SRL prior to surgery in order to improve post-surgery biochemical outcomes. Theoretically, patients with severe cardiac and respiratory complications due to acromegaly could potentially benefit from pre-operative SRLs in order to reduce peri-operative morbidity. Further investigation and investment in large randomized long-term clinical trials are needed to define the precise role and duration of pre-surgical medical treatment in acromegaly patients.
Collapse
|
30
|
Sagvand BT, Khairi S, Haghshenas A, Swearingen B, Tritos NA, Miller KK, Klibanski A, Nachtigall LB. Monotherapy with lanreotide depot for acromegaly: long-term clinical experience in a pituitary center. Pituitary 2016; 19:437-47. [PMID: 27155600 DOI: 10.1007/s11102-016-0724-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
PURPOSE Long-acting somatostatin analogs are one of the main classes of medical therapy used for acromegaly and most patients require ongoing treatment. Few studies have evaluated the long-term efficacy and safety of lanreotide depot beyond 2 years. The goal of this study was to provide a long-term longitudinal assessment of efficacy and safety of lanreotide depot in lanreotide responders compared to a surgically cured control group. METHODS In this retrospective longitudinal case-control study, patients with acromegaly receiving lanreotide depot monotherapy continuously for at least 24 months (N = 24) and surgically cured patients (N = 39) were compared. Serum IGF-1, pituitary MRIs, lanreotide dose, co-morbidities and adverse effects were assessed longitudinally. RESULTS In the lanreotide group, IGF-1 remained normal and unchanged over 6 years; comparable to the surgery only group. There was no difference in prevalence of normal IGF-1 between the lanreotide and surgery only groups at 6 months (100 vs. 97 %), 6 years (89 vs. 90 %) and at last follow-up (96 vs. 92 %). Tumor size remained stable (79 %) or decreased (21 %) in the lanreotide group. In the surgery only group, tumor size remained unchanged in all patients. Hemoglobin A1C did not differ between lanreotide and surgery only groups (baseline 5.8 vs. 6.1 %; last follow-up 6.0 vs. 5.7 %). Two (8 %) of the lanreotide and none of the surgery only group developed new diabetes mellitus. CONCLUSION Lanreotide depot maintains normalization of IGF-1 in 89 % of responders after 6 years, comparable to surgically cured controls, and controlled tumor size in all without significant adverse effects.
Collapse
Affiliation(s)
- Babak Torabi Sagvand
- Neuroendocrine Unit, Neuroendocrine Clinical Center, Massachusetts General Hospital, Harvard Medical School, Zero Emerson Place, Suite 112, Boston, MA, 02114, USA
| | - Shafaq Khairi
- Neuroendocrine Unit, Neuroendocrine Clinical Center, Massachusetts General Hospital, Harvard Medical School, Zero Emerson Place, Suite 112, Boston, MA, 02114, USA
| | - Arezoo Haghshenas
- Neuroendocrine Unit, Neuroendocrine Clinical Center, Massachusetts General Hospital, Harvard Medical School, Zero Emerson Place, Suite 112, Boston, MA, 02114, USA
| | - Brooke Swearingen
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Nicholas A Tritos
- Neuroendocrine Unit, Neuroendocrine Clinical Center, Massachusetts General Hospital, Harvard Medical School, Zero Emerson Place, Suite 112, Boston, MA, 02114, USA
| | - Karen K Miller
- Neuroendocrine Unit, Neuroendocrine Clinical Center, Massachusetts General Hospital, Harvard Medical School, Zero Emerson Place, Suite 112, Boston, MA, 02114, USA
| | - Anne Klibanski
- Neuroendocrine Unit, Neuroendocrine Clinical Center, Massachusetts General Hospital, Harvard Medical School, Zero Emerson Place, Suite 112, Boston, MA, 02114, USA
| | - Lisa B Nachtigall
- Neuroendocrine Unit, Neuroendocrine Clinical Center, Massachusetts General Hospital, Harvard Medical School, Zero Emerson Place, Suite 112, Boston, MA, 02114, USA.
| |
Collapse
|
31
|
Abstract
INTRODUCTION In nearly all cases, acromegaly is caused by excess GH from a pituitary adenoma, resulting in elevated circulating levels of GH and, subsequently, IGF-1. Treatment goals are to eliminate morbidity and restore the increased mortality to normal rates. Therapeutic strategies aim to minimize tumor mass and normalize GH and IGF-1 levels. Somatostatin analogues are the medical treatment of choice in acromegaly, as first-line or post-surgical therapy, and have proven efficacy in pituitary tumor volume reduction (TVR). METHODS Here we review the effects of somatostatin analogue therapy on pituitary tumor volume in patients with acromegaly. RESULTS TVR with somatostatin analogues may be mediated by direct anti-proliferative effects via activation of somatostatin receptors, or by indirect effects, such as angiogenesis inhibition, and is more pronounced when they are administered as first-line therapy. Various studies of first-line treatment with octreotide LAR have shown significant TVR in ≥73% of patients. First-line treatment with lanreotide Autogel has shown evidence of TVR, although more studies are needed. In a recent randomized, double-blind, 12-month trial in 358 medical-treatment-naïve acromegaly patients, significant TVR was achieved by 81% of patients administered pasireotide LAR and 77% administered octreotide LAR. Pre-operative somatostatin analogue therapy may also induce TVR and improve post-operative disease control compared with surgery alone. TVR is progressive with prolonged treatment, and decreased IGF-1 levels may be its best predictor, followed by age and degree of GH decrease. However, TVR does not always correlate with degree of biochemical control. CONCLUSION Somatostatin analogues (first- or second-line treatment) are the mainstay of medical therapy and, as first-line medical therapy, are associated with significant pituitary TVR in most patients.
Collapse
Affiliation(s)
- Annamaria Colao
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, via S Pansini 5, 80131, Naples, Italy.
| | - Renata S Auriemma
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, via S Pansini 5, 80131, Naples, Italy
| | - Rosario Pivonello
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, via S Pansini 5, 80131, Naples, Italy
| |
Collapse
|
32
|
Abstract
Morbidity and mortality rates in patients with active acromegaly are higher than the general population. Adequate biochemical control restores mortality to normal rates. Now, medical therapy has an increasingly important role in the treatment of patients with acromegaly. Somatostatin receptor ligands (SRLs) are considered the standard medical therapy, either after surgery or as a first-line therapy when surgery is deemed ineffective or is contraindicated. Overall, octreotide and lanreotide are first-generation SRLs and are effective in ~20%-70% of patients. Pegvisomant, a growth hormone receptor antagonist, controls insulin-like growth factor 1 in 65%-90% of cases. Consequently, a subset of patients (nonresponders) requires other treatment options. Drug combination therapy offers the potential for more efficacious disease control. However, the development of new medical therapies remains essential. Here, emphasis is placed on new medical therapies to control acromegaly. There is a focus on pasireotide long-acting release (LAR) (Signifor LAR®), which was approved in 2014 by the US Food and Drug Administration and the European Medicine Agency for the treatment of acromegaly. Pasireotide LAR is a long-acting somatostatin multireceptor ligand. In a Phase III clinical trial in patients with acromegaly (naïve to medical therapy or uncontrolled on a maximum dose of first-generation SRLs), 40 and 60 mg of intramuscular pasireotide LAR achieved better biochemical disease control than octreotide LAR, and tumor shrinkage was noted in both pasireotide groups. Pasireotide LAR tolerability was similar to other SRLs, except for a greater frequency and degree of hyperglycemia and diabetes mellitus. Baseline glucose may predict hyperglycemia occurrence after treatment, and careful monitoring of glycemic status and appropriate treatment is required. A precise definition of patients with acromegaly who will derive the greatest therapeutic benefit from pasireotide LAR remains to be established. Lastly, novel therapies and new potential delivery modalities (oral octreotide) are summarized.
Collapse
Affiliation(s)
- Daniel Cuevas-Ramos
- Department of Endocrinology and Metabolism, Neuroendocrinology Clinic, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Maria Fleseriu
- Department of Medicine (Endocrinology), Portland, OR, USA
- Department of Neurological Surgery, Northwest Pituitary Center, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
33
|
Balti EV, Akwo EA, Fezeu L, Kengne AP, Sobngwi E, Mbanya JC. Somatostatin analogues, dopamine agonists or growth hormone antagonists for pituitary adenoma-induced acromegaly. Hippokratia 2015. [DOI: 10.1002/14651858.cd008292.pub2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Eric V Balti
- University of Yaounde I; National Obesity Centre and HoPiT Research Group, FMSB; PO Box 7535 Yaounde Cameroon
| | - Elvis A Akwo
- University of Yaounde I; National Obesity Centre and HoPiT Research Group, FMSB; PO Box 7535 Yaounde Cameroon
| | - Leopold Fezeu
- University of Yaounde I; National Obesity Centre and HoPiT Research Group, FMSB; PO Box 7535 Yaounde Cameroon
| | - Andre Pascal Kengne
- University of Yaounde I; National Obesity Centre and HoPiT Research Group, FMSB; PO Box 7535 Yaounde Cameroon
| | - Eugene Sobngwi
- University of Yaounde I; National Obesity Centre and HoPiT Research Group, FMSB; PO Box 7535 Yaounde Cameroon
| | - Jean Claude Mbanya
- University of Yaounde I; National Obesity Centre and HoPiT Research Group, FMSB; PO Box 7535 Yaounde Cameroon
| |
Collapse
|
34
|
Scherbarth F, Diedrich V, Dumbell RA, Schmid HA, Steinlechner S, Barrett P. Somatostatin receptor activation is involved in the control of daily torpor in a seasonal mammal. Am J Physiol Regul Integr Comp Physiol 2015; 309:R668-74. [DOI: 10.1152/ajpregu.00191.2015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 07/05/2015] [Indexed: 01/21/2023]
Abstract
Siberian hamsters ( Phodopus sungorus) show spontaneous daily torpor only after ∼2 mo in winter-like short photoperiods (SP). Although some SP-induced hormonal changes have been demonstrated to be necessary for the occurrence of seasonal torpor, the whole set of preconditions is still unknown. Recent findings provide evidence that the hypothalamic pituitary growth axis is involved in endocrine responses to SP exposure in the photoperiodic hamsters. To examine whether suppression of growth hormone (GH) and insulin-like growth factor-1 (IGF-1) secretion affects the incidence of daily torpor, we used two somatostatin receptor agonists, pasireotide (SOM230) and octreotide, with different affinity profiles for receptor subtypes. Pasireotide strikingly increased the torpor frequency in male hamsters compared with sham-treated controls, and torpor duration was often increased, which in some cases exceeded 12 h. In contrast, administration of octreotide reduced the body weight of SP hamsters but had only a marginal effect on torpor frequency in males and no effect in females. Together with measured concentrations of circulating IGF-1, the present results strongly suggest that reduced activity of the GH/IGF-1 axis is not critical for stimulation of torpor expression but activation of specific somatostatin receptors is critical. This putative role for certain somatostatin receptor subtypes in torpor induction provides a promising new approach to unravel the endocrine mechanisms of torpor regulation.
Collapse
Affiliation(s)
- Frank Scherbarth
- Department of Zoology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Victoria Diedrich
- Department of Zoology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Rebecca A. Dumbell
- Rowett Institute for Nutrition and Health, Aberdeen, United Kingdom; and
| | - Herbert A. Schmid
- Oncology Department, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Stephan Steinlechner
- Department of Zoology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Perry Barrett
- Rowett Institute for Nutrition and Health, Aberdeen, United Kingdom; and
| |
Collapse
|
35
|
Sakai K, Yonezawa T, Yamawaki H, Oyamada T. Alteration of somatostatin receptor 2 expression in canine mammary gland tumor. J Vet Med Sci 2015; 77:1319-22. [PMID: 25985817 PMCID: PMC4638304 DOI: 10.1292/jvms.15-0002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Somatostatin receptor 2 (SSTR2) is a negative regulator of cell proliferation in human
breast cancer. Since there is little information about SSTR2 in canine mammary gland tumor
(MGT), we clarified its distribution and expression level in normal mammary gland, benign
MGT and malignant MGT. SSTR2 expression determined by immunohistochemical staining was
observed in the cytoplasm of luminal epithelial cells. The intensity was negatively
correlated with malignancy: normal tissues and some of the benign tumors had the highest
levels, while the malignant tumors had little or no SSTR2 expression. As for the Western
blotting, SSTR2 protein level in benign tumors was significantly lower than the normal
mammary gland. On the other hand, SSTR2 protein levels in two of three malignant tumors
were higher than the other groups. These results suggest that SSTR2 expression alters
according to the malignancy of canine MGT.
Collapse
Affiliation(s)
- Kosei Sakai
- Department of Veterinary Pathology, School of Veterinary Medicine, Kitasato University, Aomori 034-8628, Japan
| | | | | | | |
Collapse
|
36
|
Lee M, Lupp A, Mendoza N, Martin N, Beschorner R, Honegger J, Schlegel J, Shively T, Pulz E, Schulz S, Roncaroli F, Pellegata NS. SSTR3 is a putative target for the medical treatment of gonadotroph adenomas of the pituitary. Endocr Relat Cancer 2015; 22:111-9. [PMID: 25515731 DOI: 10.1530/erc-14-0472] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Gonadotroph pituitary adenomas (GPAs) often present as invasive macroadenomas not amenable to complete surgical resection. Radiotherapy is the only post-operative option for patients with large invasive or recurrent lesions. No medical treatment is available for these patients. The somatostatin analogs (SSAs) octreotide and lanreotide that preferentially target somatostatin receptor type 2 (SSTR2) have little effect on GPAs. It is widely accepted that the expression of specific SSTR subtypes determines the response to SSAs. Given that previous studies on mRNA and protein expression of SSTRs in GPAs have generated conflicting results, we investigated the expression of SSTR2, SSTR3, and SSTR5 (the main targets of available SSAs) in a clinically and pathologically well-characterized cohort of 108 patients with GPAs. A total of 118 samples were examined by immunohistochemistry using validated and specific MABs. Matched primary and recurrent tissues were available for ten patients. The results obtained were validated in an independent cohort of 27 GPAs. We observed that SSTR3 was significantly more abundant than SSTR2 (P<0.0001) in GPAs, while full-length SSTR5 was only expressed in few tumors. Expression of SSTR3 was similar in primary and recurrent adenomas, was high in potentially aggressive lesions, and did not change significantly in adenomas that recurred after irradiation. In conclusion, low levels of expression of SSTR2 may account for the limited response of GPAs to octreotide and lanreotide. Given the potent anti-proliferative, pro-apoptotic, and anti-angiogenic activities of SSTR3, targeting this receptor with a multireceptor ligand SSA such as pasireotide may be indicated for potentially aggressive GPAs.
Collapse
Affiliation(s)
- Misu Lee
- Institute of PathologyHelmholtz Zentrum München, D-85764 Neuherberg, GermanyDepartment of Pharmacology and ToxicologyJena University Hospital, Friedrich Schiller University, Jena, GermanyDepartments of NeurosurgeryMedicineImperial College, St Dunstans Road, London W6 8RP, UKDepartment of NeuropathologyInstitute for Pathology and NeuropathologyDepartment of NeurosurgeryUniversity of Tübingen, Tübingen, GermanyInstitute of PathologyTechnical University of Munich, München, Germany
| | - Amelie Lupp
- Institute of PathologyHelmholtz Zentrum München, D-85764 Neuherberg, GermanyDepartment of Pharmacology and ToxicologyJena University Hospital, Friedrich Schiller University, Jena, GermanyDepartments of NeurosurgeryMedicineImperial College, St Dunstans Road, London W6 8RP, UKDepartment of NeuropathologyInstitute for Pathology and NeuropathologyDepartment of NeurosurgeryUniversity of Tübingen, Tübingen, GermanyInstitute of PathologyTechnical University of Munich, München, Germany
| | - Nigel Mendoza
- Institute of PathologyHelmholtz Zentrum München, D-85764 Neuherberg, GermanyDepartment of Pharmacology and ToxicologyJena University Hospital, Friedrich Schiller University, Jena, GermanyDepartments of NeurosurgeryMedicineImperial College, St Dunstans Road, London W6 8RP, UKDepartment of NeuropathologyInstitute for Pathology and NeuropathologyDepartment of NeurosurgeryUniversity of Tübingen, Tübingen, GermanyInstitute of PathologyTechnical University of Munich, München, Germany
| | - Niamh Martin
- Institute of PathologyHelmholtz Zentrum München, D-85764 Neuherberg, GermanyDepartment of Pharmacology and ToxicologyJena University Hospital, Friedrich Schiller University, Jena, GermanyDepartments of NeurosurgeryMedicineImperial College, St Dunstans Road, London W6 8RP, UKDepartment of NeuropathologyInstitute for Pathology and NeuropathologyDepartment of NeurosurgeryUniversity of Tübingen, Tübingen, GermanyInstitute of PathologyTechnical University of Munich, München, Germany
| | - Rudi Beschorner
- Institute of PathologyHelmholtz Zentrum München, D-85764 Neuherberg, GermanyDepartment of Pharmacology and ToxicologyJena University Hospital, Friedrich Schiller University, Jena, GermanyDepartments of NeurosurgeryMedicineImperial College, St Dunstans Road, London W6 8RP, UKDepartment of NeuropathologyInstitute for Pathology and NeuropathologyDepartment of NeurosurgeryUniversity of Tübingen, Tübingen, GermanyInstitute of PathologyTechnical University of Munich, München, Germany
| | - Jürgen Honegger
- Institute of PathologyHelmholtz Zentrum München, D-85764 Neuherberg, GermanyDepartment of Pharmacology and ToxicologyJena University Hospital, Friedrich Schiller University, Jena, GermanyDepartments of NeurosurgeryMedicineImperial College, St Dunstans Road, London W6 8RP, UKDepartment of NeuropathologyInstitute for Pathology and NeuropathologyDepartment of NeurosurgeryUniversity of Tübingen, Tübingen, GermanyInstitute of PathologyTechnical University of Munich, München, Germany
| | - Jürgen Schlegel
- Institute of PathologyHelmholtz Zentrum München, D-85764 Neuherberg, GermanyDepartment of Pharmacology and ToxicologyJena University Hospital, Friedrich Schiller University, Jena, GermanyDepartments of NeurosurgeryMedicineImperial College, St Dunstans Road, London W6 8RP, UKDepartment of NeuropathologyInstitute for Pathology and NeuropathologyDepartment of NeurosurgeryUniversity of Tübingen, Tübingen, GermanyInstitute of PathologyTechnical University of Munich, München, Germany
| | - Talia Shively
- Institute of PathologyHelmholtz Zentrum München, D-85764 Neuherberg, GermanyDepartment of Pharmacology and ToxicologyJena University Hospital, Friedrich Schiller University, Jena, GermanyDepartments of NeurosurgeryMedicineImperial College, St Dunstans Road, London W6 8RP, UKDepartment of NeuropathologyInstitute for Pathology and NeuropathologyDepartment of NeurosurgeryUniversity of Tübingen, Tübingen, GermanyInstitute of PathologyTechnical University of Munich, München, Germany
| | - Elke Pulz
- Institute of PathologyHelmholtz Zentrum München, D-85764 Neuherberg, GermanyDepartment of Pharmacology and ToxicologyJena University Hospital, Friedrich Schiller University, Jena, GermanyDepartments of NeurosurgeryMedicineImperial College, St Dunstans Road, London W6 8RP, UKDepartment of NeuropathologyInstitute for Pathology and NeuropathologyDepartment of NeurosurgeryUniversity of Tübingen, Tübingen, GermanyInstitute of PathologyTechnical University of Munich, München, Germany
| | - Stefan Schulz
- Institute of PathologyHelmholtz Zentrum München, D-85764 Neuherberg, GermanyDepartment of Pharmacology and ToxicologyJena University Hospital, Friedrich Schiller University, Jena, GermanyDepartments of NeurosurgeryMedicineImperial College, St Dunstans Road, London W6 8RP, UKDepartment of NeuropathologyInstitute for Pathology and NeuropathologyDepartment of NeurosurgeryUniversity of Tübingen, Tübingen, GermanyInstitute of PathologyTechnical University of Munich, München, Germany
| | - Federico Roncaroli
- Institute of PathologyHelmholtz Zentrum München, D-85764 Neuherberg, GermanyDepartment of Pharmacology and ToxicologyJena University Hospital, Friedrich Schiller University, Jena, GermanyDepartments of NeurosurgeryMedicineImperial College, St Dunstans Road, London W6 8RP, UKDepartment of NeuropathologyInstitute for Pathology and NeuropathologyDepartment of NeurosurgeryUniversity of Tübingen, Tübingen, GermanyInstitute of PathologyTechnical University of Munich, München, Germany
| | - Natalia S Pellegata
- Institute of PathologyHelmholtz Zentrum München, D-85764 Neuherberg, GermanyDepartment of Pharmacology and ToxicologyJena University Hospital, Friedrich Schiller University, Jena, GermanyDepartments of NeurosurgeryMedicineImperial College, St Dunstans Road, London W6 8RP, UKDepartment of NeuropathologyInstitute for Pathology and NeuropathologyDepartment of NeurosurgeryUniversity of Tübingen, Tübingen, GermanyInstitute of PathologyTechnical University of Munich, München, Germany
| |
Collapse
|
37
|
Vilar L, Fleseriu M, Naves LA, Albuquerque JL, Gadelha PS, dos Santos Faria M, Nascimento GC, Montenegro RM, Montenegro RM. Can we predict long-term remission after somatostatin analog withdrawal in patients with acromegaly? Results from a multicenter prospective trial. Endocrine 2014; 46:577-84. [PMID: 24272601 DOI: 10.1007/s12020-013-0094-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2013] [Accepted: 10/17/2013] [Indexed: 10/26/2022]
Abstract
Somatostatin analogs (SSAs) represent the mainstay of therapy in acromegaly. One of the potential disadvantages is the expected need to maintain therapy indefinitely in previously non-irradiated patients. The aim of this multicenter prospective open trial was to evaluate the likelihood of successful discontinuation of SSA therapy in well-controlled acromegalic patients who fulfilled very strict criteria: two or more years of treatment with the long-acting SSA octreotide LAR (OCT-LAR), a stable dose and injections interval every 4 weeks or longer for the previous year, GH levels <2.5 ng/ml and normal IGF-1 levels for age, a tumor remnant <10 mm, no history of radiotherapy, and no use of cabergoline or pegvisomant over the previous 6 months. Disease recurrence was defined as an increase of IGF-1 to levels above 1.2-fold the upper limit of normal (ULN). Out of 220 patients, 20 patients (12 women and 8 men; mean age, 48.1 ± 10.3 years; age range, 27-64) treated for 2.74 ± 0.64 years (range, 2.0-4.4) were included in this prospective study and OCT-LAR therapy was stopped. Four patients (20 %) remained without clinical and biochemical/neuroradiological evidence of disease recurrence after 12-18 months of follow-up. Sixteen patients (80 %) relapsed biochemically within 9 months after drug withdrawal and restarted OCT-LAR at the same previous dose. Compared to recurring subjects, non-recurring patients had significantly lower mean IGF-1 (× ULN) levels but there were some overlapping values in both groups. No other characteristic could be identified as a predictor of successful OCT-LAR discontinuation. Our findings demonstrated that OCT-LAR withdrawal, though rare, is possible in well-selected acromegalic patients treated for at least 2 years and considered optimally controlled in hormonal and neuroradiological terms.
Collapse
Affiliation(s)
- Lucio Vilar
- Division of Endocrinology, Hospital das Clínicas, Federal University of Pernambuco, Rua Clovis Silveira Barros, 84/1202, Boa Vista, Recife, CEP 50050-270, Brazil,
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Cuevas-Ramos D, Fleseriu M. Somatostatin receptor ligands and resistance to treatment in pituitary adenomas. J Mol Endocrinol 2014; 52:R223-40. [PMID: 24647046 DOI: 10.1530/jme-14-0011] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Somatostatin (SST), an inhibitory polypeptide with two biologically active forms SST14 and SST28, inhibits GH, prolactin (PRL), TSH, and ACTH secretion in the anterior pituitary gland. SST also has an antiproliferative effect inducing cell cycle arrest and apoptosis. Such actions are mediated through five G-protein-coupled somatostatin receptors (SSTR): SSTR1-SSTR5. In GH-secreting adenomas, SSTR2 expression predominates, and somatostatin receptor ligands (SRLs; octreotide and lanreotide) directed to SSTR2 are presently the mainstays of medical therapy. However, about half of patients show incomplete biochemical remission, but the definition of resistance per se remains controversial. We summarize here the determinants of SRL resistance in acromegaly patients, including clinical, imaging features as well as molecular (mutations, SSTR variants, and polymorphisms), and histopathological (granulation pattern, and proteins and receptor expression) predictors. The role of SSTR5 may explain the partial responsiveness to SRLs in patients with adequate SSTR2 density in the cell membrane. In patients with ACTH-secreting pituitary adenomas, i.e. Cushing's disease (CD), SSTR5 is the most abundant receptor expressed and tumors show low SSTR2 density due to hypercortisolism-induced SSTR2 down-regulation. Clinical studies with pasireotide, a multireceptor-targeted SRL with increased SSTR5 activity, lead to approval of pasireotide for treatment of patients with CD. Other SRL delivery modes (oral octreotide), multireceptor-targeted SRL (somatoprim) or chimeric compounds targeting dopamine D2 receptors and SSTR2 (dopastatin), are briefly discussed.
Collapse
Affiliation(s)
- Daniel Cuevas-Ramos
- Department of MedicinePituitary Center, Cedars-Sinai Medical Center, Los Angeles, California, USANorthwest Pituitary Center and Departments of Medicine and Neurological SurgeryOregon Health and Science University, 3181 SW Sam Jackson Park Road (BTE 472), Portland, Oregon 97239, USA
| | - Maria Fleseriu
- Department of MedicinePituitary Center, Cedars-Sinai Medical Center, Los Angeles, California, USANorthwest Pituitary Center and Departments of Medicine and Neurological SurgeryOregon Health and Science University, 3181 SW Sam Jackson Park Road (BTE 472), Portland, Oregon 97239, USA
| |
Collapse
|
39
|
Abstract
INTRODUCTION Acromegaly is a rare disease that severely impacts patients' health all the while, being a slowly progressing illness. In the past decades, advancements in treatment modalities, especially development of new drugs, as well as focused guidelines has improved management of acromegaly. Still, many patients are considered not sufficiently treated and there remains an ongoing need for further development. AREAS COVERED This article reviews new medical treatments currently under clinical investigation (such as pasireotide, oral octreotide and somatoprim) and under experimental development (such as octreotide implants, CAM2029 and ATL-1103). EXPERT OPINION As it seems unlikely that one single agent may achieve cure in 100% of cases, there is an urgent need for new agents that help patients where current medication fails. Imperatively, this means we have to improve our understanding of the underlying pathogenetic and molecular mechanisms.
Collapse
Affiliation(s)
- Sylvère Störmann
- Klinikum der Universität München, Medizinische Klinik und Poliklinik IV , Ziemssenstr. 1, 80336 München , Germany +49 0 89 5160 2111 ; +49 0 89 5160 2194 ;
| | | |
Collapse
|
40
|
Nakamura Y, Ishii J, Kondo A. Bright fluorescence monitoring system utilizing Zoanthus sp. green fluorescent protein (ZsGreen) for human G-protein-coupled receptor signaling in microbial yeast cells. PLoS One 2013; 8:e82237. [PMID: 24340008 PMCID: PMC3855394 DOI: 10.1371/journal.pone.0082237] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Accepted: 10/22/2013] [Indexed: 11/18/2022] Open
Abstract
G-protein-coupled receptors (GPCRs) are currently the most important pharmaceutical targets for drug discovery because they regulate a wide variety of physiological processes. Consequently, simple and convenient detection systems for ligands that regulate the function of GPCR have attracted attention as powerful tools for new drug development. We previously developed a yeast-based fluorescence reporter ligand detection system using flow cytometry. However, using this conventional detection system, fluorescence from a cell expressing GFP and responding to a ligand is weak, making detection of these cells by fluorescence microscopy difficult. We here report improvements to the conventional yeast fluorescence reporter assay system resulting in the development of a new highly-sensitive fluorescence reporter assay system with extremely bright fluorescence and high signal-to-noise (S/N) ratio. This new system allowed the easy detection of GPCR signaling in yeast using fluorescence microscopy. Somatostatin receptor and neurotensin receptor (implicated in Alzheimer's disease and Parkinson's disease, respectively) were chosen as human GPCR(s). The facile detection of binding to these receptors by cognate peptide ligands was demonstrated. In addition, we established a highly sensitive ligand detection system using yeast cell surface display technology that is applicable to peptide screening, and demonstrate that the display of various peptide analogs of neurotensin can activate signaling through the neurotensin receptor in yeast cells. Our system could be useful for identifying lead peptides with agonistic activity towards targeted human GPCR(s).
Collapse
Affiliation(s)
- Yasuyuki Nakamura
- Department of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, Kobe, Japan
| | - Jun Ishii
- Organization of Advanced Science and Technology, Kobe University, Kobe, Japan
| | - Akihiko Kondo
- Department of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, Kobe, Japan
- * E-mail:
| |
Collapse
|
41
|
Nakamura Y, Takemoto N, Ishii J, Kondo A. Simultaneous method for analyzing dimerization and signaling of G-protein-coupled receptor in yeast by dual-color reporter system. Biotechnol Bioeng 2013; 111:586-96. [DOI: 10.1002/bit.25125] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2013] [Revised: 09/10/2013] [Accepted: 10/01/2013] [Indexed: 11/08/2022]
Affiliation(s)
- Yasuyuki Nakamura
- Department of Chemical Science and Engineering, Graduate School of Engineering; Kobe University; 1-1 Rokkodai, Nada Kobe 657-8501 Japan
| | - Norika Takemoto
- Department of Chemical Science and Engineering, Graduate School of Engineering; Kobe University; 1-1 Rokkodai, Nada Kobe 657-8501 Japan
| | - Jun Ishii
- Organization of Advanced Science and Technology; Kobe University; Kobe Japan
| | - Akihiko Kondo
- Department of Chemical Science and Engineering, Graduate School of Engineering; Kobe University; 1-1 Rokkodai, Nada Kobe 657-8501 Japan
| |
Collapse
|
42
|
Kharmate G, Rajput PS, Lin YC, Kumar U. Inhibition of tumor promoting signals by activation of SSTR2 and opioid receptors in human breast cancer cells. Cancer Cell Int 2013; 13:93. [PMID: 24059654 PMCID: PMC3852783 DOI: 10.1186/1475-2867-13-93] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Accepted: 08/23/2013] [Indexed: 12/31/2022] Open
Abstract
Background Somatostatin receptors (SSTRs) and opioid receptors (ORs) belong to the superfamily of G-protein coupled receptors and function as negative regulators of cell proliferation in breast cancer. In the present study, we determined the changes in SSTR subtype 2 (SSTR2) and μ, δ and κ-ORs expression, signaling cascades and apoptosis in three different breast cancer cells namely MCF-7, MDA-MB231 and T47D. Methods Immunocytochemistry and western blot analysis were employed to study the colocalization and changes in MAPKs (ERK1/2 and p38), cell survival pathway (PI3K/AKT) and tumor suppressor proteins (PTEN and p53) in breast cancer cell lines. The nature of cell death upon activation of SSTR2 or OR was analysed using flow cytometry analysis. Results The activation of SSTR2 and ORs modulate MAPKs (ERK1/2 and p38) in cell dependent and possibly estrogen receptor (ER) dependent manner. The activation of tumor suppressor proteins phosphatase and tensin homolog (PTEN) and p53 antagonized the PI3K/AKT cell survival pathway. Flow cytometry analyses reveal increased necrosis as opposed to apoptosis in MCF-7 and T47D cells when compared to ER negative MDA-MB231 cells. Furthermore, receptor and agonist dependent expression of ORs in SSTR2 immunoprecipitate suggest that SSTR2 and ORs might interact as heterodimers and inhibit epidermal growth factor receptor phosphorylation. Conclusion Taken together, findings indicate a new role for SSTR2/ORs in modulation of signaling pathways involved in cancer progression and provide novel therapeutic approaches in breast cancer treatment.
Collapse
Affiliation(s)
- Geetanjali Kharmate
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC V6T1Z3, Canada.
| | | | | | | |
Collapse
|
43
|
Pedro Becker C. Tratamiento médico de tumores hipofisarios. REVISTA MÉDICA CLÍNICA LAS CONDES 2013. [DOI: 10.1016/s0716-8640(13)70218-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
44
|
Dadzie DD, Lee EJ, Monteleone CA, Schneider SH. Desensitization treatment for hypersensitivity reaction to octreotide in an acromegalic patient. Pituitary 2012; 15 Suppl 1:S68-71. [PMID: 22618955 DOI: 10.1007/s11102-012-0400-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Octreotide is widely used as medical therapy for acromegaly. It is known to markedly reduce growth hormone levels, improve symptoms and reduce tumor size. Common side effects include gastrointestinal symptoms, hepatobiliary disorders, dizziness, headaches, bradycardia, hyperglycemia or hypoglycemia and thyroid dysfunction. Although urticaria, allergy/hypersensitivity reactions and anaphylaxis have been noted as possible adverse reactions, there is a lack of data showing a causal relationship between octreotide and hypersensitivity reactions and there is no information on management when continued use of this medication is essential. We now report a case of a 60 year old male with acromegaly who had presented with a cutaneous hypersensitivity reaction to octreotide. In addition he failed treatment with surgery, radiation, and dopamine agonist and could no longer afford to continue treatment with pegvisomant. The patient underwent desensitization treatment for his octreotide allergy and was able to resume treatment without any further side effects. We believe this case represents the first report of successful desensitization treatment for octreotide allergy in an acromegalic patient.
Collapse
Affiliation(s)
- Daphne D Dadzie
- Division of Endocrinology, Metabolism and Nutrition, UMDNJ-Robert Wood Johnson University Hospital, New Brunswick, NJ 08901, USA.
| | | | | | | |
Collapse
|
45
|
Mercieca C, Gruppetta M, Vassallo J. Epidemiology, treatment trends and outcomes of acromegaly. Eur J Intern Med 2012; 23:e206-7. [PMID: 22921824 DOI: 10.1016/j.ejim.2012.07.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2012] [Revised: 07/22/2012] [Accepted: 07/30/2012] [Indexed: 11/16/2022]
|
46
|
Abstract
Acromegaly is a chronic disease characterized by the presence of a pituitary growth hormone (GH)-producing tumour, excessive secretion of growth hormone, raised levels of insulin-like growth factor I (IGF-I) and characteristic clinical presentation of acral enlargement. Over the past two decades, major advances have occurred in the understanding of some aspects of acromegaly--such as the biology of pituitary tumours, the physiology, molecular mechanisms of GH secretion and IGF-I generation, and the pathogenesis of comorbidities. Moreover, new approaches to diagnosis and surveillance (both in terms of screening and follow-up) of acromegaly have led to increases in the number of patients diagnosed with active disease, many of whom would previously have been missed. The development of sensitive assays for detecting plasma GH and IGF-I levels, as well as the widespread use of MRI for visualization of small tumours, have been major contributing factors to these improvements. Treatment advances have resulted in improved cure rates and disease control through novel neurosurgical techniques and pharmacological approaches. This Review summarizes and discusses the changes in our understanding of the epidemiology, diagnosis, treatment, and follow-up of acromegaly and its comorbidities.
Collapse
Affiliation(s)
- Antônio Ribeiro-Oliveira
- Federal University of Minas Gerais, Department of Internal Medicine, Rua Alfredo Balena 110, Belo Horizonte, MG 30330-120, Brazil
| | | |
Collapse
|
47
|
Tutuncu Y, Berker D, Isik S, Ozuguz U, Akbaba G, Kucukler FK, Aydin Y, Guler S. Comparison of octreotide LAR and lanreotide autogel as post-operative medical treatment in acromegaly. Pituitary 2012; 15:398-404. [PMID: 21863263 PMCID: PMC3443341 DOI: 10.1007/s11102-011-0335-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Long-acting somatostatin analogs are frequently used as adjuvant treatment of acromegaly patients after noncurative surgery. This sudy aims to compare the efficacy of octreotide long-acting release (OCT) and lanreotide Autogel (LAN) in acromegaly patients. Sixty-eight patients not cured by transsphenoidal endoscopic or microscopic pituitary surgery between 2003 and 2009 were retrospectively analyzed (25 men; 43 women; mean age 41.1 ± 10.9 years [range 18-65 years]). The patients were assigned randomly to OCT (n = 36) and LAN (n = 32) groups. Evaluations included insulin-like growth factor I (IGF-I) and growth hormone (GH) after oral glucose tolerance test (OGTT) 3, 6, 12 and 18 months after starting medical treatment; pituitary magnetic resonance imaging was performed before treatment and after 3 and 12 months. Patients achieving IGF-I levels within the age and gender normal range and GH level <1 μg/l following OGTT were considered a 'biochemical cure'. Mean IGF-I and GH values and tumor volumes (cm(3)) in the LAN and OCT groups were similar in the post-operative period before initiation of medical treatment. A statistically significant decrease in GH and IGF-I levels was obtained for both treatment groups at each follow-up visit compared to the previous value. Tumor shrinkage after 12 months of treatment was statistically significant in both groups but the percentage tumor shrinkage (28.5% vs. 34.9%, P = 0.166) and rate of patients achieving biochemical cure (63.9 and 78.1%, P = 0.454) were similar between OCT and LAN groups, respectively. OCT and LAN treatment options have similar efficacy for ensuring biochemical cure and tumor shrinkage in acromegaly patients who had noncurative surgery.
Collapse
Affiliation(s)
- Yasemin Tutuncu
- Department of Endocrinology and Metabolism, Ministry of Health, Ankara Numune Research and Training Hospital, Ankara, Turkey.
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Moloney KJ, Mercado JU, Ludlam WH, Mayberg MR. Pasireotide (SOM230): a novel pituitary-targeted medical therapy for the treatment of patients with Cushing's disease. Expert Rev Endocrinol Metab 2012; 7:491-502. [PMID: 30780888 DOI: 10.1586/eem.12.49] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Cushing's disease (CD) is a rare and debilitating condition resulting from extended exposure to excessive glucocorticoids caused by an adrenocorticotropic hormone-secreting pituitary adenoma. First-line treatment for most patients with CD is trans-sphenoidal adenomectomy. Postsurgical remission remains problematic; however, due to the difficulty of removing the tumor. Until recently, there were no approved medical treatments for Cushing's syndrome, but recent data on pasireotide (SOM230; a novel somatostatin analog) demonstrate restored hormone levels and improvements in quality of life, with a safety profile similar to that of other somatostatin analogs, except for incidence of hyperglycemia. Pasireotide represents an exciting, novel, pituitary-targeted medical therapy for patients with CD who are not surgical candidates, or for those who experience postsurgical recurrence.
Collapse
Affiliation(s)
- Kelley J Moloney
- a Seattle Pituitary Center, Swedish Neuroscience Institute, Seattle, WA, USA
- c Seattle Pituitary Center, Swedish Neuroscience Institute, Seattle, WA, USA.
| | - Jennifer U Mercado
- a Seattle Pituitary Center, Swedish Neuroscience Institute, Seattle, WA, USA
| | | | - Marc R Mayberg
- a Seattle Pituitary Center, Swedish Neuroscience Institute, Seattle, WA, USA
| |
Collapse
|
49
|
Immunohistochemical Distribution of Somatostatin and Somatostatin Receptor Subtypes (SSTR1–5) in Hypothalamus of ApoD Knockout Mice Brain. J Mol Neurosci 2012; 48:684-95. [DOI: 10.1007/s12031-012-9792-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Accepted: 04/26/2012] [Indexed: 01/09/2023]
|
50
|
Somvanshi RK, Kumar U. Pathophysiology of GPCR Homo- and Heterodimerization: Special Emphasis on Somatostatin Receptors. Pharmaceuticals (Basel) 2012; 5:417-46. [PMID: 24281555 PMCID: PMC3763651 DOI: 10.3390/ph5050417] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Revised: 04/18/2012] [Accepted: 04/19/2012] [Indexed: 12/19/2022] Open
Abstract
G-protein coupled receptors (GPCRs) are cell surface proteins responsible for translating >80% of extracellular reception to intracellular signals. The extracellular information in the form of neurotransmitters, peptides, ions, odorants etc is converted to intracellular signals via a wide variety of effector molecules activating distinct downstream signaling pathways. All GPCRs share common structural features including an extracellular N-terminal, seven-transmembrane domains (TMs) linked by extracellular/intracellular loops and the C-terminal tail. Recent studies have shown that most GPCRs function as dimers (homo- and/or heterodimers) or even higher order of oligomers. Protein-protein interaction among GPCRs and other receptor proteins play a critical role in the modulation of receptor pharmacology and functions. Although ~50% of the current drugs available in the market target GPCRs, still many GPCRs remain unexplored as potential therapeutic targets, opening immense possibility to discover the role of GPCRs in pathophysiological conditions. This review explores the existing information and future possibilities of GPCRs as tools in clinical pharmacology and is specifically focused for the role of somatostatin receptors (SSTRs) in pathophysiology of diseases and as the potential candidate for drug discovery.
Collapse
Affiliation(s)
- Rishi K Somvanshi
- Faculty of Pharmaceutical Sciences, Division of Pharmacology and Toxicology, The University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.
| | | |
Collapse
|