1
|
Hu C, Chen L, Ding Y, Ye M, Tang Q. Metabolic changes in neuroendocrine neoplasms. Cell Mol Life Sci 2025; 82:205. [PMID: 40377669 PMCID: PMC12084448 DOI: 10.1007/s00018-025-05656-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 02/05/2025] [Accepted: 03/11/2025] [Indexed: 05/18/2025]
Abstract
Neuroendocrine neoplasms (NENs) are a group of highly heterogeneous neoplasms originating from neuroendocrine cells with a gradually increased incidence. Metabolic change is one of the recognized markers of tumor progression, which has been extensively and systematically studied in other malignant tumors. However, metabolic change in NENs has been relatively poorly studied, and systematic reviews are lacking. We reviewed the relationship between metabolic changes and NENs from the aspects of glucose metabolism, lipid metabolism, metabolic syndrome, amino acid metabolism and metabolomics, and discussed the potential therapeutic strategies of metabolic changes for NENs.
Collapse
Affiliation(s)
- Chunhua Hu
- Shanghai Key Laboratory of Gut Microecology and Associated Major Diseases Research, Digestive Disease Research and Clinical Translation Center, Department of Gastroenterology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Neuroendocrine Tumor Diagnosis and Treatment Center, Jiangsu Province Hospital, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Lingyi Chen
- Neuroendocrine Tumor Diagnosis and Treatment Center, Jiangsu Province Hospital, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Yi Ding
- Neuroendocrine Tumor Diagnosis and Treatment Center, Jiangsu Province Hospital, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Mujie Ye
- Neuroendocrine Tumor Diagnosis and Treatment Center, Jiangsu Province Hospital, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China.
| | - Qiyun Tang
- Neuroendocrine Tumor Diagnosis and Treatment Center, Jiangsu Province Hospital, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China.
| |
Collapse
|
2
|
Porcel-Pastrana F, Montero-Hidalgo AJ, G-García ME, Gil-Duque I, Prats-Escribano A, Gahete MD, Sarmento-Cabral A, Luque RM, León-González AJ. Cellular and Molecular Evidence of the Synergistic Antitumour Effects of Hydroxytyrosol and Metformin in Prostate Cancer. Int J Mol Sci 2025; 26:1341. [PMID: 39941109 PMCID: PMC11818903 DOI: 10.3390/ijms26031341] [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] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 01/30/2025] [Accepted: 01/31/2025] [Indexed: 02/16/2025] Open
Abstract
Prostate cancer (PCa) is the tumour pathology with the second highest incidence among men worldwide. PCa is strongly influenced by obesity (OB), which increases its aggressiveness. Hence, some metabolic drugs like metformin have emerged as potential anti-tumour agents against several endocrine-related cancers. Likewise, a high adherence to the Mediterranean diet has been associated with lower rates of OB and a reduction in PCa aggressiveness since this diet contains phenolic bioactive compounds such as hydroxytyrosol (HT) that is mainly present in extra virgin olive oil. Thus, we decided to analyse the therapeutic potential of the combination of HT + metformin in different PCa cell models. Specifically, combinations of different doses of HT and metformin were evaluated by analysing the proliferation rate of LNCaP, 22Rv1, DU-145, and PC-3 cells using the SynergicFinder method. The results revealed a synergistic effect of HT + metformin in significantly reducing proliferation, especially in LNCaP cells. This anti-tumour effect of HT + metformin was also confirmed in migration and tumoursphere formation assays in LNCaP. The effects on the cell cycle and apoptosis were also assessed by flow-cytometry, and a cycle arrest in the G1 phase and an increase in late apoptosis were observed with the combination of HT + metformin. The phosphorylation levels of critical components of different oncogenic pathways were measured which revealed that the combination of HT + metformin significantly reduced the activity of multiple components of the MAPK, AKT, and TGF-β pathways. Overall, the combination of HT + metformin might represent a new therapeutic avenue for the management of PCa patients, an observation that certainly warrants further investigation through a well-designed clinical trial.
Collapse
Affiliation(s)
- Francisco Porcel-Pastrana
- Maimonides Institute for Biomedical Research of Córdoba (IMIBIC), 14004 Córdoba, Spain; (F.P.-P.); (A.J.M.-H.); (M.E.G.-G.); (I.G.-D.); (A.P.-E.); (M.D.G.); (A.S.-C.)
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14014 Córdoba, Spain
- Hospital Universitario Reina Sofía (HURS), 14004 Córdoba, Spain
| | - Antonio J. Montero-Hidalgo
- Maimonides Institute for Biomedical Research of Córdoba (IMIBIC), 14004 Córdoba, Spain; (F.P.-P.); (A.J.M.-H.); (M.E.G.-G.); (I.G.-D.); (A.P.-E.); (M.D.G.); (A.S.-C.)
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14014 Córdoba, Spain
- Hospital Universitario Reina Sofía (HURS), 14004 Córdoba, Spain
| | - Miguel E. G-García
- Maimonides Institute for Biomedical Research of Córdoba (IMIBIC), 14004 Córdoba, Spain; (F.P.-P.); (A.J.M.-H.); (M.E.G.-G.); (I.G.-D.); (A.P.-E.); (M.D.G.); (A.S.-C.)
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14014 Córdoba, Spain
- Hospital Universitario Reina Sofía (HURS), 14004 Córdoba, Spain
| | - Ignacio Gil-Duque
- Maimonides Institute for Biomedical Research of Córdoba (IMIBIC), 14004 Córdoba, Spain; (F.P.-P.); (A.J.M.-H.); (M.E.G.-G.); (I.G.-D.); (A.P.-E.); (M.D.G.); (A.S.-C.)
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14014 Córdoba, Spain
- Hospital Universitario Reina Sofía (HURS), 14004 Córdoba, Spain
| | - Antonio Prats-Escribano
- Maimonides Institute for Biomedical Research of Córdoba (IMIBIC), 14004 Córdoba, Spain; (F.P.-P.); (A.J.M.-H.); (M.E.G.-G.); (I.G.-D.); (A.P.-E.); (M.D.G.); (A.S.-C.)
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14014 Córdoba, Spain
- Hospital Universitario Reina Sofía (HURS), 14004 Córdoba, Spain
| | - Manuel D. Gahete
- Maimonides Institute for Biomedical Research of Córdoba (IMIBIC), 14004 Córdoba, Spain; (F.P.-P.); (A.J.M.-H.); (M.E.G.-G.); (I.G.-D.); (A.P.-E.); (M.D.G.); (A.S.-C.)
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14014 Córdoba, Spain
- Hospital Universitario Reina Sofía (HURS), 14004 Córdoba, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBERobn), 28019 Madrid, Spain
| | - André Sarmento-Cabral
- Maimonides Institute for Biomedical Research of Córdoba (IMIBIC), 14004 Córdoba, Spain; (F.P.-P.); (A.J.M.-H.); (M.E.G.-G.); (I.G.-D.); (A.P.-E.); (M.D.G.); (A.S.-C.)
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14014 Córdoba, Spain
- Hospital Universitario Reina Sofía (HURS), 14004 Córdoba, Spain
| | - Raúl M. Luque
- Maimonides Institute for Biomedical Research of Córdoba (IMIBIC), 14004 Córdoba, Spain; (F.P.-P.); (A.J.M.-H.); (M.E.G.-G.); (I.G.-D.); (A.P.-E.); (M.D.G.); (A.S.-C.)
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14014 Córdoba, Spain
- Hospital Universitario Reina Sofía (HURS), 14004 Córdoba, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBERobn), 28019 Madrid, Spain
| | - Antonio J. León-González
- Maimonides Institute for Biomedical Research of Córdoba (IMIBIC), 14004 Córdoba, Spain; (F.P.-P.); (A.J.M.-H.); (M.E.G.-G.); (I.G.-D.); (A.P.-E.); (M.D.G.); (A.S.-C.)
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14014 Córdoba, Spain
- Hospital Universitario Reina Sofía (HURS), 14004 Córdoba, Spain
- Department of Pharmacology, School of Pharmacy, University of Seville, 41012 Seville, Spain
| |
Collapse
|
3
|
Modica R, Liccardi A, Minotta R, Benevento E, Cannavale G, Di Iasi G, Colao A. Neuroendocrine Tumors and Survival Rates in Multiple Endocrine Neoplasia Type 1 Patients: Impact of Gender Difference. Neuroendocrinology 2024:1-7. [PMID: 39467535 DOI: 10.1159/000542143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 10/17/2024] [Indexed: 10/30/2024]
Abstract
INTRODUCTION Multiple endocrine neoplasia type 1 (MEN-1) is the most common inherited syndrome associated with NET development and gender-specific differences are emerging in neuroendocrine tumors (NETs). This study aimed to analyze gender difference in a single cohort of MEN-1 patients focusing on duodeno-pancreatic (DP)-NET and survival rates. METHODS MEN-1 patients referred to the Endocrinology Unit of the "Federico II" University of Naples, ENETS CoE, were retrospectively evaluated. RESULTS Among 100 MEN-1 patients enrolled, 59 (59%) were female and 41 (41%) male, and mean age at diagnosis was 39.4 years (range 5-86). No statistically significant association was identified between MEN-1 clinical manifestations and gender (primary hyperparathyroidism [PHPT] p: 1.0, DP-NET p: 0.83, pituitary adenoma [PA] p: 0.84, lung NET p: 0.64, and thymic NET p: 0.10), and similarly, age at diagnosis of MEN-1 and its individual manifestations was similar between genders. Survival analysis revealed no statistically significant difference between genders in DP-NET patients regarding progression disease p: 1.0 and death p: 1.0. Mean progression-free survival (PFS) of patients with DP-NET was 98.6 months (range 3-288), and mean overall survival (OS) was 130.1 months (range 3-444 months), without differences between genders (PFS p: 0.67 and OS p: 0.60). The Kaplan-Meier survival curves for PFS and OS showed no differences between genders (PFS p: 0.92; OS p: 0.87). CONCLUSION In this MEN-1 cohort, no gender differences in the occurrence of PHPT, PA, DP-NET, lung NET, and thymic NET, nor in survival outcomes including PFS and OS within the DP-NET subcohort, emerged. Therefore, this study supports similar screening and follow-up for both male and female MEN-1 patients.
Collapse
Affiliation(s)
- Roberta Modica
- Department of Clinical Medicine and Surgery, Endocrinology, Diabetology and Andrology Unit, Federico II University of Naples, Naples, Italy
| | - Alessia Liccardi
- Department of Clinical Medicine and Surgery, Endocrinology, Diabetology and Andrology Unit, Federico II University of Naples, Naples, Italy
| | - Roberto Minotta
- Department of Clinical Medicine and Surgery, Endocrinology, Diabetology and Andrology Unit, Federico II University of Naples, Naples, Italy
| | - Elio Benevento
- Department of Clinical Medicine and Surgery, Endocrinology, Diabetology and Andrology Unit, Federico II University of Naples, Naples, Italy
| | - Giuseppe Cannavale
- Department of Clinical Medicine and Surgery, Endocrinology, Diabetology and Andrology Unit, Federico II University of Naples, Naples, Italy
| | - Gianfranco Di Iasi
- Department of Clinical Medicine and Surgery, Endocrinology, Diabetology and Andrology Unit, Federico II University of Naples, Naples, Italy
| | - Annamaria Colao
- Department of Clinical Medicine and Surgery, Endocrinology, Diabetology and Andrology Unit, Federico II University of Naples, Naples, Italy,
- UNESCO Chair "Education for Health and Sustainable Development", Federico II University, Naples, Italy,
| |
Collapse
|
4
|
Modica R, La Salvia A, Liccardi A, Cozzolino A, Di Sarno A, Russo F, Colao A, Faggiano A. Dyslipidemia, lipid-lowering agents and neuroendocrine neoplasms: new horizons. Endocrine 2024; 85:520-531. [PMID: 38509261 PMCID: PMC11291585 DOI: 10.1007/s12020-024-03767-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 02/29/2024] [Indexed: 03/22/2024]
Abstract
PURPOSE Neuroendocrine neoplasms (NENs) are a heterogeneous group of malignancies originating from cells with a neuroendocrine phenotype. The complex relationship between lipid metabolism and cancer is gaining interest and a potential anti-cancer effect of lipid lowering agents is being considered. This review aims to discuss the current understanding and treatment of dyslipidaemia in NENs, focusing on the role of lipid lowering agents, including new therapeutic approaches, and future perspectives as possible tool in cancer prevention and tumor-growth control. METHODS We performed an electronic-based search using PubMed updated until December 2023, summarizing the available evidence both in basic and clinical research about lipid lowering agents in NENs. RESULTS Dyslipidemia is an important aspect to be considered in NENs management, although randomized studies specifically addressing this topic are lacking, unlike other cancer types. Available data mainly regard statins, and in vitro studies have demonstrated direct antitumor effects, including antiproliferative effects in some cancers, supporting possible pleiotropic effects also in NENs, but data remain conflicting. Ezetimibe, omega 3-fatty acids, fibrates and inhibitors of proprotein convertase subtilisin/kexin type 9 (PCSK9) may enhance the regulation of lipid homeostasis, as demonstrated in other cancers. CONCLUSIONS Targeting dyslipidemia in NENs should be part of the multidisciplinary management and an integrated approach may be the best option for both metabolic and tumor control. Whether lipid lowering agents may directly contribute to tumor control remains to be confirmed with specific studies, focusing on association with other metabolic risk, disease stage and primary site.
Collapse
Affiliation(s)
- Roberta Modica
- Endocrinology, Diabetology and Andrology Unit, Department of Clinical Medicine and Surgery, Federico II University of Naples, 80131, Naples, Italy.
| | - Anna La Salvia
- National Center for Drug Research and Evaluation, National Institute of Health (ISS), 00161, Rome, Italy
| | - Alessia Liccardi
- Endocrinology, Diabetology and Andrology Unit, Department of Clinical Medicine and Surgery, Federico II University of Naples, 80131, Naples, Italy
| | - Alessia Cozzolino
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Antonella Di Sarno
- Endocrinology, Diabetology and Andrology Unit, Department of Clinical Medicine and Surgery, Federico II University of Naples, 80131, Naples, Italy
| | - Flaminia Russo
- Endocrinology Unit, Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, ENETS Center of Excellence, Sapienza University of Rome, 00189, Rome, Italy
| | - Annamaria Colao
- Endocrinology, Diabetology and Andrology Unit, Department of Clinical Medicine and Surgery, Federico II University of Naples, 80131, Naples, Italy
- UNESCO Chair, Education for Health and Sustainable Development, Federico II University, 80131, Naples, Italy
| | - Antongiulio Faggiano
- Endocrinology Unit, Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, ENETS Center of Excellence, Sapienza University of Rome, 00189, Rome, Italy
| |
Collapse
|
5
|
Ruggeri RM, Altieri B, Razzore P, Retta F, Sperti E, Scotto G, Brizzi MP, Zumstein L, Pia A, Lania A, Lavezzi E, Nappo G, Laffi A, Albertelli M, Boschetti M, Hasballa I, Veresani A, Prinzi N, Pusceddu S, Oldani S, Nichetti F, Modica R, Minotta R, Liccardi A, Cannavale G, Grossrubatscher EM, Tarsitano MG, Zamponi V, Zatelli MC, Zanata I, Mazzilli R, Appetecchia M, Davì MV, Guarnotta V, Giannetta E, La Salvia A, Fanciulli G, Malandrino P, Isidori AM, Colao A, Faggiano A. Gender-related differences in patients with carcinoid syndrome: new insights from an Italian multicenter cohort study. J Endocrinol Invest 2024; 47:959-971. [PMID: 37837555 DOI: 10.1007/s40618-023-02213-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 09/25/2023] [Indexed: 10/16/2023]
Abstract
BACKGROUND The incidence of neuroendocrine neoplasm (NEN) and related carcinoid syndrome (CaS) has increased markedly in recent decades, and women appear to be more at risk than men. As per other tumors, gender may be relevant in influencing the clinical and prognostic characteristics of NEN-associated CS. However, specific data on carcinoid syndrome (CaS) are still lacking. PURPOSE To evaluate gender differences in clinical presentation and outcome of CaS. METHODS Retrospective analysis of 144 CaS patients from 20 Italian high-volume centers was conducted. Clinical presentation, tumor characteristics, therapies, and outcomes (progression-free survival, PFS, overall survival, OS) were correlated to gender. RESULTS Ninety (62.5%) CaS patients were male. There was no gender difference in the site of primary tumor, tumor grade and clinical stage, as well as in treatments. Men were more frequently smokers (37.2%) and alcohol drinkers (17.8%) than women (9.5%, p = 0.002, and 3.7%, p = 0.004, respectively). Concerning clinical presentation, women showed higher median number of symptoms (p = 0.0007), more frequent abdominal pain, tachycardia, and psychiatric disorders than men (53.3% vs 70.4%, p = 0.044; 6.7% vs 31.5%, p = 0.001; 50.9% vs. 26.7%, p = 0.003, respectively). Lymph node metastases at diagnosis were more frequent in men than in women (80% vs 64.8%; p = 0.04), but no differences in terms of PFS (p = 0.51) and OS (p = 0.64) were found between gender. CONCLUSIONS In this Italian cohort, CaS was slightly more frequent in males than females. Gender-related differences emerged in the clinical presentation of CaS, as well as gender-specific risk factors for CaS development. A gender-driven clinical management of these patients should be advisable.
Collapse
Affiliation(s)
- R M Ruggeri
- Endocrinology Unit, Department of Human Pathology of Adulthood and Childhood DETEV, University of Messina, 98125, Messina, Italy
| | - B Altieri
- Division of Endocrinology and Diabetes, Department of Internal Medicine, University of Würzburg, Würzburg, Germany
| | - P Razzore
- SC Endocrinologia, Diabetologia e Malattie del Metabolismo, AO Ordine Mauriziano di Torino, Largo Turati, 62 10128, Turin, Italy
| | - F Retta
- SC Endocrinologia, Diabetologia e Malattie del Metabolismo, AO Ordine Mauriziano di Torino, Largo Turati, 62 10128, Turin, Italy
| | - E Sperti
- SCDU Oncologia, AO Ordine Mauriziano di Torino, Largo Turati, 62 10128, Turin, Italy
| | - G Scotto
- Department of Oncology, University Hospital San Luigi Gonzaga, University of Turin, Orbassano, Turin, Italy
| | - M P Brizzi
- Department of Oncology, University Hospital San Luigi Gonzaga, University of Turin, Orbassano, Turin, Italy
| | - L Zumstein
- Department of Oncology, University Hospital San Luigi Gonzaga, University of Turin, Orbassano, Turin, Italy
| | - A Pia
- Internal Medicine, Department of Clinical and Biological Sciences, S. Luigi Hospital, University of Turin, Turin, Italy
| | - A Lania
- Department of Biomedical Sciences, Humanitas University, 20089, Pieve Emanuele, Italy
- Endocrinology, Diabetology and Andrology Unit, IRCCS Humanitas Research Hospital, 20089, Rozzano, Italy
| | - E Lavezzi
- Endocrinology, Diabetology and Andrology Unit, IRCCS Humanitas Research Hospital, 20089, Rozzano, Italy
| | - G Nappo
- Department of Biomedical Sciences, Humanitas University, 20089, Pieve Emanuele, Italy
- Pancreas Surgery Unit, IRCCS Humanitas Research Hospital, 20089, Rozzano, Italy
| | - A Laffi
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - M Albertelli
- Endocrinology Unit, Department of Internal Medicine and Medical Specialties (DIMI), University of Genova, 16132, Genoa, Italy
- Endocrinology Unit, IRCCC Ospedale Policlinico San Martino, 16132, Genoa, Italy
| | - M Boschetti
- Endocrinology Unit, Department of Internal Medicine and Medical Specialties (DIMI), University of Genova, 16132, Genoa, Italy
- Endocrinology Unit, IRCCC Ospedale Policlinico San Martino, 16132, Genoa, Italy
| | - I Hasballa
- Endocrinology Unit, Department of Internal Medicine and Medical Specialties (DIMI), University of Genova, 16132, Genoa, Italy
| | - A Veresani
- Endocrinology Unit, Department of Internal Medicine and Medical Specialties (DIMI), University of Genova, 16132, Genoa, Italy
| | - N Prinzi
- Medical Oncology, Foundation IRCCS National Cancer Institute, Milan, Italy
- First Department of Internal Medicine, San Matteo Hospital Foundation, Padua, Italy
| | - S Pusceddu
- Medical Oncology, Foundation IRCCS National Cancer Institute, Milan, Italy
| | - S Oldani
- Medical Oncology, Foundation IRCCS National Cancer Institute, Milan, Italy
| | - F Nichetti
- Medical Oncology, Foundation IRCCS National Cancer Institute, Milan, Italy
| | - R Modica
- Endocrinology, Diabetology and Andrology Unit, Department of Clinical Medicine and Surgery, Federico II University of Naples, Naples, Italy
| | - R Minotta
- Endocrinology, Diabetology and Andrology Unit, Department of Clinical Medicine and Surgery, Federico II University of Naples, Naples, Italy
| | - A Liccardi
- Endocrinology, Diabetology and Andrology Unit, Department of Clinical Medicine and Surgery, Federico II University of Naples, Naples, Italy
| | - G Cannavale
- Endocrinology, Diabetology and Andrology Unit, Department of Clinical Medicine and Surgery, Federico II University of Naples, Naples, Italy
| | | | - M G Tarsitano
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - V Zamponi
- Endocrinology Unit, Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, Sapienza University of Rome, ENETS Center of Excellence, Rome, Italy.
| | - M C Zatelli
- Section of Endocrinology, Geriatrics and Internal Medicine, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - I Zanata
- Section of Endocrinology, Geriatrics and Internal Medicine, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - R Mazzilli
- Endocrinology Unit, Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, Sapienza University of Rome, ENETS Center of Excellence, Rome, Italy
| | - M Appetecchia
- Oncological Endocrinology Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - M V Davì
- Department of Medicine, Section of Endocrinology, University and Hospital Trust of Verona, Verona, Italy
| | - V Guarnotta
- Dipartimento di Promozione della Salute, Materno-Infantile, Medicina Interna e Specialistica di Eccellenza "G. D'Alessandro" (PROMISE), Sezione di Malattie Endocrine, del Ricambio e della Nutrizione, Università di Palermo, 90127, Palermo, Italy
| | - E Giannetta
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - A La Salvia
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - G Fanciulli
- Neuroendocrine Tumour Unit, Department of Medicine, Surgery and Pharmacy, University of Sassari-Endocrine Unit, AOU Sassari, Sassari, Italy
| | - P Malandrino
- Endocrinology Unit, Department of Clinical and Experimental Medicine, University of Catania and Garibaldi, Nesima Medical Center, Catania, Italy
| | - A M Isidori
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - A Colao
- Endocrinology, Diabetology and Andrology Unit, Department of Clinical Medicine and Surgery, Federico II University of Naples, Naples, Italy
- UNESCO Chair on Health Education and Sustainable Development, Federico II University, 80138, Naples, Italy
| | - A Faggiano
- Endocrinology Unit, Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, Sapienza University of Rome, ENETS Center of Excellence, Rome, Italy
| |
Collapse
|
6
|
Cigrovski Berkovic M, Coppola A, Sesa V, Mrzljak A, Lai Q. Metformin and pancreatic neuroendocrine tumors: A systematic review and meta-analysis. World J Gastroenterol 2024; 30:759-769. [PMID: 38515954 PMCID: PMC10950617 DOI: 10.3748/wjg.v30.i7.759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 12/18/2023] [Accepted: 01/23/2024] [Indexed: 02/21/2024] Open
Abstract
BACKGROUND Most patients with advanced pancreatic neuroendocrine tumors (pNETs) die due to tumor progression. Therefore, identifying new therapies with low toxicity and good tolerability to use concomitantly with the established pNET treatment is relevant. In this perspective, metformin is emerging as a molecule of interest. Retrospective studies have described metformin, a widely used agent for the treatment of patients with type 2 diabetes mellitus (T2DM), to be effective in modulating different tumor-related events, including cancer incidence, recurrence and survival by inhibiting mTOR phosphorylation. This systematic review evaluates the role of T2DM and metformin in the insurgence and post-treatment outcomes in patients with pNET. AIM To systematically analyze and summarize evidence related to the diagnostic and prognostic value of T2DM and metformin for predicting the insurgence and post-treatment outcomes of pNET. METHODS A systematic review of the published literature was undertaken, focusing on the role of T2DM and metformin in insurgence and prognosis of pNET, measured through outcomes of tumor-free survival (TFS), overall survival and progression-free survival. RESULTS A total of 13 studies (5674 patients) were included in this review. Analysis of 809 pNET cases from five retrospective studies (low study heterogeneity with I² = 0%) confirms the correlation between T2DM and insurgence of pNET (OR = 2.13, 95%CI = 1.56-4.55; P < 0.001). The pooled data from 1174 pNET patients showed the correlation between T2DM and post-treatment TFS in pNET patients (hazard ratio = 1.84, 95%CI = 0.78-2.90; P < 0.001). The study heterogeneity was intermediate, with I² = 51%. A few studies limited the possibility of performing pooled analysis in the setting of metformin; therefore, results were heterogeneous, with no statistical relevance to the use of this drug in the diagnosis and prognosis of pNET. CONCLUSION T2DM represents a risk factor for the insurgence of pNET and is a significant predictor of poor post-treatment TFS of pNET patients. Unfortunately, a few studies with heterogeneous results limited the possibility of exploring the effect of metformin in the diagnosis and prognosis of pNET.
Collapse
Affiliation(s)
- Maja Cigrovski Berkovic
- Department for Sport and Exercise Medicine, Faculty of Kinesiology University of Zagreb, Zagreb 10000, Croatia
| | | | - Vibor Sesa
- Department of Gastroenterology and Hepatology, Liver Transplant Center, University Hospital Centre Zagreb, Zagreb 10000, Croatia
| | - Anna Mrzljak
- Department of Gastroenterology and Hepatology, Liver Transplant Center, University Hospital Centre Zagreb, Zagreb 10000, Croatia
- Department of Medicine, School of Medicine, Zagreb 10000, Croatia
| | - Quirino Lai
- General Surgery and Organ Transplantation Unit, Department of Surgery, Sapienza University of Rome, Rome 00018, Italy
| |
Collapse
|
7
|
Herrera-Martínez AD, Castillo-Peinado LLS, Molina-Puerta MJ, Calañas-Continente A, Membrives A, Castilla J, Camacho Cardenosa M, Casado-Díaz A, Gálvez-Moreno MA, Gahete MD, Quesada Gómez JM, Bouillon R, Priego-Capote F, Luque RM. Bariatric surgery and calcifediol treatment, Gordian knot of severe-obesity-related comorbidities treatment. Front Endocrinol (Lausanne) 2023; 14:1243906. [PMID: 37867510 PMCID: PMC10588639 DOI: 10.3389/fendo.2023.1243906] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 08/11/2023] [Indexed: 10/24/2023] Open
Abstract
Background Obesity (OB) is a chronic metabolic disease with important associated comorbidities and mortality. Vitamin D supplementation is frequently administered after bariatric surgery (BS), so as to reduce OB-related complications, maybe including chronic inflammation. Aim This study aimed to explore relations between vitamin D metabolites and components of the inflammasome machinery in OB before and after BS and their relations with the improvement of metabolic comorbidities. Patients and methods Epidemiological/clinical/anthropometric/biochemical evaluation was performed in patients with OB at baseline and 6 months after BS. Evaluation of i) vitamin-D metabolites in plasma and ii) components of the inflammasome machinery and inflammatory-associated factors [NOD-like-receptors (NLRs), inflammasome-activation-components, cytokines and inflammation/apoptosis-related components, and cell-cycle and DNA-damage regulators] in peripheral blood mononuclear cells (PBMCs) was performed at baseline and 6 months after BS. Clinical and molecular correlations/associations were analyzed. Results Significant correlations between vitamin D metabolites and inflammasome-machinery components were observed at baseline, and these correlations were significantly reduced 6 months after BS in parallel to a decrease in inflammation markers, fat mass, and body weight. Treatment with calcifediol remarkably increased 25OHD levels, despite 24,25(OH)2D3 remained stable after BS. Several inflammasome-machinery components were associated with improvement in metabolic comorbidities, especially hypertension and dyslipidemia. Conclusion The beneficial effects of vitamin D on OB-related comorbidities after BS patients are associated with significant changes in the molecular expression of key inflammasome-machinery components. The expression profile of these inflammasome components can be dynamically modulated in PBMCs after BS and vitamin D supplementation, suggesting that this profile could likely serve as a sensor and early predictor of the reversal of OB-related complications after BS.
Collapse
Affiliation(s)
- Aura D. Herrera-Martínez
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
- Endocrinology and Nutrition Service, Reina Sofia University Hospital, Córdoba, Spain
| | - Laura L. S. Castillo-Peinado
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
- Department of Analytical Chemistry, University of Córdoba, Córdoba, Spain
- Chemical Institute for Energy and Environment (IQUEMA), University of Córdoba, Córdoba, Spain
- CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - María J. Molina-Puerta
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
- Endocrinology and Nutrition Service, Reina Sofia University Hospital, Córdoba, Spain
| | - Alfonso Calañas-Continente
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
- Endocrinology and Nutrition Service, Reina Sofia University Hospital, Córdoba, Spain
| | - Antonio Membrives
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
- General Surgery Service, Reina Sofia University Hospital, Córdoba, Spain
| | - Juan Castilla
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
- General Surgery Service, Reina Sofia University Hospital, Córdoba, Spain
| | | | - Antonio Casado-Díaz
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
- CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - María A. Gálvez-Moreno
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
- Endocrinology and Nutrition Service, Reina Sofia University Hospital, Córdoba, Spain
| | - Manuel D. Gahete
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain
| | - José Manuel Quesada Gómez
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
- Endocrinology and Nutrition Service, Reina Sofia University Hospital, Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain
| | - Roger Bouillon
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, Catholic University of Leuven, Leuven, Belgium
| | - Feliciano Priego-Capote
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
- Department of Analytical Chemistry, University of Córdoba, Córdoba, Spain
- Chemical Institute for Energy and Environment (IQUEMA), University of Córdoba, Córdoba, Spain
- CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Raúl M. Luque
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain
| |
Collapse
|
8
|
Zhang LY, Yin YH, Wang XJ. Advances in the mechanism of action of metformin in pituitary tumors. World J Meta-Anal 2023; 11:144-150. [DOI: 10.13105/wjma.v11.i5.144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/20/2023] [Accepted: 04/14/2023] [Indexed: 06/16/2023] Open
|
9
|
Fuentes-Fayos AC, G-García ME, Pérez-Gómez JM, Montero-Hidalgo AJ, Martín-Colom J, Doval-Rosa C, Blanco-Acevedo C, Torres E, Toledano-Delgado Á, Sánchez-Sánchez R, Peralbo-Santaella E, Ortega-Salas RM, Jiménez-Vacas JM, Tena-Sempere M, López M, Castaño JP, Gahete MD, Solivera J, Luque RM. Metformin and simvastatin exert additive antitumour effects in glioblastoma via senescence-state: clinical and translational evidence. EBioMedicine 2023; 90:104484. [PMID: 36907105 PMCID: PMC10024193 DOI: 10.1016/j.ebiom.2023.104484] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 01/13/2023] [Accepted: 02/03/2023] [Indexed: 03/12/2023] Open
Abstract
BACKGROUND Glioblastoma is one of the most devastating and incurable cancers due to its aggressive behaviour and lack of available therapies, being its overall-survival from diagnosis ∼14-months. Thus, identification of new therapeutic tools is urgently needed. Interestingly, metabolism-related drugs (e.g., metformin/statins) are emerging as efficient antitumour agents for several cancers. Herein, we evaluated the in vitro/in vivo effects of metformin and/or statins on key clinical/functional/molecular/signalling parameters in glioblastoma patients/cells. METHODS An exploratory-observational-randomized retrospective glioblastoma patient cohort (n = 85), human glioblastoma/non-tumour brain human cells (cell lines/patient-derived cell cultures), mouse astrocytes progenitor cell cultures, and a preclinical xenograft glioblastoma mouse model were used to measure key functional parameters, signalling-pathways and/or antitumour progression in response to metformin and/or simvastatin. FINDINGS Metformin and simvastatin exerted strong antitumour actions in glioblastoma cell cultures (i.e., proliferation/migration/tumoursphere/colony-formation/VEGF-secretion inhibition and apoptosis/senescence induction). Notably, their combination additively altered these functional parameters vs. individual treatments. These actions were mediated by the modulation of key oncogenic signalling-pathways (i.e., AKT/JAK-STAT/NF-κB/TGFβ-pathways). Interestingly, an enrichment analysis uncovered a TGFβ-pathway activation, together with AKT inactivation, in response to metformin + simvastatin combination, which might be linked to an induction of the senescence-state, the associated secretory-phenotype, and to the dysregulation of spliceosome components. Remarkably, the antitumour actions of metformin + simvastatin combination were also observed in vivo [i.e., association with longer overall-survival in human, and reduction in tumour-progression in a mouse model (reduced tumour-size/weight/mitosis-number, and increased apoptosis)]. INTERPRETATION Altogether, metformin and simvastatin reduce aggressiveness features in glioblastomas, being this effect significantly more effective (in vitro/in vivo) when both drugs are combined, offering a clinically relevant opportunity that should be tested for their use in humans. FUNDING Spanish Ministry of Science, Innovation and Universities; Junta de Andalucía; CIBERobn (CIBER is an initiative of Instituto de Salud Carlos III, Spanish Ministry of Health, Social Services and Equality).
Collapse
Affiliation(s)
- Antonio C Fuentes-Fayos
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004, Cordoba, Spain; Department of Cell Biology, Physiology, 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.
| | - Miguel E G-García
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004, Cordoba, Spain; Department of Cell Biology, Physiology, 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
| | - Jesús M Pérez-Gómez
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004, Cordoba, Spain; Department of Cell Biology, Physiology, 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
| | - Antonio J Montero-Hidalgo
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004, Cordoba, Spain; Department of Cell Biology, Physiology, 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
| | - Julia Martín-Colom
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004, Cordoba, Spain; Department of Cell Biology, Physiology, Immunology, University of Cordoba, 14004, Cordoba, Spain; Department of Neurosurgery, Reina Sofia University Hospital, 14004, Cordoba, Spain
| | - Carlos Doval-Rosa
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004, Cordoba, Spain; Department of Cell Biology, Physiology, Immunology, University of Cordoba, 14004, Cordoba, Spain; Department of Neurosurgery, Reina Sofia University Hospital, 14004, Cordoba, Spain
| | - Cristóbal Blanco-Acevedo
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004, Cordoba, Spain; Department of Cell Biology, Physiology, Immunology, University of Cordoba, 14004, Cordoba, Spain; Department of Neurosurgery, Reina Sofia University Hospital, 14004, Cordoba, Spain
| | - Encarnación Torres
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004, Cordoba, Spain; Department of Cell Biology, Physiology, 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
| | - Álvaro Toledano-Delgado
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004, Cordoba, Spain; Department of Cell Biology, Physiology, Immunology, University of Cordoba, 14004, Cordoba, Spain; Department of Neurosurgery, Reina Sofia University Hospital, 14004, Cordoba, Spain
| | - Rafael Sánchez-Sánchez
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004, Cordoba, Spain; Department of Cell Biology, Physiology, Immunology, University of Cordoba, 14004, Cordoba, Spain; Pathology Service, Reina Sofia University Hospital, 14004, Cordoba, Spain
| | - Esther Peralbo-Santaella
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004, Cordoba, Spain; Flow Cytometry Unit, Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), 14004, Cordoba, Spain
| | - Rosa M Ortega-Salas
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004, Cordoba, Spain; Department of Cell Biology, Physiology, Immunology, University of Cordoba, 14004, Cordoba, Spain; Pathology Service, Reina Sofia University Hospital, 14004, Cordoba, Spain
| | - Juan M Jiménez-Vacas
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004, Cordoba, Spain; Department of Cell Biology, Physiology, 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
| | - Manuel Tena-Sempere
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004, Cordoba, Spain; Department of Cell Biology, Physiology, 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
| | - Miguel López
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004, Cordoba, Spain; NeurObesity Group, Department of Physiology, CiMUS, University of Santiago de Compostela, Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
| | - Justo P Castaño
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004, Cordoba, Spain; Department of Cell Biology, Physiology, 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
| | - Manuel D Gahete
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004, Cordoba, Spain; Department of Cell Biology, Physiology, 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
| | - Juan Solivera
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004, Cordoba, Spain; Department of Cell Biology, Physiology, Immunology, University of Cordoba, 14004, Cordoba, Spain; Department of Neurosurgery, Reina Sofia University Hospital, 14004, Cordoba, Spain
| | - Raúl M Luque
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004, Cordoba, Spain; Department of Cell Biology, Physiology, 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.
| |
Collapse
|
10
|
Vitale G, Carra S, Alessi Y, Campolo F, Pandozzi C, Zanata I, Colao A, Faggiano A. Carcinoid Syndrome: Preclinical Models and Future Therapeutic Strategies. Int J Mol Sci 2023; 24:ijms24043610. [PMID: 36835022 PMCID: PMC9961914 DOI: 10.3390/ijms24043610] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/27/2023] [Accepted: 02/08/2023] [Indexed: 02/15/2023] Open
Abstract
Carcinoid syndrome represents a debilitating paraneoplastic disease, caused by the secretion of several substances, occurring in about 10-40% of patients with well-differentiated neuroendocrine tumors (NETs). The main signs and symptoms associated with carcinoid syndrome are flushing, diarrhea, hypotension, tachycardia, bronchoconstriction, venous telangiectasia, dyspnea and fibrotic complications (mesenteric and retroperitoneal fibrosis, and carcinoid heart disease). Although there are several drugs available for the treatment of carcinoid syndrome, the lack of therapeutic response, poor tolerance or resistance to drugs are often reported. Preclinical models are indispensable tools for investigating the pathogenesis, mechanisms for tumor progression and new therapeutic approaches for cancer. This paper provides a state-of-the-art overview of in vitro and in vivo models in NETs with carcinoid syndrome, highlighting the future developments and therapeutic approaches in this field.
Collapse
Affiliation(s)
- Giovanni Vitale
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20122 Milan, Italy
- Laboratory of Geriatric and Oncologic Neuroendocrinology Research, IRCCS, Istituto Auxologico Italiano, 20100 Milan, Italy
- Correspondence: ; Tel.: +39-02-6191-12023; Fax: +39-02-6191-13033
| | - Silvia Carra
- Laboratory of Endocrine and Metabolic Research, IRCCS, Istituto Auxologico Italiano, 20100 Milan, Italy
| | - Ylenia Alessi
- Endocrine Unit, University Hospital “Gaetano Martino” of Messina, 98125 Messina, Italy
| | - Federica Campolo
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Carla Pandozzi
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Isabella Zanata
- Section of Endocrinology and Internal Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Annamaria Colao
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80138 Naples, Italy
| | - Antongiulio Faggiano
- Endocrinology Unit, Department of Clinical and Molecular Medicine, Sant’Andrea Hospital, ENETS Center of Excellence, Sapienza University of Rome, 00189 Rome, Italy
| | | |
Collapse
|
11
|
Cantone MC, Gaudenzi G, Carra S, Saronni D, Dicitore A. Obesity and GEP-NET: which to hit first? Minerva Med 2022; 113:913-915. [PMID: 36695409 DOI: 10.23736/s0026-4806.22.08314-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- Maria C Cantone
- Laboratory of Geriatric and Oncologic Neuroendocrinology Research, IRCCS Auxologic Italian Institute, Milan, Italy
| | - Germano Gaudenzi
- Laboratory of Geriatric and Oncologic Neuroendocrinology Research, IRCCS Auxologic Italian Institute, Milan, Italy
| | - Silvia Carra
- Laboratory of Endocrine and Metabolic Research, IRCCS Auxologic Italian Institute, Milan, Italy
| | - Davide Saronni
- Department of Medical Biotechnology and Translational Medicine, Translational Medicine (BIOMETRA), University of Milan, Milan, Italy
| | - Alessandra Dicitore
- Department of Medical Biotechnology and Translational Medicine, Translational Medicine (BIOMETRA), University of Milan, Milan, Italy -
| |
Collapse
|
12
|
Exploring the Relationship between Obesity, Metabolic Syndrome and Neuroendocrine Neoplasms. Metabolites 2022; 12:metabo12111150. [DOI: 10.3390/metabo12111150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/13/2022] [Accepted: 11/17/2022] [Indexed: 11/22/2022] Open
Abstract
Obesity is a major burden for modern medicine, with many links to negative health outcomes, including the increased incidence of certain cancer types. Interestingly, some studies have supported the concept of an “Obesity Paradox”, where some cancer patients living with obesity have been shown to have a better prognosis than non-obese patients. Neuroendocrine neoplasms (NENs) are malignancies originating from neuroendocrine cells, in some cases retaining important functional properties with consequences for metabolism and nutritional status. In this review, we summarize the existing evidence demonstrating that obesity is both a risk factor for developing NENs as well as a good prognostic factor. We further identify the limitations of existing studies and further avenues of research that will be necessary to optimize the metabolic and nutritional status of patients living with NENs to ensure improved outcomes.
Collapse
|
13
|
Hernandez-Rienda L, del Olmo-García MI, Merino-Torres JF. Impact of Diabetes Mellitus in Patients with Pancreatic Neuro-Endocrine Tumors: Causes, Consequences, and Future Perspectives. Metabolites 2022; 12:1103. [PMID: 36422243 PMCID: PMC9698930 DOI: 10.3390/metabo12111103] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/03/2022] [Accepted: 11/07/2022] [Indexed: 09/05/2023] Open
Abstract
Diabetes mellitus (DM) and pancreatic neuroendocrine tumors (pNETs) are two entities closely linked together. DM has been described as a risk factor for the development of pNETs and for the aggressiveness of the disease. On the other hand, DM due to pNETs is frequently undiagnosed or misclassified as type 2 DM when it is due to type 3 DM. In addition, metformin, a commonly prescribed drug for type 2 DM, has an antiproliferative property and is gaining increasing attention as an antitumor agent. This review article presents the findings published in the last few years on pNETs and DMs. Emphasis will be placed on DM as a risk factor, pNET as a risk factor for the development of type 3 DM, the management of type 3 DM on pNET, and DM as a prognostic factor in patients with pNET, as well as the future clinical implications of DM in these patients. The coexistence of DM and pNET is extensively presented. It is important to perform future clinical trials, which are necessary to establish the role of metformin on pNET disease. Increasing awareness among professionals managing pNET on the importance of a correct DM diagnosis and management of the disease must be a priority due to the implications on mortality and comorbidities it may have in these patients.
Collapse
Affiliation(s)
- Lorena Hernandez-Rienda
- Endocrinology and Nutrition Department, University and Politecnic Hospital La Fe, 46026 Valencia, Spain
- Joint Research Unit on Endocrinology, Nutrition and Clinical Dietetics, Health Research Institute Hospital La Fe-University of Valencia, 46026 Valencia, Spain
| | - Maria Isabel del Olmo-García
- Endocrinology and Nutrition Department, University and Politecnic Hospital La Fe, 46026 Valencia, Spain
- Joint Research Unit on Endocrinology, Nutrition and Clinical Dietetics, Health Research Institute Hospital La Fe-University of Valencia, 46026 Valencia, Spain
| | - Juan Francisco Merino-Torres
- Endocrinology and Nutrition Department, University and Politecnic Hospital La Fe, 46026 Valencia, Spain
- Joint Research Unit on Endocrinology, Nutrition and Clinical Dietetics, Health Research Institute Hospital La Fe-University of Valencia, 46026 Valencia, Spain
- Department of Medicine, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain
| |
Collapse
|
14
|
Morphofunctional and Molecular Assessment of Nutritional Status in Head and Neck Cancer Patients Undergoing Systemic Treatment: Role of Inflammasome in Clinical Nutrition. Cancers (Basel) 2022; 14:cancers14030494. [PMID: 35158762 PMCID: PMC8833422 DOI: 10.3390/cancers14030494] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 01/07/2022] [Accepted: 01/10/2022] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Malnutrition in patients with head and neck cancer is associated with worse clinical evolution and prognosis. Accurate nutritional assessments allow for early-identification of patients at risk of malnutrition. We aimed to perform a novel morphofunctional nutritional evaluation, including molecular analysis in patients with head and neck cancer who are undergoing systemic treatment. A morphofunctional nutritional assessment includes bioimpedance, anthropometric, ultrasound and biochemical measurements. We observed that malnutrition induces a profound alteration in the gene-expression pattern of inflammasome-machinery components, which are related with clinical nutritional parameters. This molecular analysis should be further studied as potential targets for nutrition-focused treatment strategies in cancer patients. Abstract Malnutrition in patients with head and neck cancer is frequent, multifactorial and widely associated with clinical evolution and prognosis. Accurate nutritional assessments allow for early identification of patients at risk of malnutrition in order to start nutritional support and prevent sarcopenia. We aimed to perform a novel morphofunctional nutritional evaluation and explore changes in inflammasome-machinery components in 45 patients with head and neck cancer who are undergoing systemic treatment. To this aim, an epidemiological/clinical/anthropometric/biochemical evaluation was performed. Serum RCP, IL6 and molecular expression of inflammasome-components and inflammatory-associated factors (NOD-like-receptors, inflammasome-activation-components, cytokines and inflammation/apoptosis-related components, cell-cycle and DNA-damage regulators) were evaluated in peripheral-blood mononuclear-cells (PBMCs). Clinical-molecular correlations/associations were analyzed. Coherent and complementary information was obtained in the morphofunctional nutritional assessment of the patients when bioimpedance, anthropometric and ultrasound data were analyzed. These factors were also correlated with different biochemical and molecular parameters, revealing the complementary aspect of the whole evaluation. Serum reactive C protein (RCP) and IL6 were the most reliable parameters for determining patients with decreased standardized phase angle, which is associated with increased mortality in patients with solid malignancies. Several inflammasome-components were dysregulated in patients with malnutrition, decreased phase angle and dependency grade or increased circulating inflammation markers. A molecular fingerprint based on gene-expression of certain inflammasome factors (p27/CCL2/ASC) in PBMCs accurately differentiated patients with and without malnutrition. In conclusion, malnutrition induces a profound alteration in the gene-expression pattern of inflammasome-machinery components in PBMCs. A comprehensive nutritional assessment including novel morphofunctional techniques and molecular markers allows a broad characterization of the nutritional status in cancer patients. Profile of certain inflammasome-components should be further studied as potential targets for nutrition-focused treatment strategies in cancer patients.
Collapse
|
15
|
Herrera-Martínez Y, Alzas Teomiro C, León Idougourram S, Molina Puertas MJ, Calañas Continente A, Serrano Blanch R, Castaño JP, Gálvez Moreno MÁ, Gahete MD, Luque RM, Herrera-Martínez AD. Sarcopenia and Ghrelin System in the Clinical Outcome and Prognosis of Gastroenteropancreatic Neuroendocrine Neoplasms. Cancers (Basel) 2021; 14:cancers14010111. [PMID: 35008278 PMCID: PMC8750458 DOI: 10.3390/cancers14010111] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/11/2021] [Accepted: 12/13/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Malnutrition and sarcopenia affect clinical outcomes in cancer patients. Nutritional evaluation in patients with neuroendocrine neoplasms (NENs) is not routinely performed. Currently, the evaluation of sarcopenia using CT scans is the gold standard in cancer patients, additionally, anthropometric, biochemical and molecular analysis of patients with gastroenteropancreatic NENs at diagnosis was perfomed. The expression levels of key ghrelin system components were assessed in 63 tumor samples. Results: Nutritional parameters were similar in GEP-NEN tumors of different origin. Relapsed disease was associated with decreased BMI. Patients who presented with weight loss at diagnosis had significantly lower overall survival (108 (25–302) vs. 263 (79–136) months). Ghrelin O-acyltransferase (GOAT) enzyme expression was higher in these patients. The prevalence of sarcopenia using CT images reached 87.2%. Mortality was observed only in patients with sarcopenia. Muscle evaluation was correlated with biochemical parameters but not with the expression of ghrelin system components. Conclusion: Survival is related to the nutritional status of patients with GEP-NENs and also to the molecular expression of some relevant ghrelin system components. Routine nutritional evaluation should be performed in these patients, in order to prescribe appropriate nutritional support, when necessary, for increasing quality of life and improving clinical outcomes. Abstract Background: Malnutrition and sarcopenia affect clinical outcomes and treatment response in cancer patients. Patients with neuroendocrine neoplasms (NENs) may present with additional symptoms related to tumor localization in the gastrointestinal tract and hormone secretion, increasing the risk and effects of sarcopenia. Aim: To explore the presence of malnutrition and sarcopenia in gastroenteropancreatic (GEP)-NEN patients, their relation to tumor characteristics, patient outcomes, survival and the molecular expression of ghrelin system components in the tumor. Patients and methods: One-hundred-and-four patients were included. Anthropometric, biochemical and CT-scans at diagnosis were evaluated. The expression levels of key ghrelin system components were assessed in 63 tumor samples. Results: Nutritional parameters were similar in GEP-NEN tumors of different origin. Relapsed disease was associated with decreased BMI. Patients who presented with weight loss at diagnosis had significantly lower overall survival (108 (25–302) vs. 263 (79–136) months). Ghrelin O-acyltransferase (GOAT) enzyme expression was higher in these patients. The prevalence of sarcopenia using CT images reached 87.2%. Mortality was observed only in patients with sarcopenia. Muscle evaluation was correlated with biochemical parameters but not with the expression of ghrelin system components. Conclusion: Survival is related to the nutritional status of patients with GEP-NENs and also to the molecular expression of some relevant ghrelin system components. Routine nutritional evaluation should be performed in these patients, in order to prescribe appropriate nutritional support, when necessary, for increasing quality of life and improving clinical outcomes.
Collapse
Affiliation(s)
| | - Carlos Alzas Teomiro
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Reina Sofia University Hospital, 14004 Cordova, Spain; (C.A.T.); (S.L.I.); (M.J.M.P.); (A.C.C.); (R.S.B.); (J.P.C.); (M.Á.G.M.); (M.D.G.); (R.M.L.)
- Endocrinology and Nutrition Service, Reina Sofia University Hospital, 14004 Cordova, Spain
| | - Soraya León Idougourram
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Reina Sofia University Hospital, 14004 Cordova, Spain; (C.A.T.); (S.L.I.); (M.J.M.P.); (A.C.C.); (R.S.B.); (J.P.C.); (M.Á.G.M.); (M.D.G.); (R.M.L.)
- Endocrinology and Nutrition Service, Reina Sofia University Hospital, 14004 Cordova, Spain
| | - María José Molina Puertas
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Reina Sofia University Hospital, 14004 Cordova, Spain; (C.A.T.); (S.L.I.); (M.J.M.P.); (A.C.C.); (R.S.B.); (J.P.C.); (M.Á.G.M.); (M.D.G.); (R.M.L.)
- Endocrinology and Nutrition Service, Reina Sofia University Hospital, 14004 Cordova, Spain
| | - Alfonso Calañas Continente
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Reina Sofia University Hospital, 14004 Cordova, Spain; (C.A.T.); (S.L.I.); (M.J.M.P.); (A.C.C.); (R.S.B.); (J.P.C.); (M.Á.G.M.); (M.D.G.); (R.M.L.)
- Endocrinology and Nutrition Service, Reina Sofia University Hospital, 14004 Cordova, Spain
| | - Raquel Serrano Blanch
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Reina Sofia University Hospital, 14004 Cordova, Spain; (C.A.T.); (S.L.I.); (M.J.M.P.); (A.C.C.); (R.S.B.); (J.P.C.); (M.Á.G.M.); (M.D.G.); (R.M.L.)
- Medical Oncology Service, Reina Sofia University Hospital, 14004 Cordova, Spain
| | - Justo P. Castaño
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Reina Sofia University Hospital, 14004 Cordova, Spain; (C.A.T.); (S.L.I.); (M.J.M.P.); (A.C.C.); (R.S.B.); (J.P.C.); (M.Á.G.M.); (M.D.G.); (R.M.L.)
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, 14014 Cordova, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, 14004 Cordova, Spain
| | - María Ángeles Gálvez Moreno
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Reina Sofia University Hospital, 14004 Cordova, Spain; (C.A.T.); (S.L.I.); (M.J.M.P.); (A.C.C.); (R.S.B.); (J.P.C.); (M.Á.G.M.); (M.D.G.); (R.M.L.)
- Endocrinology and Nutrition Service, Reina Sofia University Hospital, 14004 Cordova, Spain
| | - Manuel D. Gahete
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Reina Sofia University Hospital, 14004 Cordova, Spain; (C.A.T.); (S.L.I.); (M.J.M.P.); (A.C.C.); (R.S.B.); (J.P.C.); (M.Á.G.M.); (M.D.G.); (R.M.L.)
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, 14014 Cordova, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, 14004 Cordova, Spain
| | - Raúl M. Luque
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Reina Sofia University Hospital, 14004 Cordova, Spain; (C.A.T.); (S.L.I.); (M.J.M.P.); (A.C.C.); (R.S.B.); (J.P.C.); (M.Á.G.M.); (M.D.G.); (R.M.L.)
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, 14014 Cordova, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, 14004 Cordova, Spain
| | - Aura D. Herrera-Martínez
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Reina Sofia University Hospital, 14004 Cordova, Spain; (C.A.T.); (S.L.I.); (M.J.M.P.); (A.C.C.); (R.S.B.); (J.P.C.); (M.Á.G.M.); (M.D.G.); (R.M.L.)
- Endocrinology and Nutrition Service, Reina Sofia University Hospital, 14004 Cordova, Spain
- Correspondence:
| |
Collapse
|
16
|
Pusceddu S, Vernieri C, Di Maio M, Prinzi N, Torchio M, Corti F, Coppa J, Buzzoni R, Di Bartolomeo M, Milione M, Regnault B, Truong Thanh XM, Mazzaferro V, de Braud F. Impact of Diabetes and Metformin Use on Enteropancreatic Neuroendocrine Tumors: Post Hoc Analysis of the CLARINET Study. Cancers (Basel) 2021; 14:cancers14010069. [PMID: 35008233 PMCID: PMC8750688 DOI: 10.3390/cancers14010069] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/17/2021] [Accepted: 12/20/2021] [Indexed: 12/20/2022] Open
Abstract
The prognostic role of diabetes mellitus (DM) in advanced enteropancreatic neuroendocrine tumors (NETs) is unclear. Progression free survival (PFS) was assessed in post-hoc analyses of the 96-week, phase III, double-blind, placebo-controlled CLARINET study of lanreotide 120 mg in patients with advanced non-functional enteropancreatic NETs with DM (with/without metformin) and without DM. Of 204 patients, there were 79 with DM (lanreotide, n = 42 {metformin, n = 14}; placebo, n = 37 {metformin, n = 10}) and 125 without DM (lanreotide, n = 59; placebo, n = 66). Median PFS was 96.0 and 98.0 weeks with and without DM, respectively (hazard ratio 1.20 {95% confidence interval 0.79 to 1.82}; p = 0.380). No difference in PFS was observed in lanreotide-treated patients with/without DM (p = 0.8476). In the placebo group, median PFS was numerically shorter with versus without DM (p = 0.052) and was significantly longer in patients with DM and metformin (85.7 weeks) versus without metformin (38.7 weeks; p = 0.009). Multivariable Cox analyses showed that DM at baseline was not associated with PFS (p = 0.079); lanreotide was significantly associated with lower disease progression risk (p = 0.017). Lanreotide efficacy was confirmed in patients with advanced enteropancreatic NETs, regardless of diabetic status; DM was not a negative prognostic factor. A potential antitumor effect of metformin was observed in patients receiving placebo.
Collapse
Affiliation(s)
- Sara Pusceddu
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale Tumori (ENETS Center of Excellence), 20133 Milan, Italy; (C.V.); (N.P.); (M.T.); (F.C.); (M.D.B.); (M.M.); (F.d.B.)
- Correspondence:
| | - Claudio Vernieri
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale Tumori (ENETS Center of Excellence), 20133 Milan, Italy; (C.V.); (N.P.); (M.T.); (F.C.); (M.D.B.); (M.M.); (F.d.B.)
- IFOM, The FIRC Institute of Molecular Oncology, 20139 Milan, Italy
| | - Massimo Di Maio
- Department of Oncology, University of Turin, A.O. Ordine Mauriziano, 10124 Turin, Italy;
| | - Natalie Prinzi
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale Tumori (ENETS Center of Excellence), 20133 Milan, Italy; (C.V.); (N.P.); (M.T.); (F.C.); (M.D.B.); (M.M.); (F.d.B.)
| | - Martina Torchio
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale Tumori (ENETS Center of Excellence), 20133 Milan, Italy; (C.V.); (N.P.); (M.T.); (F.C.); (M.D.B.); (M.M.); (F.d.B.)
| | - Francesca Corti
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale Tumori (ENETS Center of Excellence), 20133 Milan, Italy; (C.V.); (N.P.); (M.T.); (F.C.); (M.D.B.); (M.M.); (F.d.B.)
| | - Jorgelina Coppa
- Department of Gastro-Entero-Pancreatic Surgical and Liver Transplantation, Fondazione IRCCS Istituto Nazionale Tumori (ENETS Center of Excellence), 20133 Milan, Italy;
| | - Roberto Buzzoni
- Oncology, Clinica San Carlo Paderno Dugnan, 20037 Milan, Italy;
| | - Maria Di Bartolomeo
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale Tumori (ENETS Center of Excellence), 20133 Milan, Italy; (C.V.); (N.P.); (M.T.); (F.C.); (M.D.B.); (M.M.); (F.d.B.)
| | - Massimo Milione
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale Tumori (ENETS Center of Excellence), 20133 Milan, Italy; (C.V.); (N.P.); (M.T.); (F.C.); (M.D.B.); (M.M.); (F.d.B.)
- First Division of Pathology, Department of Pathology and Laboratory Medicine, IRCCS Foundation National Cancer Institute, 20133 Milan, Italy
| | | | | | - Vincenzo Mazzaferro
- Department of Surgery, Fondazione IRCCS Istituto Nazionale Tumori (ENETS Center of Excellence), 20133 Milan, Italy;
- Oncology, Università Degli Studi di Milano, 20122 Milan, Italy
| | - Filippo de Braud
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale Tumori (ENETS Center of Excellence), 20133 Milan, Italy; (C.V.); (N.P.); (M.T.); (F.C.); (M.D.B.); (M.M.); (F.d.B.)
- Oncology, Università Degli Studi di Milano, 20122 Milan, Italy
| |
Collapse
|
17
|
Natalicchio A, Faggiano A, Zatelli MC, Argentiero A, D'Oronzo S, Marrano N, Beretta GD, Acquati S, Adinolfi V, Di Bartolo P, Danesi R, Ferrari P, Gori S, Morviducci L, Russo A, Tuveri E, Montagnani M, Gallo M, Silvestris N, Giorgino F. Metabolic disorders and gastroenteropancreatic-neuroendocrine tumors (GEP-NETs): How do they influence each other? An Italian Association of Medical Oncology (AIOM)/ Italian Association of Medical Diabetologists (AMD)/ Italian Society of Endocrinology (SIE)/ Italian Society of Pharmacology (SIF) multidisciplinary consensus position paper. Crit Rev Oncol Hematol 2021; 169:103572. [PMID: 34954047 DOI: 10.1016/j.critrevonc.2021.103572] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/14/2021] [Accepted: 12/16/2021] [Indexed: 12/13/2022] Open
Abstract
Gastroenteropancreatic neuroendocrine tumors (GEP-NETs) are a heterogeneous group of malignancies derived from neuroendocrine cells that can occur anywhere along the gastrointestinal tract. GEP-NETs incidence has been steadily increasing over the past decades, in parallel with the increasing incidence of the metabolic syndrome (MetS). It is not yet fully known whether the MetS components (such as obesity, dyslipidemia and type 2 diabetes) could be involved in the etiology of GEP-NETs or could influence their outcomes. In this review, a panel of experts of the Italian Association of Medical Oncology (AIOM), Italian Association of Medical Diabetologists (AMD), Italian Society of Endocrinology (SIE), and Italian Society of Pharmacology (SIF) provides a critical view of the experimental and clinical evidence about the association of GEP-NETs risk, outcomes, and therapies with the metabolic disorders typical of MetS. The potential therapeutic strategies for an optimal management of patients with both GEP-NETs and MetS are also discussed.
Collapse
Affiliation(s)
- Annalisa Natalicchio
- Department of Emergency and Organ Transplantation, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy.
| | - Antongiulio Faggiano
- Endocrinology Unit, Department of Clinical & Molecular Medicine, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy.
| | - Maria Chiara Zatelli
- Section of Endocrinology & Internal Medicine, Department of Medical Sciences, University of Ferrara, Ferrara, Italy.
| | | | - Stella D'Oronzo
- Department of Biomedical Sciences and Human Oncology, University of Bari Aldo Moro, Bari, Italy.
| | - Nicola Marrano
- Department of Emergency and Organ Transplantation, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy.
| | | | - Silvia Acquati
- Endocrinology Unit, Ospedale Pierantoni-Morgagni, Forlì, Italy.
| | - Valerio Adinolfi
- Endocrinology and Diabetology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy.
| | - Paolo Di Bartolo
- Diabetology Clinic, Rete Clinica di Diabetologia Aziendale - Dipartimento, Internistico di Ravenna - AUSL Romagna, Ravenna, Italy.
| | - Romano Danesi
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Italy.
| | - Pietro Ferrari
- Palliative Care Unit, Istituti Clinici Scientifici Maugeri SPA SB, IRCCS (PV), Italy.
| | - Stefania Gori
- Oncologia Medica, IRCCS Ospedale Sacro Cuore Don Calabria di Negrar, Verona, Italy.
| | - Lelio Morviducci
- Diabetology and Nutrition Unit, Department of Medical Specialities, ASL Roma 1 - S. Spirito Hospital, Rome, Italy.
| | - Antonio Russo
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Italy.
| | - Enzo Tuveri
- Diabetology, Endocrinology and Metabolic Diseases Service, ATS Sardegna - ASSL Carbonia-Iglesias, Italy.
| | - Monica Montagnani
- Department of Biomedical Sciences and Human Oncology, University of Bari Aldo Moro, Bari, Italy.
| | - Marco Gallo
- Endocrinology and Metabolic Diseases Unit, AO SS Antonio e Biagio e Cesare Arrigo, Alessandria, Italy.
| | - Nicola Silvestris
- Department of Emergency and Organ Transplantation, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy; Department of Biomedical Sciences and Human Oncology, University of Bari Aldo Moro, Bari, Italy.
| | - Francesco Giorgino
- Department of Emergency and Organ Transplantation, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy.
| |
Collapse
|
18
|
Tulipano G. Integrated or Independent Actions of Metformin in Target Tissues Underlying Its Current Use and New Possible Applications in the Endocrine and Metabolic Disorder Area. Int J Mol Sci 2021; 22:13068. [PMID: 34884872 PMCID: PMC8658259 DOI: 10.3390/ijms222313068] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/18/2021] [Accepted: 11/29/2021] [Indexed: 12/14/2022] Open
Abstract
Metformin is considered the first-choice drug for type 2 diabetes treatment. Actually, pleiotropic effects of metformin have been recognized, and there is evidence that this drug may have a favorable impact on health beyond its glucose-lowering activity. In summary, despite its long history, metformin is still an attractive research opportunity in the field of endocrine and metabolic diseases, age-related diseases, and cancer. To this end, its mode of action in distinct cell types is still in dispute. The aim of this work was to review the current knowledge and recent findings on the molecular mechanisms underlying the pharmacological effects of metformin in the field of metabolic and endocrine pathologies, including some endocrine tumors. Metformin is believed to act through multiple pathways that can be interconnected or work independently. Moreover, metformin effects on target tissues may be either direct or indirect, which means secondary to the actions on other tissues and consequent alterations at systemic level. Finally, as to the direct actions of metformin at cellular level, the intracellular milieu cooperates to cause differential responses to the drug between distinct cell types, despite the primary molecular targets may be the same within cells. Cellular bioenergetics can be regarded as the primary target of metformin action. Metformin can perturb the cytosolic and mitochondrial NAD/NADH ratio and the ATP/AMP ratio within cells, thus affecting enzymatic activities and metabolic and signaling pathways which depend on redox- and energy balance. In this context, the possible link between pyruvate metabolism and metformin actions is extensively discussed.
Collapse
Affiliation(s)
- Giovanni Tulipano
- Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| |
Collapse
|
19
|
Thapi S, Baeg K, Kim MK, Gallagher EJ. Survival of Patients With Gastroenteropancreatic Neuroendocrine Tumors and Diabetes Mellitus. Pancreas 2021; 50:1293-1297. [PMID: 34860814 PMCID: PMC8647768 DOI: 10.1097/mpa.0000000000001911] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
OBJECTIVES Diabetes mellitus (DM) is associated with an increased risk of gastroenteropancreatic neuroendocrine tumors (GEP-NETs), but the association between DM and GEP-NET survival is unknown. We evaluated disease characteristics and survival in individuals with DM and GEP-NETs. METHODS Using the Surveillance, Epidemiology, and End Results registry linked to Medicare (SEER-Medicare) claims database, we examined sociodemographics, GEP-NET characteristics, and treatment in patients with and without DM before GEP-NET diagnosis. We compared survival using univariate and multivariate analyses. RESULTS We identified 1858 individuals with GEP-NETs: 478 (25.7%) with DM and 1380 (74.3%) without. Significant differences in race (P = 0.002) were found between the DM and non-DM groups. Compared with individuals without DM, those with DM had more gastric (9.7% vs 14.9%), duodenal (6.5% vs 10.0%), and pancreatic (17.0% vs 21.8%), and less jejunal/ileal (18.1% vs 12.8%) NETs (P < 0.0001). Patients with DM had earlier stages (stage I, 37.0%; stage IV, 30.8%) than those without (stage I, 30.6%; stage IV, 36.4%; P = 0.0012). We found no difference in survival (multivariate hazard ratio, 0.97; 95% confidence interval, 0.76-1.23) between groups. CONCLUSIONS Among patients with and without DM before GEP-NET diagnosis, we found differences in tumor location and stage, but not survival.
Collapse
Affiliation(s)
| | - Kiwoon Baeg
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Michelle K. Kim
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Emily J. Gallagher
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
- Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
20
|
León-González AJ, Jiménez-Vacas JM, Fuentes-Fayos AC, Sarmento-Cabral A, Herrera-Martínez AD, Gahete MD, Luque RM. Role of metformin and other metabolic drugs in the prevention and therapy of endocrine-related cancers. Curr Opin Pharmacol 2021; 60:17-26. [PMID: 34311387 DOI: 10.1016/j.coph.2021.06.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/08/2021] [Accepted: 06/15/2021] [Indexed: 12/11/2022]
Abstract
Metabolic syndrome is associated with chronic diseases, including type 2 diabetes, cardiovascular diseases, and cancer. This review summarizes the current evidence on the antitumor effects of some relevant drugs currently used to manage metabolic-related pathologies (i.e. insulin and its analogs, metformin, statins, etc.) in endocrine-related cancers including breast cancer, prostate cancer, pituitary cancer, ovarian cancer, and neuroendocrine neoplasms. Although current evidence does not provide a clear antitumor role of several of these drugs, metformin seems to be a promising chemopreventive and adjuvant agent in cancer management, modulating tumor cell metabolism and microenvironment, through both AMP-activated protein kinase-dependent and -independent mechanisms. Moreover, its combination with statins might represent a promising therapeutic strategy to tackle the progression of endocrine-related tumors. However, further studies are needed to endorse the clinical relevance of these drugs as adjuvants for cancer chemotherapy.
Collapse
Affiliation(s)
- Antonio J León-González
- Maimonides Institute of Biomedical Research of Cordoba, 14004 Cordoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14014 Cordoba, Spain; Reina Sofia University Hospital, 14004 Cordoba, Spain; CIBER Physiopathology of Obesity and Nutrition, 14004 Cordoba, Spain; Department of Pharmacology, School of Pharmacy, University of Seville, 41012 Seville, Spain
| | - Juan M Jiménez-Vacas
- Maimonides Institute of Biomedical Research of Cordoba, 14004 Cordoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14014 Cordoba, Spain; Reina Sofia University Hospital, 14004 Cordoba, Spain; CIBER Physiopathology of Obesity and Nutrition, 14004 Cordoba, Spain
| | - Antonio C Fuentes-Fayos
- Maimonides Institute of Biomedical Research of Cordoba, 14004 Cordoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14014 Cordoba, Spain; Reina Sofia University Hospital, 14004 Cordoba, Spain; CIBER Physiopathology of Obesity and Nutrition, 14004 Cordoba, Spain
| | - Andre Sarmento-Cabral
- Maimonides Institute of Biomedical Research of Cordoba, 14004 Cordoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14014 Cordoba, Spain; Reina Sofia University Hospital, 14004 Cordoba, Spain; CIBER Physiopathology of Obesity and Nutrition, 14004 Cordoba, Spain
| | - Aura D Herrera-Martínez
- Maimonides Institute of Biomedical Research of Cordoba, 14004 Cordoba, Spain; Reina Sofia University Hospital, 14004 Cordoba, Spain; CIBER Physiopathology of Obesity and Nutrition, 14004 Cordoba, Spain; Endocrinology and Nutrition Service, Reina Sofia University Hospital, Córdoba, Spain
| | - Manuel D Gahete
- Maimonides Institute of Biomedical Research of Cordoba, 14004 Cordoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14014 Cordoba, Spain; Reina Sofia University Hospital, 14004 Cordoba, Spain; CIBER Physiopathology of Obesity and Nutrition, 14004 Cordoba, Spain
| | - Raúl M Luque
- Maimonides Institute of Biomedical Research of Cordoba, 14004 Cordoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14014 Cordoba, Spain; Reina Sofia University Hospital, 14004 Cordoba, Spain; CIBER Physiopathology of Obesity and Nutrition, 14004 Cordoba, Spain.
| |
Collapse
|
21
|
Cunha Júnior AD, Bragagnoli AC, Costa FO, Carvalheira JBC. Repurposing metformin for the treatment of gastrointestinal cancer. World J Gastroenterol 2021; 27:1883-1904. [PMID: 34007128 PMCID: PMC8108031 DOI: 10.3748/wjg.v27.i17.1883] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/13/2021] [Accepted: 04/07/2021] [Indexed: 02/06/2023] Open
Abstract
Diabetes mellitus type 2 and cancer share many risk factors. The pleiotropic insulin-dependent and insulin-independent effects of metformin might inhibit pathways that are frequently amplified in neoplastic tissue. Particularly, modulation of inflammation, metabolism, and cell cycle arrest are potential therapeutic cancer targets utilized by metformin to boost the anti-cancer effects of chemotherapy. Studies in vitro and in vivo models have demonstrated the potential of metformin as a chemo- and radiosensitizer, besides its chemopreventive and direct therapeutic activity in digestive system (DS) tumors. Hence, these aspects have been considered in many cancer clinical trials. Case-control and cohort studies and associated meta-analyses have evaluated DS cancer risk and metformin usage, especially in colorectal cancer, pancreatic cancer, and hepatocellular carcinoma. Most clinical studies have demonstrated the protective role of metformin in the risk for DS cancers and survival rates. On the other hand, the ability of metformin to enhance the actions of chemotherapy for gastric and biliary cancers is yet to be investigated. This article reviews the current findings on the anti-cancer mechanisms of metformin and its apparatus from pre-clinical and ongoing studies in DS malignancies.
Collapse
Affiliation(s)
- Ademar Dantas Cunha Júnior
- Department of Internal Medicine, Division of Oncology, University of Campinas (UNICAMP), Campinas 13083-970, São Paulo, Brazil
| | | | - Felipe Osório Costa
- Department of Internal Medicine, Division of Oncology, University of Campinas (UNICAMP), Campinas 13083-970, São Paulo, Brazil
| | | |
Collapse
|
22
|
Ni K, Yang JY, Baeg K, Leiter AC, Mhango G, Gallagher EJ, Wisnivesky JP, Kim MK. Association between somatostatin analogues and diabetes mellitus in gastroenteropancreatic neuroendocrine tumor patients: A Surveillance, Epidemiology, and End Results-Medicare analysis of 5235 patients. Cancer Rep (Hoboken) 2021; 4:e1387. [PMID: 33835729 PMCID: PMC8551991 DOI: 10.1002/cnr2.1387] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/04/2021] [Accepted: 03/09/2021] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Gastroenteropancreatic neuroendocrine tumors (GEP-NETs) are increasingly common malignancies and tend to have favorable long-term prognoses. Somatostatin analogues (SSA) are a first-line treatment for many NETs. Short-term experiments suggest an association between SSAs and hyperglycemia. However, it is unknown whether there is a relationship between SSAs and clinically significant hyperglycemia causing development of diabetes mellitus (DM), a chronic condition with significant morbidity and mortality. AIM In this study, we aimed to compare risk of developing DM in patients treated with SSA vs no SSA treatment. METHODS AND RESULTS Using the Surveillance, Epidemiology, and End Results (SEER) database and linked Medicare claims (1991-2016), we identified patients age 65+ with no prior DM diagnosis and a GEP-NET in the stomach, small intestine, appendix, colon, rectum, or pancreas. We used χ2 tests to compare SSA-treated and SSA-untreated patients and multivariable Cox regression to assess risk factors for developing DM. Among 8464 GEP-NET patients, 5235 patients had no prior DM and were included for analysis. Of these, 784 (15%) patients received SSAs. In multivariable analysis, the hazard ratio of developing DM with SSA treatment was 1.19, which was not statistically significant (95% CI 0.95-1.49). Significant risk factors for DM included black race, Hispanic ethnicity, prior pancreatic surgery, prior chemotherapy, tumor size >2 cm, pancreas tumors, and higher Charlson scores. CONCLUSION DM was very common in GEP-NET patients, affecting 53% of our cohort. Despite prior studies suggesting an association between SSAs and hyperglycemia, our analysis found similar risk of DM in SSA-treated and SSA-untreated GEP-NET patients. Further studies are needed to better understand this relationship. As NET patients have increasingly prolonged survival, it is crucial to identify chronic conditions such as DM that these patients may be at elevated risk for.
Collapse
Affiliation(s)
- Katherine Ni
- Department of MedicineIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Jeong Yun Yang
- Department of MedicineIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Kiwoon Baeg
- Division of GastroenterologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Amanda C. Leiter
- Division of EndocrinologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Grace Mhango
- Division of GastroenterologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Emily J. Gallagher
- Division of EndocrinologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA,Tisch Cancer Institute at Mount SinaiIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Juan P. Wisnivesky
- Division of Pulmonary, Critical Care, and Sleep MedicineIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Michelle K. Kim
- Division of GastroenterologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| |
Collapse
|
23
|
Kusne YN, Kosiorek HE, Buras MR, Verona PM, Coppola KE, Rone KA, Cook CB, Karlin NJ. Implications of neuroendocrine tumor and diabetes mellitus on patient outcomes and care: a matched case-control study. Future Sci OA 2021; 7:FSO684. [PMID: 34046189 PMCID: PMC8147757 DOI: 10.2144/fsoa-2020-0190] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Aim: We aimed to determine the impact of diabetes mellitus (DM) on survival of patients with neuroendocrine tumors (NETs) and of NETs on glycemic control. Patients & methods: Patients with newly diagnosed NETs with/without DM were matched 1:1 by age, sex and diagnosis year (2005–2017), and survival compared (Kaplan–Meier and Cox proportional hazards). Mixed models compared hemoglobin A1c (HbA1c) and glucose during the year after cancer diagnosis. Results: Three-year overall survival was 72% (95% CI: 60–86%) for DM patients versus 80% (95% CI: 70–92%) for non-DM patients (p = 0.82). Hazard ratio was 1.33 (95% CI: 0.56–3.16; p = 0.51); mean DM HbA1c, 7.3%. Conclusion: DM did not adversely affect survival of patients with NET. NET and its treatment did not affect glycemic control. The aim of this study was to evaluate the effect of diabetes mellitus (DM) on survival of patients with neuroendocrine tumor (NET) and to determine whether NET affected glycemic control. From an institutional cancer registry, 118 patients with NET were identified and grouped by DM (n = 59) or no DM (n = 59). The two groups were matched by age, sex and year of NET diagnosis. DM did not decrease survival, and NET did not significantly affect glycemic control in patients with DM.
Collapse
Affiliation(s)
- Yael N Kusne
- Department of Internal Medicine, Mayo Clinic, Scottsdale 85259, Arizona
| | | | | | - Patricia M Verona
- Enterprise Technology Services, Mayo Clinic, Scottsdale 85259, Arizona
| | - Kyle E Coppola
- Mayo Clinic Cancer Center, Mayo Clinic, Scottsdale 85259, Arizona
| | - Kelley A Rone
- Division of Hematology & Medical Oncology, Mayo Clinic Hospital, Phoenix 85054, Arizona
| | - Curtiss B Cook
- Division of Endocrinology, Mayo Clinic, Scottsdale 85259, Arizona
| | - Nina J Karlin
- Mayo Clinic Cancer Center, Mayo Clinic, Scottsdale 85259, Arizona.,Division of Hematology & Medical Oncology, Mayo Clinic Hospital, Phoenix 85054, Arizona
| |
Collapse
|
24
|
Jiménez-Vacas JM, Herrero-Aguayo V, Montero-Hidalgo AJ, Sáez-Martínez P, Gómez-Gómez E, León-González AJ, Fuentes-Fayos AC, Yubero-Serrano EM, Requena-Tapia MJ, López M, Castaño JP, Gahete MD, Luque RM. Clinical, Cellular, and Molecular Evidence of the Additive Antitumor Effects of Biguanides and Statins in Prostate Cancer. J Clin Endocrinol Metab 2021; 106:e696-e710. [PMID: 33247590 DOI: 10.1210/clinem/dgaa877] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Indexed: 02/06/2023]
Abstract
CONTEXT Prostate cancer (PCa) is one of the leading causes of cancer-related death among the male population worldwide. Unfortunately, current medical treatments fail to prevent PCa progression in a high percentage of cases; therefore, new therapeutic tools to tackle PCa are urgently needed. Biguanides and statins have emerged as antitumor agents for several endocrine-related cancers. OBJECTIVE To evaluate: (1) the putative in vivo association between metformin and/or statins treatment and key tumor and clinical parameters and (2) the direct effects of different biguanides (metformin/buformin/phenformin), statins (atorvastatin/simvastatin/lovastatin), and their combination, on key functional endpoints and associated signalling mechanisms. METHODS An exploratory/observational retrospective cohort of patients with PCa (n = 75) was analyzed. Moreover, normal and tumor prostate cells (normal [RWPE-cells/primary prostate cell cultures]; tumor [LNCaP/22RV1/PC3/DU145 cell lines]) were used to measure proliferation/migration/tumorsphere-formation/signalling pathways. RESULTS The combination of metformin+statins in vivo was associated to lower Gleason score and longer biochemical recurrence-free survival. Moreover, biguanides and statins exerted strong antitumor actions (ie, inhibition of proliferation/migration/tumorsphere formation) on PCa cells, and that their combination further decreased; in addition, these functional parameters compared with the individual treatments. These actions were mediated through modulation of key oncogenic and metabolic signalling pathways (ie, AR/mTOR/AMPK/AKT/ERK) and molecular mediators (MKI67/cMYC/androgen receptor/cell-cycle inhibitors). CONCLUSIONS Biguanides and statins significantly reduced tumor aggressiveness in PCa, with this effect being more potent (in vitro and in vivo) when both compounds are combined. Therefore, given the demonstrated clinical safety of biguanides and statins, our results suggest a potential therapeutic role of these compounds, especially their combination, for the treatment of PCa.
Collapse
Affiliation(s)
- Juan M Jiménez-Vacas
- Maimonides I nstitute for Biomedical Research of Córdoba (IMIBIC), Cordoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Cordoba, Spain
- Hospital Universitario Reina Sofía (HURS), Cordoba, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, (CIBERobn), Cordoba, Spain
| | - Vicente Herrero-Aguayo
- Maimonides I nstitute for Biomedical Research of Córdoba (IMIBIC), Cordoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Cordoba, Spain
- Hospital Universitario Reina Sofía (HURS), Cordoba, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, (CIBERobn), Cordoba, Spain
| | - Antonio J Montero-Hidalgo
- Maimonides I nstitute for Biomedical Research of Córdoba (IMIBIC), Cordoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Cordoba, Spain
- Hospital Universitario Reina Sofía (HURS), Cordoba, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, (CIBERobn), Cordoba, Spain
| | - Prudencio Sáez-Martínez
- Maimonides I nstitute for Biomedical Research of Córdoba (IMIBIC), Cordoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Cordoba, Spain
- Hospital Universitario Reina Sofía (HURS), Cordoba, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, (CIBERobn), Cordoba, Spain
| | - Enrique Gómez-Gómez
- Maimonides I nstitute for Biomedical Research of Córdoba (IMIBIC), Cordoba, Spain
- Hospital Universitario Reina Sofía (HURS), Cordoba, Spain
- Urology Service, HURS/IMIBIC, Cordoba, Spain
| | - Antonio J León-González
- Maimonides I nstitute for Biomedical Research of Córdoba (IMIBIC), Cordoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Cordoba, Spain
- Hospital Universitario Reina Sofía (HURS), Cordoba, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, (CIBERobn), Cordoba, Spain
| | - Antonio C Fuentes-Fayos
- Maimonides I nstitute for Biomedical Research of Córdoba (IMIBIC), Cordoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Cordoba, Spain
- Hospital Universitario Reina Sofía (HURS), Cordoba, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, (CIBERobn), Cordoba, Spain
| | - Elena M Yubero-Serrano
- Maimonides I nstitute for Biomedical Research of Córdoba (IMIBIC), Cordoba, Spain
- Hospital Universitario Reina Sofía (HURS), Cordoba, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, (CIBERobn), Cordoba, Spain
- Unidad de Gestión Clinica Medicina Interna, Lipids and Atherosclerosis Unit, Reina Sofia University Hospital, Córdoba, Spain
| | - María J Requena-Tapia
- Maimonides I nstitute for Biomedical Research of Córdoba (IMIBIC), Cordoba, Spain
- Hospital Universitario Reina Sofía (HURS), Cordoba, Spain
- Urology Service, HURS/IMIBIC, Cordoba, Spain
| | - Miguel López
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, (CIBERobn), Cordoba, Spain
- NeurObesity Group, Department of Physiology, CiMUS, University of Santiago de Compostela, Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
| | - Justo P Castaño
- Maimonides I nstitute for Biomedical Research of Córdoba (IMIBIC), Cordoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Cordoba, Spain
- Hospital Universitario Reina Sofía (HURS), Cordoba, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, (CIBERobn), Cordoba, Spain
| | - Manuel D Gahete
- Maimonides I nstitute for Biomedical Research of Córdoba (IMIBIC), Cordoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Cordoba, Spain
- Hospital Universitario Reina Sofía (HURS), Cordoba, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, (CIBERobn), Cordoba, Spain
| | - Raúl M Luque
- Maimonides I nstitute for Biomedical Research of Córdoba (IMIBIC), Cordoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Cordoba, Spain
- Hospital Universitario Reina Sofía (HURS), Cordoba, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, (CIBERobn), Cordoba, Spain
| |
Collapse
|
25
|
Pusceddu S, Vernieri C, Prinzi N, Torchio M, Coppa J, Antista M, Niger M, Milione M, Giacomelli L, Corti F, Prisciandaro M, Monteleone M, Colombo E, Di Bartolomeo M, de Braud F. The potential role of metformin in the treatment of patients with pancreatic neuroendocrine tumors: a review of preclinical to clinical evidence. Therap Adv Gastroenterol 2020; 13:1756284820927271. [PMID: 32821286 PMCID: PMC7406937 DOI: 10.1177/1756284820927271] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 04/15/2020] [Indexed: 02/04/2023] Open
Abstract
The incidence of pancreatic neuroendocrine tumors (panNETs) has increased worldwide in the last two decades. Given the indolent nature of these tumors, several patients are diagnosed with metastatic disease, which partially impairs the long-term efficacy of currently available treatments and reduces survival rates. The search for new therapeutic strategies for cancer patients has pushed towards the retrospective analysis of studies involving patients who concomitantly received other drugs together with standard anticancer agents. In this light, several retrospective analyses have shown that metformin use is associated with improved prognosis in patients with different tumor types treated with standard antitumor agents. Metformin, the cornerstone oral agent for the treatment of type 2 diabetes, plays a role in modulating glucose cell metabolism. Its potential ability to interfere with tumors may derive from the tight relationship between metabolic reprogramming in cancer cells and tumor progression. Indications for metformin use as an anticancer drug result from pre-clinical and clinical observations. In particular, metformin use in diabetic patients with advanced panNETs has been associated with better progression-free survival in patients treated with somatostatin analogues with or without metformin.
Collapse
Affiliation(s)
| | - Claudio Vernieri
- Fondazione IRCCS Istituto Nazionale dei Tumori, ENETS Center of Excellence, Milan, Italy The FIRC Institute of Molecular Oncology (IFOM), Milan, Italy
| | - Natalie Prinzi
- Fondazione IRCCS Istituto Nazionale dei Tumori, ENETS Center of Excellence, Milan, Italy
| | - Martina Torchio
- Fondazione IRCCS Istituto Nazionale dei Tumori, ENETS Center of Excellence, Milan, Italy
| | - Jorgelina Coppa
- Fondazione IRCCS Istituto Nazionale dei Tumori, ENETS Center of Excellence, Milan, Italy
| | - Maria Antista
- Fondazione IRCCS Istituto Nazionale dei Tumori, ENETS Center of Excellence, Milan, Italy
| | - Monica Niger
- Fondazione IRCCS Istituto Nazionale dei Tumori, ENETS Center of Excellence, Milan, Italy
| | - Massimo Milione
- Fondazione IRCCS Istituto Nazionale dei Tumori, ENETS Center of Excellence, Milan, Italy
| | - Luca Giacomelli
- Polistudium SRL (Milan, Italy) and Department of Surgical Sciences and Integrated Diagnostics, University of Genoa, Genoa, Italy
| | - Francesca Corti
- Fondazione IRCCS Istituto Nazionale dei Tumori, ENETS Center of Excellence, Milan, Italy
| | - Michele Prisciandaro
- Fondazione IRCCS Istituto Nazionale dei Tumori, ENETS Center of Excellence, Milan, Italy
| | - Michela Monteleone
- Fondazione IRCCS Istituto Nazionale dei Tumori, ENETS Center of Excellence, Milan, Italy
| | - Elena Colombo
- Fondazione IRCCS Istituto Nazionale dei Tumori, ENETS Center of Excellence, Milan, Italy
| | - Maria Di Bartolomeo
- Fondazione IRCCS Istituto Nazionale dei Tumori, ENETS Center of Excellence, Milan, Italy
| | - Filppo de Braud
- Fondazione IRCCS Istituto Nazionale dei Tumori, ENETS Center of Excellence, Milan, Italy University of Milan, Milan, Italy
| |
Collapse
|
26
|
Parween S, Rihs S, Flück CE. Metformin inhibits the activation of melanocortin receptors 2 and 3 in vitro: A possible mechanism for its anti-androgenic and weight balancing effects in vivo? J Steroid Biochem Mol Biol 2020; 200:105684. [PMID: 32360359 DOI: 10.1016/j.jsbmb.2020.105684] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 04/10/2020] [Accepted: 04/14/2020] [Indexed: 12/13/2022]
Abstract
Metformin is recommended as one of the first-line drugs for the treatment of type 2 diabetes and the metabolic syndrome. In addition to its insulin sensitizing effects, it has been shown to attenuate androgen excess in women with polycystic ovary syndrome (PCOS) or congenital adrenal hyperplasia (CAH), as well as to ameliorate obesity. The mechanisms of metformin action seem manifold. Preclinical studies suggest that it inhibits the cellular stress response at the level of the mitochondrial OXPHOS system and through AMPK dependent and independent mechanisms. Recent studies have shown that metformin decreases ACTH secretion from pituitary and reduces ACTH-stimulated adrenal secretion. In this study we investigated its specific effect through the melanocortin receptor 2 (MC2R) on signaling targeting adrenal steroidogenesis. To assess this effect, we used mouse adrenal OS3 cells, which do not express the MC2R. Cells were transfected with the MC2R and stimulated by ACTH. Downstream cyclic AMP production was then assessed by a co-transfected cAMP-responsive vector producing luciferase that was measured by a dual luciferase assay. The amount of luciferase produced in this assay corresponds to the amount of receptor activation with varying amount of ACTH. The effect of metformin was then tested in this system. We found a significant inhibition of ACTH induced MC2R activation and signaling with 10 mM metformin. The ACTH concentration response curve (CRC) was half-log shifted and a ∼30 % reduction in maximum receptor response (Rmax) to ACTH in presence of metformin was observed. This effect was dose dependent with an IC50 of 4.2 mM. qRT-PCR analyses showed that metformin decreased ACTH induced MC2R expression. Metformin did not affect cell viability and basal cAMP levels. We also tested the effect of metformin on homologous melanocortin receptors (MCRs). No significant effect was found on MC1R and MC4R activity. However, a log shift of EC50 of ACTH stimulation on MC3R was observed with metformin treatment. Metformin also inhibited melanocortin stimulating hormone (αMSH) induced MC3R activity. In conclusion, we show that metformin acts on MC2R and MC3R signaling directly. The role of MC2R for steroidogenesis is well established. MC3R is involved in energy balance and seems to act as a rheostat when the metabolism is challenged. Our study may explain how metformin helps in weight loss and attenuates the excess response to ACTH in androgen excess disorders such as PCOS and CAH.
Collapse
MESH Headings
- Adrenocorticotropic Hormone/pharmacology
- Androgen Antagonists/pharmacology
- Animals
- Cell Line
- Cell Survival/drug effects
- Hypoglycemic Agents/pharmacology
- Metformin/pharmacology
- Mice
- Receptor, Melanocortin, Type 2/antagonists & inhibitors
- Receptor, Melanocortin, Type 2/genetics
- Receptor, Melanocortin, Type 2/metabolism
- Receptor, Melanocortin, Type 3/antagonists & inhibitors
- Receptor, Melanocortin, Type 3/metabolism
- Weight Loss
Collapse
Affiliation(s)
- Shaheena Parween
- Pediatric Endocrinology, Diabetology, and Metabolism, Department of Pediatrics, University Children's Hospital Bern, 3010, Bern, Switzerland; Department of Biomedical Research, University of Bern, 3010, Bern, Switzerland
| | - Silvia Rihs
- Pediatric Endocrinology, Diabetology, and Metabolism, Department of Pediatrics, University Children's Hospital Bern, 3010, Bern, Switzerland; Department of Biomedical Research, University of Bern, 3010, Bern, Switzerland
| | - Christa E Flück
- Pediatric Endocrinology, Diabetology, and Metabolism, Department of Pediatrics, University Children's Hospital Bern, 3010, Bern, Switzerland; Department of Biomedical Research, University of Bern, 3010, Bern, Switzerland.
| |
Collapse
|
27
|
Lu M, Xing H, Cheng L, Liu H, Lang L, Yang T, Zhao X, Xu H, Ding P. A dual-functional buformin-mimicking poly(amido amine) for efficient and safe gene delivery. J Drug Target 2020; 28:923-932. [PMID: 32312081 DOI: 10.1080/1061186x.2020.1729770] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Biguanides (i.e. metformin, phenformin and buformin) are antidiabetic drugs with potential antitumor effects. Herein, a polycationic polymer, N,N'-bis(cystamine)acrylamide-buformin (CBA-Bu), containing multiple biodegradable disulphide bonds and buformin-mimicking side chains was synthesised. CBA-Bu was equipped with high efficiency and safety profile for gene delivery, meanwhile exhibiting potential antitumor efficacy. As a gene vector, CBA-Bu was able to condense plasmid DNA (pDNA) into nano-sized (<200 nm), positively-charged (>30 mV) uniform polyplexes that were well resistant to heparin and DNase I. Due to the reduction responsiveness of the disulphide bonds, CBA-Bu/pDNA polyplexes could release the loaded pDNA in the presence of dithiothreitol, and induce extremely low cytotoxicity in NIH/3T3 and U87 MG cells. The transfection results showed that CBA-Bu had a cellular uptake efficiency comparable to 25 kDa PEI, while a significantly higher gene expression level. Additionally, CBA-Bu had a lower IC50 value than its non-biguanide counterpart in two cancer cell lines. Furthermore, CBA-Bu could activate AMPK and inhibit mTOR pathways in U87 MG cells, a mechanism involved in the antitumor effect of biguanides. Taken together, CBA-Bu represented an advanced gene vector combining desirable gene delivery capability with potential antitumor activity, which was promising to achieve enhanced therapeutic efficacy in antitumor gene therapy.
Collapse
Affiliation(s)
- Mei Lu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Haonan Xing
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Lin Cheng
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Hui Liu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Lang Lang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Tianzhi Yang
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, Husson University, Bangor, Maine, USA
| | - Xiaoyun Zhao
- School of life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
| | - Hui Xu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Pingtian Ding
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
28
|
Scott AT, Weitz M, Breheny PJ, Ear PH, Darbro B, Brown BJ, Braun TA, Li G, Umesalma S, Kaemmer CA, Maharjan CK, Quelle DE, Bellizzi AM, Chandrasekharan C, Dillon JS, O'Dorisio TM, Howe JR. Gene Expression Signatures Identify Novel Therapeutics for Metastatic Pancreatic Neuroendocrine Tumors. Clin Cancer Res 2020; 26:2011-2021. [PMID: 31937620 PMCID: PMC7165057 DOI: 10.1158/1078-0432.ccr-19-2884] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 12/19/2019] [Accepted: 01/10/2020] [Indexed: 12/24/2022]
Abstract
PURPOSE Pancreatic neuroendocrine tumors (pNETs) are uncommon malignancies noted for their propensity to metastasize and comparatively favorable prognosis. Although both the treatment options and clinical outcomes have improved in the past decades, most patients will die of metastatic disease. New systemic therapies are needed. EXPERIMENTAL DESIGN Tissues were obtained from 43 patients with well-differentiated pNETs undergoing surgery. Gene expression was compared between primary tumors versus liver and lymph node metastases using RNA-Seq. Genes that were selectively elevated at only one metastatic site were filtered out to reduce tissue-specific effects. Ingenuity pathway analysis (IPA) and the Connectivity Map (CMap) identified drugs likely to antagonize metastasis-specific targets. The biological activity of top identified agents was tested in vitro using two pNET cell lines (BON-1 and QGP-1). RESULTS A total of 902 genes were differentially expressed in pNET metastases compared with primary tumors, 626 of which remained in the common metastatic profile after filtering. Analysis with IPA and CMap revealed altered activity of factors involved in survival and proliferation, and identified drugs targeting those pathways, including inhibitors of mTOR, PI3K, MEK, TOP2A, protein kinase C, NF-kB, cyclin-dependent kinase, and histone deacetylase. Inhibitors of MEK and TOP2A were consistently the most active compounds. CONCLUSIONS We employed a complementary bioinformatics approach to identify novel therapeutics for pNETs by analyzing gene expression in metastatic tumors. The potential utility of these drugs was confirmed by in vitro cytotoxicity assays, suggesting drugs targeting MEK and TOP2A may be highly efficacious against metastatic pNETs. This is a promising strategy for discovering more effective treatments for patients with pNETs.
Collapse
Affiliation(s)
- Aaron T Scott
- Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Michelle Weitz
- College of Public Health, Department of Biostatistics, University of Iowa, Iowa City, IA
| | - Patrick J Breheny
- College of Public Health, Department of Biostatistics, University of Iowa, Iowa City, IA
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA
| | - Po Hien Ear
- Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Benjamin Darbro
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA
- Stead Family Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Bart J Brown
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA
- Center for Bioinformatics and Computational Biology, College of Engineering, University of Iowa, Iowa City, IA
| | - Terry A Braun
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA
- Center for Bioinformatics and Computational Biology, College of Engineering, University of Iowa, Iowa City, IA
| | - Guiying Li
- Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Shaikamjad Umesalma
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Courtney A Kaemmer
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Chandra K Maharjan
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Dawn E Quelle
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA
- Department of Pathology, Carver College of Medicine University of Iowa, Iowa City, IA
| | - Andrew M Bellizzi
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA
- Department of Pathology, Carver College of Medicine University of Iowa, Iowa City, IA
| | - Chandrikha Chandrasekharan
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Joseph S Dillon
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Thomas M O'Dorisio
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - James R Howe
- Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA.
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA
| |
Collapse
|
29
|
de Mestier L, Védie AL, Faron M, Cros J, Rebours V, Hentic O, Do Cao C, Bardet P, Lévy P, Sauvanet A, Ruszniewski P, Hammel P. The Postoperative Occurrence or Worsening of Diabetes Mellitus May Increase the Risk of Recurrence in Resected Pancreatic Neuroendocrine Tumors. Neuroendocrinology 2020; 110:967-976. [PMID: 31791037 DOI: 10.1159/000505158] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Accepted: 11/29/2019] [Indexed: 12/12/2022]
Abstract
INTRODUCTION The goal of this retrospective study was to investigate the potential link between diabetes mellitus (DM) and the recurrence of pancreatic neuroendocrine tumors (PanNET) following curative intent surgery. METHODS We included patients who underwent surgical resection of nonmetastatic well-differentiated PanNET. Exacerbation of DM was defined as the postoperative occurrence of DM or worsening of preexisting DM. We explored the variables associated with PanNET recurrence-free survival (RFS). RFS was compared in a subset of patients with and without DM operated on by anatomical resection, after matching for the main prognostic factors. The impact of antidiabetic therapy on RFS was assessed. RESULTS A total of 268 patients (median age 54.7, 40% men) were included. Most PanNET were sporadic (85%), G1 (61%), pT1/pT2 (79%), and pN0 (76%). Postoperative DM exacerbation occurred in 38 patients (14%), including 27 with new-onset DM. On multivariable analysis, DM exacerbation was independently associated with an increased risk of PanNET recurrence (HR 2.35, 95% CI [1.24-4.47], p = 0.009) after adjustment for age, multiplicity of tumors, grade, pT, and pN stages. Similar results were found when 27 patients with and 48 patients without DM exacerbation, matched for grade, pT stage and pN stage, were compared (HR 3.03, 95% CI [1.05-8.77], p = 0.032). The postoperative use of metformin tended to decrease the risk of recurrence (HR 0.59, 95% CI 0.24-1.47, p = 0.26). CONCLUSION Patients with postoperative DM exacerbation may have an increased risk of PanNET recurrence. Closer follow-up might be beneficial in these patients. The protective role of metformin should be further explored.
Collapse
Affiliation(s)
- Louis de Mestier
- Department of Gastroenterology and Pancreatology, ENETS Centre of Excellence, Beaujon Hospital (APHP), Clichy, France,
- Université de Paris, Paris, France,
- INSERM U1149, Centre de Recherche sur l'Inflammation, Clichy/Paris, France,
| | - Anne-Laure Védie
- Department of Gastroenterology and Pancreatology, ENETS Centre of Excellence, Beaujon Hospital (APHP), Clichy, France
- Université de Paris, Paris, France
| | - Matthieu Faron
- Department of Surgical Oncology and Department of Biostatistics and Epidemiology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Jérôme Cros
- Université de Paris, Paris, France
- INSERM U1149, Centre de Recherche sur l'Inflammation, Clichy/Paris, France
- Department of Pathology, ENETS Centre of Excellence, Beaujon/Bichat Hospitals (APHP), Clichy/Paris, France
| | - Vinciane Rebours
- Department of Gastroenterology and Pancreatology, ENETS Centre of Excellence, Beaujon Hospital (APHP), Clichy, France
- Université de Paris, Paris, France
- INSERM U1149, Centre de Recherche sur l'Inflammation, Clichy/Paris, France
| | - Olivia Hentic
- Department of Gastroenterology and Pancreatology, ENETS Centre of Excellence, Beaujon Hospital (APHP), Clichy, France
| | - Christine Do Cao
- Department of Endocrinology, Diabetology and Metabolism, Lille University Hospital, Lille, France
| | - Pascal Bardet
- Department of Gastroenterology and Pancreatology, ENETS Centre of Excellence, Beaujon Hospital (APHP), Clichy, France
| | - Philippe Lévy
- Department of Gastroenterology and Pancreatology, ENETS Centre of Excellence, Beaujon Hospital (APHP), Clichy, France
- Université de Paris, Paris, France
| | - Alain Sauvanet
- Université de Paris, Paris, France
- Department of Hepato-Bilio-Pancreatic Surgery, ENETS Centre of Excellence, Beaujon/Bichat Hospitals (APHP), Clichy, France
| | - Philippe Ruszniewski
- Department of Gastroenterology and Pancreatology, ENETS Centre of Excellence, Beaujon Hospital (APHP), Clichy, France
- Université de Paris, Paris, France
- INSERM U1149, Centre de Recherche sur l'Inflammation, Clichy/Paris, France
| | - Pascal Hammel
- Université de Paris, Paris, France
- Department of Digestive Oncology, ENETS Centre of Excellence, Beaujon Hospital (APHP), Clichy, France
| |
Collapse
|
30
|
Zhuge X, Wang Y, Chen X, Guo C. Diabetes in Patients With Pancreatic Neuroendocrine Neoplasms. Front Endocrinol (Lausanne) 2020; 11:615082. [PMID: 33424776 PMCID: PMC7786385 DOI: 10.3389/fendo.2020.615082] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 11/18/2020] [Indexed: 01/18/2023] Open
Abstract
OBJECTIVE Diabetes mellitus (DM) is probably a risk factor for pancreatic neuroendocrine neoplasms (PNENs). However, the prevalence of DM in PNEN patients remains inconclusive. In the present study we observed the prevalence of DM and possible risk factors in PNEN patients. METHODS After excluding those with insulinoma, a total of 197 patients with PNENs were included. The demographic data, pathological characteristics, and data of blood biochemical tests were recorded. DM was considered if there was evidence of a fasting plasma glucose level of ≥7.0 mmol/L or a 2-h plasma glucose level of ≥11.1 mmol/L, or a history of DM at the time of PNEN diagnosis. Impaired fasting glucose was considered if fasting plasma glucose level was between 6.1 and 7.0 mmol/L. RESULTS The prevalence of DM, new-onset DM, and impaired fasting glucose were 17.26, 9.14, and 7.1%, respectively. The prevalence of DM was 26.0% in patients ≥60 years old (19/73) and 12.1% in patients <60 years old. Multivariable logistic regression analysis demonstrated that age, tumor size, and nerve invasion were independent risk factors for DM and impaired fasting glucose + DM (p < 0.05). Age, organs and nerve invasion were independent risk factors for impaired fasting glucose. Low high-density lipoprotein (HDL) was also a risk factor for incident of DM (OR = 0.15, 95%CI: 0.03-0.66). G2/G3 was an independent risk factor for DM in women. CONCLUSION Our data shows that the prevalence of DM is 17.26% in patients with PNENs and is 26.0% in patients ≥60 years of age after excluding insulinoma. Age, nerve invasion, tumor size, and HDL are risk factors for DM in PNEN patients.
Collapse
Affiliation(s)
- Xiaoling Zhuge
- Department of Laboratory Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yajie Wang
- Department of Radiology, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiao Chen
- Department of Radiology, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- *Correspondence: Xiao Chen, ; Chuangen Guo,
| | - Chuangen Guo
- Department of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Xiao Chen, ; Chuangen Guo,
| |
Collapse
|
31
|
Zhang X, Liu P, Shang Y, Kerndl H, Kumstel S, Gong P, Vollmar B, Zechner D. Metformin and LW6 impairs pancreatic cancer cells and reduces nuclear localization of YAP1. J Cancer 2020; 11:479-487. [PMID: 31897243 PMCID: PMC6930432 DOI: 10.7150/jca.33029] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 08/27/2019] [Indexed: 01/15/2023] Open
Abstract
The poor survival rate of pancreatic cancer is still a major challenge for the clinicians and their patients. In this study, we evaluated the efficacy of metformin, an inhibitor of oxidative phosphorylation, in combination with LW6, which impairs malate dehydrogenase 2 activities, in treating pancreatic cancer cells. We observed that this combinational therapy significantly reduced cell proliferation, migration, and significantly induced cell death when compared to cells treated by each monotherapy or Sham. In addition, we found that the combination of metformin and LW6 increased the phosphorylation of yes-associated protein 1 at serine 127 and attenuated the nuclear localization of this transcription factor. This combinatorial treatment also decreased the level of cellular yes-associated protein 1. This suggests that metformin in combination with LW6 impairs pancreatic cancer cells and reduces nuclear localization of yes-associated protein 1.
Collapse
Affiliation(s)
- Xianbin Zhang
- Institute for Experimental Surgery, Rostock University Medical Center, Schillingallee 69a, 18059, Rostock, Germany.,Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jiyan Road 440, 250117, Jinan, China
| | - Peng Liu
- Department of General Surgery, Shenzhen University General Hospital, Xueyuan Road 1098, 518055, Shenzhen, China
| | - Yuru Shang
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jiyan Road 440, 250117, Jinan, China.,Molecular Oncology and Immunotherapy, Department of General Surgery, Rostock University Medical Center, Schillingallee 69, 18059, Rostock, Germany
| | - Hagen Kerndl
- Institute for Experimental Surgery, Rostock University Medical Center, Schillingallee 69a, 18059, Rostock, Germany
| | - Simone Kumstel
- Institute for Experimental Surgery, Rostock University Medical Center, Schillingallee 69a, 18059, Rostock, Germany
| | - Peng Gong
- Department of General Surgery, Shenzhen University General Hospital, Xueyuan Road 1098, 518055, Shenzhen, China
| | - Brigitte Vollmar
- Institute for Experimental Surgery, Rostock University Medical Center, Schillingallee 69a, 18059, Rostock, Germany
| | - Dietmar Zechner
- Institute for Experimental Surgery, Rostock University Medical Center, Schillingallee 69a, 18059, Rostock, Germany
| |
Collapse
|
32
|
The anti-cancer effects of phenformin in thyroid cancer cell lines and in normal thyrocytes. Oncotarget 2019; 10:6432-6443. [PMID: 31741708 PMCID: PMC6849649 DOI: 10.18632/oncotarget.27266] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 09/24/2019] [Indexed: 12/26/2022] Open
Abstract
Phenformin is a biguanide drug which, besides the original anti-diabetic effect, also exerts anti-cancer effects. The aim of this study was to further characterize these latter in terms of both cell-viability and modulation of the secretion of the pro-tumorigenic chemokine CXCL8. Normal human thyrocytes in primary cultures (NHT) and thyroid cancer cell lines, TPC-1 and 8505C (RET/PTC and BRAFV600E mutated, respectively) were treated with increasing concentrations of phenformin at different times. Cell-viability was assessed by WST-1 and further characterized by AnnexinV/PI staining and cell proliferation colony-assay. CXCL8 levels were measured in cell supernatants. Phenformin reduced cell-viability in TPC-1 and 8505C and their ability to form colonies. In NHT cells, phenformin affected cell-viability only at the maximal dose but interestingly it inhibited CXCL8 secretion at all the concentrations not affecting cell-viability. Phenformin had no effect on CXCL8 secretion in thyroid cancer cell lines. Thus, phenformin exerts anti-cancer effects on both cancer cells (cell death induction) and surrounding normal cells (inhibition of CXCL8 secretion). These results highlight that the anti-cancer effects of phenformin are multifaceted and effective on both solid and soluble components of the tumor-microenvironment.
Collapse
|
33
|
Vernieri C, Pusceddu S, de Braud F. Impact of Metformin on Systemic Metabolism and Survival of Patients With Advanced Pancreatic Neuroendocrine Tumors. Front Oncol 2019; 9:902. [PMID: 31616628 PMCID: PMC6763756 DOI: 10.3389/fonc.2019.00902] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 08/30/2019] [Indexed: 12/22/2022] Open
Affiliation(s)
- Claudio Vernieri
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.,IFOM, The FIRC Institute of Molecular Oncology, Milan, Italy
| | - Sara Pusceddu
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Filippo de Braud
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.,Oncology and Hematology-Oncology Department, University of Milan, Milan, Italy
| |
Collapse
|
34
|
Vázquez-Borrego MC, Fuentes-Fayos AC, Herrera-Martínez AD, L-López F, Ibáñez-Costa A, Moreno-Moreno P, Alhambra-Expósito MR, Barrera-Martín A, Blanco-Acevedo C, Dios E, Venegas-Moreno E, Solivera J, Gahete MD, Soto-Moreno A, Gálvez-Moreno MA, Castaño JP, Luque RM. Biguanides Exert Antitumoral Actions in Pituitary Tumor Cells Through AMPK-Dependent and -Independent Mechanisms. J Clin Endocrinol Metab 2019; 104:3501-3513. [PMID: 30860580 DOI: 10.1210/jc.2019-00056] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 03/06/2019] [Indexed: 12/17/2022]
Abstract
CONTEXT Pituitary neuroendocrine tumors (PitNETs) are a commonly underestimated pathology in terms of incidence and associated morbimortality. Currently, an appreciable subset of patients are resistant or poorly responsive to the main current medical treatments [i.e., synthetic somatostatin analogs (SSAs) and dopamine agonists]. Thus, development and optimization of novel and available medical therapies is necessary. Biguanides (metformin, buformin, and phenformin) are antidiabetic drugs that exert antitumoral actions in several tumor types, but their pharmacological effects on PitNETs are poorly known. OBJECTIVE We aimed to explore the direct effects of biguanides on key functions (cell viability, hormone release, apoptosis, and signaling pathways) in primary cell cultures from human PitNETs and cell lines. Additionally, we evaluated the effect of combined metformin with SSAs on cell viability and hormone secretion. DESIGN A total of 13 corticotropinomas, 13 somatotropinomas, 13 nonfunctioning PitNETs, 3 prolactinomas, and 2 tumoral pituitary cell lines (AtT-20 and GH3) were used to evaluate the direct effects of biguanides on cell viability, hormone release, apoptosis, and signaling pathways. RESULTS Biguanides reduced cell viability in all PitNETs and cell lines (with phenformin being the most effective biguanide) and increased apoptosis in somatotropinomas. Moreover, buformin and phenformin, but not metformin, reduced hormone secretion in a cell type-specific manner. Combination metformin/SSA therapy did not increase SSA monotherapy effectiveness. Effects of biguanides on PitNETs could involve the modulation of AMP-activated protein kinase-dependent ([Ca2+]i, PI3K/Akt) and independent (MAPK) mechanisms. CONCLUSION Altogether, our data unveil clear antitumoral effects of biguanides on PitNET cells, opening avenues to explore their potential as drugs to treat these pathologies.
Collapse
Affiliation(s)
- Mari C Vázquez-Borrego
- Maimonides Institute of Biomedical Research of Cordoba, Cordoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain
- Reina Sofia University Hospital, Cordoba, Spain
- CIBER Physiopathology of Obesity and Nutrition, Cordoba, Spain
| | - Antonio C Fuentes-Fayos
- Maimonides Institute of Biomedical Research of Cordoba, Cordoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain
- Reina Sofia University Hospital, Cordoba, Spain
- CIBER Physiopathology of Obesity and Nutrition, Cordoba, Spain
| | - Aura D Herrera-Martínez
- Maimonides Institute of Biomedical Research of Cordoba, Cordoba, Spain
- Reina Sofia University Hospital, Cordoba, Spain
- Service of Endocrinology and Nutrition, IMIBIC, Reina Sofia University Hospital, Cordoba, Spain
| | - Fernando L-López
- Maimonides Institute of Biomedical Research of Cordoba, Cordoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain
- Reina Sofia University Hospital, Cordoba, Spain
- CIBER Physiopathology of Obesity and Nutrition, Cordoba, Spain
| | - Alejandro Ibáñez-Costa
- Maimonides Institute of Biomedical Research of Cordoba, Cordoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain
- Reina Sofia University Hospital, Cordoba, Spain
- CIBER Physiopathology of Obesity and Nutrition, Cordoba, Spain
| | - Paloma Moreno-Moreno
- Maimonides Institute of Biomedical Research of Cordoba, Cordoba, Spain
- Reina Sofia University Hospital, Cordoba, Spain
- Service of Endocrinology and Nutrition, IMIBIC, Reina Sofia University Hospital, Cordoba, Spain
| | - María R Alhambra-Expósito
- Maimonides Institute of Biomedical Research of Cordoba, Cordoba, Spain
- Reina Sofia University Hospital, Cordoba, Spain
- Service of Endocrinology and Nutrition, IMIBIC, Reina Sofia University Hospital, Cordoba, Spain
| | - Ana Barrera-Martín
- Maimonides Institute of Biomedical Research of Cordoba, Cordoba, Spain
- Reina Sofia University Hospital, Cordoba, Spain
- Service of Endocrinology and Nutrition, IMIBIC, Reina Sofia University Hospital, Cordoba, Spain
| | - Cristóbal Blanco-Acevedo
- Maimonides Institute of Biomedical Research of Cordoba, Cordoba, Spain
- Reina Sofia University Hospital, Cordoba, Spain
- Service of Neurosurgery, Reina Sofia University Hospital, Cordoba, Spain
| | - Elena Dios
- Metabolism and Nutrition Unit, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla, Sevilla, Spain
| | - Eva Venegas-Moreno
- Metabolism and Nutrition Unit, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla, Sevilla, Spain
| | - Juan Solivera
- Maimonides Institute of Biomedical Research of Cordoba, Cordoba, Spain
- Reina Sofia University Hospital, Cordoba, Spain
- Service of Neurosurgery, Reina Sofia University Hospital, Cordoba, Spain
| | - Manuel D Gahete
- Maimonides Institute of Biomedical Research of Cordoba, Cordoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain
- Reina Sofia University Hospital, Cordoba, Spain
- CIBER Physiopathology of Obesity and Nutrition, Cordoba, Spain
| | - Alfonso Soto-Moreno
- Metabolism and Nutrition Unit, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla, Sevilla, Spain
| | - María A Gálvez-Moreno
- Maimonides Institute of Biomedical Research of Cordoba, Cordoba, Spain
- Reina Sofia University Hospital, Cordoba, Spain
- Service of Endocrinology and Nutrition, IMIBIC, Reina Sofia University Hospital, Cordoba, Spain
| | - Justo P Castaño
- Maimonides Institute of Biomedical Research of Cordoba, Cordoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain
- Reina Sofia University Hospital, Cordoba, Spain
- CIBER Physiopathology of Obesity and Nutrition, Cordoba, Spain
| | - Raúl M Luque
- Maimonides Institute of Biomedical Research of Cordoba, Cordoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain
- Reina Sofia University Hospital, Cordoba, Spain
- CIBER Physiopathology of Obesity and Nutrition, Cordoba, Spain
| |
Collapse
|
35
|
Thakur S, Daley B, Klubo-Gwiezdzinska J. The role of an anti-diabetic drug metformin in the treatment of endocrine tumors. J Mol Endocrinol 2019; 63:R17-R35. [PMID: 31307011 PMCID: PMC6938582 DOI: 10.1530/jme-19-0083] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 07/15/2019] [Indexed: 12/15/2022]
Abstract
Incidence of endocrine cancers is rising every year. Over the last decade, evidence has accumulated that demonstrates the anti-cancer effects of an anti-diabetic drug, metformin, in endocrine malignancies. We performed a literature review utilizing the PubMed, Medline and clinicaltrials.gov databases using the keyword 'metformin' plus the following terms: 'thyroid cancer', 'thyroid nodules', 'parathyroid', 'hyperparathyroidism', 'adrenal adenoma', 'Cushing syndrome', 'hyperaldosteronism', 'adrenocortical cancer', 'neuroendocrine tumor (NET)', 'pancreatic NET (pNET)', 'carcinoid', 'pituitary adenoma', 'pituitary neuroendocrine tumor (PitNET)', 'prolactinoma', 'pheochromocytoma/paraganglioma'. We found 37 studies describing the preclinical and clinical role of metformin in endocrine tumors. The available epidemiological data show an association between exposure of metformin and lower incidence of thyroid cancer and pNETs in diabetic patients. Metformin treatment has been associated with better response to cancer therapy in thyroid cancer and pNETs. Preclinical evidence suggests that the primary direct mechanisms of metformin action include inhibition of mitochondrial oxidative phosphorylation via inhibition of both mitochondrial complex I and mitochondrial glycerophosphate dehydrogenase, leading to metabolic stress. Decreased ATP production leads to an activation of a cellular energy sensor, AMPK, and subsequent downregulation of mTOR signaling pathway, which is associated with decreased cellular proliferation. We also describe several AMPK-independent mechanisms of metformin action, as well as the indirect mechanisms targeting insulin resistance. Overall, repositioning of metformin has emerged as a promising strategy for adjuvant therapy of endocrine tumors. The mechanisms of synergy between metformin and other anti-cancer agents need to be elucidated further to guide well-designed prospective trials on combination therapies in endocrine malignancies.
Collapse
Affiliation(s)
- Shilpa Thakur
- Metabolic Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Brianna Daley
- Metabolic Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Joanna Klubo-Gwiezdzinska
- Metabolic Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
36
|
Fuentes-Fayos AC, García-Martínez A, Herrera-Martínez AD, Jiménez-Vacas JM, Vázquez-Borrego MC, Castaño JP, Picó A, Gahete MD, Luque RM. Molecular determinants of the response to medical treatment of growth hormone secreting pituitary neuroendocrine tumors. MINERVA ENDOCRINOL 2019; 44:109-128. [PMID: 30650942 DOI: 10.23736/s0391-1977.19.02970-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Acromegaly is a chronic systemic disease mainly caused by a growth hormone (GH)-secreting pituitary neuroendocrine tumor (PitNETs), which is associated with many health complications and increased mortality when not adequately treated. Transsphenoidal surgery is considered the treatment of choice in GH-secreting PitNETs, but patients in whom surgery cannot be considered or with persistent disease after surgery require medical therapy. Treatment with available synthetic somatostatin analogues (SSAs) is considered the mainstay in the medical management of acromegaly which exert their beneficial effects through the binding to a family of G-protein coupled receptors encoded by 5 genes (SSTR1-5). However, although it has been demonstrated that the SST1-5 receptors are physically present in tumor cells, SSAs are in many cases ineffective (i.e. approximately 10-30% of patients with GH-secreting PitNET are unresponsive to SSAs), suggesting that other cellular/molecular determinants could be essential for the response to the pharmacological treatment in patients with GH-secreting PitNETs. Therefore, the scrutiny of these determinants might be used for the identification of subgroups of patients in whom an appropriate pharmacological treatment can be successfully employed (responders vs. non-responders). In this review, we will describe some of the existing, classical and novel, genetic and molecular determinants involved in the response of patients with GH-secreting PitNETs to the available therapeutic treatments, as well as new molecular/therapeutic approaches that could be potentially useful for the treatment of GH-secreting PitNETs.
Collapse
Affiliation(s)
- Antonio C Fuentes-Fayos
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Cordoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain.,Reina Sofia University Hospital (HURS), Cordoba, Spain.,CIBER Physiopathology of Obesity and Nutrition (CIBERobn), Cordoba, Spain
| | - Araceli García-Martínez
- Research Laboratory, Hospital General Universitario de Alicante-Institute for Health and Biomedical Research (ISABIAL), Alicante, Spain
| | - Aura D Herrera-Martínez
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Cordoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain.,Reina Sofia University Hospital (HURS), Cordoba, Spain.,CIBER Physiopathology of Obesity and Nutrition (CIBERobn), Cordoba, Spain
| | - Juan M Jiménez-Vacas
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Cordoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain.,Reina Sofia University Hospital (HURS), Cordoba, Spain.,CIBER Physiopathology of Obesity and Nutrition (CIBERobn), Cordoba, Spain
| | - Mari C Vázquez-Borrego
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Cordoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain.,Reina Sofia University Hospital (HURS), Cordoba, Spain.,CIBER Physiopathology of Obesity and Nutrition (CIBERobn), Cordoba, Spain
| | - Justo P Castaño
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Cordoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain.,Reina Sofia University Hospital (HURS), Cordoba, Spain.,CIBER Physiopathology of Obesity and Nutrition (CIBERobn), Cordoba, Spain
| | - Antonio Picó
- Department of Endocrinology and Nutrition, Hospital General Universitario de Alicante-ISABIAL, Miguel Hernández University, CIBERER, Alicante, Spain
| | - Manuel D Gahete
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Cordoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain.,Reina Sofia University Hospital (HURS), Cordoba, Spain.,CIBER Physiopathology of Obesity and Nutrition (CIBERobn), Cordoba, Spain
| | - Raúl M Luque
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Cordoba, Spain - .,Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain.,Reina Sofia University Hospital (HURS), Cordoba, Spain.,CIBER Physiopathology of Obesity and Nutrition (CIBERobn), Cordoba, Spain
| |
Collapse
|