1
|
The Role of Somatostatin Analogues in the Control of Diarrhea and Flushing as Markers of Carcinoid Syndrome: A Systematic Review and Meta-Analysis. J Pers Med 2023; 13:jpm13020304. [PMID: 36836538 PMCID: PMC9959441 DOI: 10.3390/jpm13020304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 01/27/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
BACKGROUND Somatostatin analogues (SSAs) are the cornerstone of treatment for carcinoid syndrome (CS)-related symptoms. The aim of this systematic review and meta-analysis is to evaluate the percentage of patients achieving partial (PR) or complete response (CR) with the use of long-acting SSAs in patients with CS. METHODS A systematic electronic literature search was conducted in PubMed, Cochrane, and Scopus to identify eligible studies. Any clinical trials reporting data on the efficacy of SSAs to alleviate symptoms in adult patients were considered as potentially eligible. RESULTS A total of 17 studies reported extractable outcomes (PR/CR) for quantitative synthesis. The pooled percentage of patients with PR/CR for diarrhea was estimated to be 0.67 (95% confidence interval (CI): 0.52-0.79, I2 = 83%). Subgroup analyses of specific drugs provided no evidence of a differential response. With regards to flushing, the pooled percentage of patients with PR/CR was estimated to be 0.68 (95% CI: 0.52-0.81, I2 = 86%). Similarly, no evidence of a significant differential response in flushing control was documented. CONCLUSIONS We estimate there is a 67-68% overall reduction in symptoms of CS associated with SSA treatment. However, significant heterogeneity was detected, possibly revealing differences in the disease course, in management and in outcome definition.
Collapse
|
2
|
Mohamed A, Wu S, Hamid M, Mahipal A, Cjakrabarti S, Bajor D, Selfridge JE, Asa SL. Management of Appendix Neuroendocrine Neoplasms: Insights on the Current Guidelines. Cancers (Basel) 2022; 15:295. [PMID: 36612291 PMCID: PMC9818268 DOI: 10.3390/cancers15010295] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/19/2022] [Accepted: 12/26/2022] [Indexed: 01/03/2023] Open
Abstract
Appendiceal neuroendocrine neoplasms (ANENs) usually present as incidental findings at the time of appendectomy for acute appendicitis. They are rare, accounting for only 0.5-1% of intestinal neoplasms; they are found in 0.3-0.9% of all appendectomy specimens. They are usually sporadic tumors. There are several histological types including well-differentiated neuroendocrine tumors (NETs), poorly differentiated neuroendocrine carcinomas (NECs), and mixed neuroendocrine-non-neuroendocrine neoplasms (MiNENs). Histologic differentiation and the grade of well-differentiated NETs correlate with clinical behavior and prognosis. Management varies based on differentiation, aggressiveness, and metastatic potential. There is debate about the optimal surgical management for localized appendiceal NETs that are impacted by many factors including the tumor size, the extent of mesoappendiceal spread, lymphovascular invasion and perineural involvement. In addition, the data to guide therapy in metastatic disease are limited due to the paucity of these tumors. Here, we review the current advances in the management of ANENs within the context of a multidisciplinary approach to these tumors.
Collapse
Affiliation(s)
- Amr Mohamed
- Division of Hematology and Medical Oncology, UH Seidman Cancer Center, 11100 Euclid Avenue, Cleveland, OH 44106, USA
| | - Sulin Wu
- Department of Internal Medicine, UH Seidman Cancer Center, 11100 Euclid Avenue, Cleveland, OH 44106, USA
- Department of Medical Genetics, Center for Human Genetics, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Mohamed Hamid
- Department of Stem Cell Biology and Regenerative Medicine, City of Hope Beckman Research Institute, Duarte, CA 91010, USA
| | - Amit Mahipal
- Division of Hematology and Medical Oncology, UH Seidman Cancer Center, 11100 Euclid Avenue, Cleveland, OH 44106, USA
| | - Sakti Cjakrabarti
- Division of Hematology and Medical Oncology, UH Seidman Cancer Center, 11100 Euclid Avenue, Cleveland, OH 44106, USA
| | - David Bajor
- Division of Hematology and Medical Oncology, UH Seidman Cancer Center, 11100 Euclid Avenue, Cleveland, OH 44106, USA
| | - J. Eva Selfridge
- Division of Hematology and Medical Oncology, UH Seidman Cancer Center, 11100 Euclid Avenue, Cleveland, OH 44106, USA
| | - Sylvia L. Asa
- Department of Pathology, UH Seidman Cancer Center, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
| |
Collapse
|
3
|
McClellan K, Chen EY, Kardosh A, Lopez CD, Del Rivero J, Mallak N, Rocha FG, Koethe Y, Pommier R, Mittra E, Pegna GJ. Therapy Resistant Gastroenteropancreatic Neuroendocrine Tumors. Cancers (Basel) 2022; 14:4769. [PMID: 36230691 PMCID: PMC9563314 DOI: 10.3390/cancers14194769] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/24/2022] [Accepted: 09/28/2022] [Indexed: 11/16/2022] Open
Abstract
Gastroenteropancreatic neuroendocrine tumors (GEP-NETs) are a heterogenous group of malignancies originating from neuroendocrine cells of the gastrointestinal tract, the incidence of which has been increasing for several decades. While there has been significant progress in the development of therapeutic options for patients with advanced or metastatic disease, these remain limited both in quantity and durability of benefit. This review examines the latest research elucidating the mechanisms of both up-front resistance and the eventual development of resistance to the primary systemic therapeutic options including somatostatin analogues, peptide receptor radionuclide therapy with lutetium Lu 177 dotatate, everolimus, sunitinib, and temozolomide-based chemotherapy. Further, potential strategies for overcoming these mechanisms of resistance are reviewed in addition to a comprehensive review of ongoing and planned clinical trials addressing this important challenge.
Collapse
Affiliation(s)
- Kristen McClellan
- School of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Emerson Y. Chen
- Division of Hematology Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Adel Kardosh
- Division of Hematology Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Charles D. Lopez
- Division of Hematology Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Jaydira Del Rivero
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nadine Mallak
- Division of Molecular Imaging and Therapy, Oregon Health & Science University, Portland, OR 97239, USA
| | - Flavio G. Rocha
- Division of Surgical Oncology, Department of Surgery, Oregon Health & Science University, Portland, OR 97239, USA
| | - Yilun Koethe
- Dotter Department of Interventional Radiology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Rodney Pommier
- Division of Surgical Oncology, Department of Surgery, Oregon Health & Science University, Portland, OR 97239, USA
| | - Erik Mittra
- Division of Molecular Imaging and Therapy, Oregon Health & Science University, Portland, OR 97239, USA
| | - Guillaume J. Pegna
- Division of Hematology Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| |
Collapse
|
4
|
Koumarianou A, Daskalakis K, Tsoli M, Kaltsas G, Pavel M. Efficacy, safety and unmet needs of evolving medical treatments for carcinoid syndrome. J Neuroendocrinol 2022; 34:e13174. [PMID: 35794780 DOI: 10.1111/jne.13174] [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: 02/18/2022] [Revised: 04/06/2022] [Accepted: 05/31/2022] [Indexed: 11/25/2022]
Abstract
This review reports on the currently available medical treatment options for the control of symptoms due to carcinoid syndrome in patients with neuroendocrine tumors. The efficacy and adverse events (AEs) of approved drugs such as somatostatin analogues (SSA), telotristat ethyl (TE) and interferon-alpha, are reviewed. Somatostatin analogues remain the standard treatment of carcinoid syndrome based on the high expression of somatostatin receptors and the resulting inhibition of secretion of bioactive compounds; their use is associated with relatively mild AEs, involving mainly the gastrointestinal system, and being usually transient. Although dose escalation of SSA remains an unapproved option, it is clinically implemented to alleviate symptoms in refractory carcinoid syndrome and supported by the most recent guidelines. The side effects associated with the increased dose are in general mild and consistent with standard dose of SSA. Telotristat ethyl, an oral inhibitor of tryptophan hydroxylase, the rate-limiting enzyme in serotonin biosynthesis, represents a rather novel innovative treatment option in patients with carcinoid syndrome suffering from diarrhea and complements the standard therapy of SSA. Given the low toxicity profile, TE may be considered an early add-on treatment to SSA in patients with uncontrolled carcinoid syndrome. However, further prolonged follow-up of patients treated with TE may be needed to exclude potential AEs, such as liver toxicity or depressed mood, in patients with long-term treatment. Interferon alpha is a cytokine with direct inhibitory effect on hormone secretion and tumor cell proliferation and an approved therapy in carcinoid syndrome but is associated with significant AEs in the majority of the patients requiring frequently dose reduction. The finding of a more favorable tolerability of pegylated interferon needs to be confirmed in a prospective study.
Collapse
Affiliation(s)
- Anna Koumarianou
- Hematology-Oncology Unit, Fourth Department of Internal Medicine, Attikon Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Kosmas Daskalakis
- Department of Surgery, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- 2nd Department of Surgery, "Korgialenio-Benakio", Red Cross General Hospital, Athens, Greece
| | - Marina Tsoli
- 1st Propaedeutic Department of Internal Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Gregory Kaltsas
- 1st Propaedeutic Department of Internal Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Marianne Pavel
- Department of Endocrinology, Universitatsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
5
|
Syguła A, Ledwon A, Hasse-Lazar K, Jurecka-Lubieniecka B, Michalik B, Paliczka-Cieślik E, Zeman M, Chmielik E, Sczasny J, Jarzab B, Handkiewicz-Junak D. In patients with well-differentiated neuroendocrine tumours, there is no apparent benefit of somatostatin analogues after disease control by peptide receptor radionuclide therapy. Eur J Nucl Med Mol Imaging 2022; 49:3841-3851. [PMID: 35503379 DOI: 10.1007/s00259-022-05792-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 04/03/2022] [Indexed: 12/13/2022]
Abstract
PURPOSE Peptide receptor radionuclide therapy (PRRT) and somatostatin analogues (SSAs) are commonly combined as primary treatment for neuroendocrine neoplasms (NEN), and SSAs given as maintenance. We sought to evaluate whether sequential therapy with PRRT followed by SSAs has progression or survival benefits in patients with NEN after disease control by PRRT. METHODS This prospective, randomised, single-centre study had as principal eligibility criteria: unresectable, locally advanced, or metastatic, histologically confirmed well-differentiated NEN; no symptoms/biochemical diagnosis of carcinoid syndrome; no SSAs or ≤ 3 months of SSAs before PRRT; and stable disease or partial or complete response after PRRT. Altogether, 115 patients were randomised 2:1 to an SSA group (n = 74) given octreotide acetate LAR every 4 weeks, or a control group (n = 41) receiving only best supportive care. Octreotide treatment was to stop upon intolerable toxicity or patient refusal, or, at physician/patient discretion, upon NEN progression. The primary endpoint was progression-free survival (PFS), the secondary endpoint, and overall survival (OS). RESULTS Median (25th-75th percentile) follow-up from the first PRRT activity to death or latest observation was 6.6 (3.18-10.22) years. During that time, 71/115 patients (62%) progressed, 52/74 (70%) in the SSA group, and 19/41 (46%) in the control group (p = 0.01). Eighty-eight/115 patients (76%) died, 58/74 (78%) in the SSA group, and 30/41 (73%) in the control group (p = 0.52). Median (95% CI) PFS was 4.7 (2.8-7.7) years in the SSA group, and 6.4 (4.1-not reached) years in controls. Overall, median OS was 6.6 years. Neither PFS nor OS differed between groups (p = 0.129, p = 0.985, respectively). CONCLUSIONS In patients with disease control after PRRT, subsequent SSA treatment appeared not to be associated with better PFS or OS. Whether to continue SSA administration upon progression after PRRT requires evaluation in a prospective, randomised, controlled multicentre study with a relatively homogeneous sample.
Collapse
Affiliation(s)
- Aleksandra Syguła
- Department of Nuclear Medicine and Endocrine Oncology, Gliwice Branch, Maria Sklodowska-Curie National Research Institute of Oncology, Wybrzeże Armii Krajowej 16, 44-101, Gliwice, Poland
| | - Aleksandra Ledwon
- Department of Nuclear Medicine and Endocrine Oncology, Gliwice Branch, Maria Sklodowska-Curie National Research Institute of Oncology, Wybrzeże Armii Krajowej 16, 44-101, Gliwice, Poland
| | - Kornelia Hasse-Lazar
- Department of Nuclear Medicine and Endocrine Oncology, Gliwice Branch, Maria Sklodowska-Curie National Research Institute of Oncology, Wybrzeże Armii Krajowej 16, 44-101, Gliwice, Poland
| | - Beata Jurecka-Lubieniecka
- Department of Nuclear Medicine and Endocrine Oncology, Gliwice Branch, Maria Sklodowska-Curie National Research Institute of Oncology, Wybrzeże Armii Krajowej 16, 44-101, Gliwice, Poland
| | - Barbara Michalik
- Department of Nuclear Medicine and Endocrine Oncology, Gliwice Branch, Maria Sklodowska-Curie National Research Institute of Oncology, Wybrzeże Armii Krajowej 16, 44-101, Gliwice, Poland
| | - Ewa Paliczka-Cieślik
- Department of Nuclear Medicine and Endocrine Oncology, Gliwice Branch, Maria Sklodowska-Curie National Research Institute of Oncology, Wybrzeże Armii Krajowej 16, 44-101, Gliwice, Poland
| | - Marcin Zeman
- The Oncologic and Reconstructive Surgery Clinic, Gliwice Branch, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, Poland
| | - Ewa Chmielik
- Tumor Pathology Department, Gliwice Branch, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, Poland
| | - Joanna Sczasny
- Radiology and Diagnostic Imaging Department, Gliwice Branch, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, Poland
| | - Barbara Jarzab
- Department of Nuclear Medicine and Endocrine Oncology, Gliwice Branch, Maria Sklodowska-Curie National Research Institute of Oncology, Wybrzeże Armii Krajowej 16, 44-101, Gliwice, Poland
| | - Daria Handkiewicz-Junak
- Department of Nuclear Medicine and Endocrine Oncology, Gliwice Branch, Maria Sklodowska-Curie National Research Institute of Oncology, Wybrzeże Armii Krajowej 16, 44-101, Gliwice, Poland.
| |
Collapse
|
6
|
Allaw MB, Switchenko JM, Khalil L, Wu C, Alese OB, Akce M, Draper A, Jones AT, El-Rayes B, Shaib W. Comparing Somatostatin Analogs in the Treatment of Advanced Gastroenteropancreatic Neuroendocrine Tumors. Oncology 2022; 100:131-139. [PMID: 35078191 PMCID: PMC9052205 DOI: 10.1159/000519605] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 08/24/2021] [Indexed: 11/19/2022]
Abstract
BACKGROUND The 2 approved somatostatin analogs (SSAs) in the first-line treatment of advanced, well-differentiated gastroenteropancreatic neuroendocrine tumors (GEP-NETs) are octreotide long-acting release (Sandostatin LAR) and somatuline depot (Lanreotide). The study's objective was to compare progression-free survival (PFS) and overall survival (OS) of patients (pts) with GEP-NETs treated with somatuline or octreotide LAR. Pts and Methods: Pts with advanced well-differentiated GEP-NET who received either SSA at Emory University between 1995 and 2019 were included after institutional review board approval. The primary end point was PFS, defined as time to disease progression (according to the Response Evaluation Criteria in Solid Tumors, version 1.1, or clinical progression) or death. The secondary end point was OS. Kaplan-Meier curves were generated, and log-rank tests were conducted to compare the survival outcomes. RESULTS A total of 105 pts were identified. The mean age was 62.1 years (SD ± 11.8). The male-to-female ratio was 51:54. The majority (N = 69, 65.7%) were white. Most pts had grade 2 (G2) disease (N = 44, 41.9%). Primary location was small bowel in 58 (55.2%), pancreas in 27 (25.7%), and other in 20 (19.0%). Functional tumors were defined in 32 pts distributed equally between the 2 groups. Distribution of treatment was similar in the 2 groups, with 54 receiving octreotide LAR and 51 receiving somatuline depot. The median PFS for the octreotide LAR and somatuline depot groups was 12 months (95% CI, 6-18 months) and 10.8 months (95% CI, 6-15.6 months), respectively, and the difference was not statistically significant (p = 0.2665). For pts with G1 disease, the median PFS for the octreotide LAR and somatuline depot was 8.4 versus 32.4 months, respectively, and the difference was not statistically significant (p = 0.159). For G2 disease, the difference in median PFS between octreotide LAR and somutaline depot groups was statistically significant (12 vs. 7.2 months, respectively; p = 0.0372). The mean follow-up time for octreotide LAR was 21.6 months versus 11.3 months for somatuline depot. CONCLUSIONS Overall, there was no difference in PFS between octreotide LAR and somatuline depot for pts with well-differentiated, metastatic GEP-NETs. A prospective study is worth designing selecting for G.
Collapse
Affiliation(s)
- Mohammed B. Allaw
- Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Jeffrey M. Switchenko
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Lana Khalil
- Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Christina Wu
- Department of Hematology and Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Olatunji B. Alese
- Department of Hematology and Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Mehmet Akce
- Department of Hematology and Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Amber Draper
- Clinical Pharmacy, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Aaron T. Jones
- Department of Hematology and Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Bassel El-Rayes
- Department of Hematology and Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Walid Shaib
- Department of Hematology and Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| |
Collapse
|
7
|
Spada F, Rossi RE, Kara E, Laffi A, Massironi S, Rubino M, Grimaldi F, Bhoori S, Fazio N. Carcinoid Syndrome and Hyperinsulinemic Hypoglycemia Associated with Neuroendocrine Neoplasms: A Critical Review on Clinical and Pharmacological Management. Pharmaceuticals (Basel) 2021; 14:539. [PMID: 34199977 PMCID: PMC8228616 DOI: 10.3390/ph14060539] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 05/31/2021] [Accepted: 06/01/2021] [Indexed: 02/05/2023] Open
Abstract
The carcinoid syndrome (CS) and hyperinsulinemic hypoglycemia (HH) represent two of the most common clinical syndromes associated with neuroendocrine neoplasms (NENs). The former is mainly related to the serotonin secretion by a small bowel NEN, whereas the latter depends on an insulin hypersecretion by a pancreatic insulinoma. Both syndromes/conditions can affect prognosis and quality of life of patients with NENs. They are often diagnosed late when patients become strongly symptomatic. Therefore, their early detection and management are a critical step in the clinical management of NEN patients. A dedicated and experienced multidisciplinary team with appropriate therapeutic strategies is needed and should be encouraged to optimize clinical outcomes. This review aims to critically analyze clinical features, evidence and treatment options of CS and HH and therefore to improve their management.
Collapse
Affiliation(s)
- Francesca Spada
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology (IEO) IRCCS, via G. Ripamonti 435, 20141 Milano, Italy; (F.S.); (A.L.); (M.R.)
| | - Roberta E. Rossi
- Hepatology and Hepato-Pancreatic-Biliary Surgery and Liver Transplantation, Fondazione IRCCS, Istituto Nazionale Tumori (INT), via G. Venezian 1, 20133 Milano, Italy; (R.E.R.); (S.B.)
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, via Festa del Perdono 7, 20122 Milano, Italy
| | - Elda Kara
- Endocrinology and Metabolism Unit, University Hospital S. Maria della Misericordia, Piazzale Santa Maria della Misericordia, 15, 33100 Udine, Italy; (E.K.); (F.G.)
| | - Alice Laffi
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology (IEO) IRCCS, via G. Ripamonti 435, 20141 Milano, Italy; (F.S.); (A.L.); (M.R.)
| | - Sara Massironi
- Division of Gastroenterology, San Gerardo Hospital, Bicocca School of Medicine, University of Milano Bicocca, 20126 Milano, Italy;
| | - Manila Rubino
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology (IEO) IRCCS, via G. Ripamonti 435, 20141 Milano, Italy; (F.S.); (A.L.); (M.R.)
| | - Franco Grimaldi
- Endocrinology and Metabolism Unit, University Hospital S. Maria della Misericordia, Piazzale Santa Maria della Misericordia, 15, 33100 Udine, Italy; (E.K.); (F.G.)
| | - Sherrie Bhoori
- Hepatology and Hepato-Pancreatic-Biliary Surgery and Liver Transplantation, Fondazione IRCCS, Istituto Nazionale Tumori (INT), via G. Venezian 1, 20133 Milano, Italy; (R.E.R.); (S.B.)
| | - Nicola Fazio
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology (IEO) IRCCS, via G. Ripamonti 435, 20141 Milano, Italy; (F.S.); (A.L.); (M.R.)
| |
Collapse
|
8
|
Patel M, Tena I, Jha A, Taieb D, Pacak K. Somatostatin Receptors and Analogs in Pheochromocytoma and Paraganglioma: Old Players in a New Precision Medicine World. Front Endocrinol (Lausanne) 2021; 12:625312. [PMID: 33854479 PMCID: PMC8039528 DOI: 10.3389/fendo.2021.625312] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 03/04/2021] [Indexed: 12/16/2022] Open
Abstract
Neuroendocrine tumors overexpress somatostatin receptors, which serve as important and unique therapeutic targets for well-differentiated advanced disease. This overexpression is a well-established finding in gastroenteropancreatic neuroendocrine tumors which has guided new medical therapies in the administration of somatostatin analogs, both "cold", particularly octreotide and lanreotide, and "hot" analogs, chelated to radiolabeled isotopes. The binding of these analogs to somatostatin receptors effectively suppresses excess hormone secretion and tumor cell proliferation, leading to stabilization, and in some cases, tumor shrinkage. Radioisotope-labeled somatostatin analogs are utilized for both tumor localization and peptide radionuclide therapy, with 68Ga-DOTATATE and 177Lu-DOTATATE respectively. Benign and malignant pheochromocytomas and paragangliomas also overexpress somatostatin receptors, irrespective of embryological origin. The pattern of somatostatin receptor overexpression is more prominent in succinate dehydrogenase subunit B gene mutation, which is more aggressive than other subgroups of this disease. While the Food and Drug Administration has approved the use of 68Ga-DOTATATE as a radiopharmaceutical for somatostatin receptor imaging, the use of its radiotherapeutic counterpart still needs approval beyond gastroenteropancreatic neuroendocrine tumors. Thus, patients with pheochromocytoma and paraganglioma, especially those with inoperable or metastatic diseases, depend on the clinical trials of somatostatin analogs. The review summarizes the advances in the utilization of somatostatin receptor for diagnostic and therapeutic approaches in the neuroendocrine tumor subset of pheochromocytoma and paraganglioma; we hope to provide a positive perspective in using these receptors as targets for treatment in this rare condition.
Collapse
Affiliation(s)
- Mayank Patel
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Isabel Tena
- Scientific Department, Medica Scientia Innovation Research (MedSIR), Barcelona, Spain
- Section of Medical Oncology, Consorcio Hospitalario Provincial of Castellon, Castellon, Spain
| | - Abhishek Jha
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - David Taieb
- Department of Nuclear Medicine, La Timone University Hospital, CERIMED, Aix-Marseille University, Marseille, France
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
- *Correspondence: Karel Pacak,
| |
Collapse
|
9
|
Bober B, Saracyn M, Kołodziej M, Kowalski Ł, Deptuła-Krawczyk E, Kapusta W, Kamiński G, Mozenska O, Bil J. Carcinoid Heart Disease: How to Diagnose and Treat in 2020? CLINICAL MEDICINE INSIGHTS-CARDIOLOGY 2020; 14:1179546820968101. [PMID: 33192110 PMCID: PMC7597558 DOI: 10.1177/1179546820968101] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 09/25/2020] [Indexed: 01/17/2023]
Abstract
Neuroendocrine tumors (NETs, originally termed “carcinoids”) create a relatively rare group of neoplasms with an approximate incidence rate of 2.5 to 5 cases per 100 000 persons. Roughly 30% to 40% of subjects with NETs develop carcinoid syndrome (CS), and 20% to 50% of subjects with CS are diagnosed with carcinoid heart disease (CaHD). The long-standing exposure to high serum serotonin concentration is one of the crucial factors in CaHD development. White plaque-like deposits on the endocardial surface of heart structures with valve leaflets and subvalvular apparatus thickening (fused and shortened chordae; thickened papillary muscles) are characteristic for CaHD. NT pro-BNP and 5-hydroxyindoleacetic acid are the 2 most useful screening markers. Long-acting somatostatin analogs are the standard of care in symptoms control. They are also the first-line treatment for tumor control in subjects with a metastatic somatostatin receptor avid disease. In cases refractory to somatostatin analogs, several options are available. We can increase a somatostatin analog to off-label doses, add telotristat ethyl or administer peptide receptor radionuclide therapy. Cardiac surgery, which mainly involves valve replacement, is presently the most efficient strategy in subjects with advanced CaHD and can relieve unmanageable symptoms or be partly responsible for better prognosis.
Collapse
Affiliation(s)
- Barbara Bober
- Department of Endocrinology and Isotope Therapy, Military Institute of Medicine, Warsaw, Mazowieckie, Poland
| | - Marek Saracyn
- Department of Endocrinology and Isotope Therapy, Military Institute of Medicine, Warsaw, Mazowieckie, Poland
| | - Maciej Kołodziej
- Department of Endocrinology and Isotope Therapy, Military Institute of Medicine, Warsaw, Mazowieckie, Poland
| | - Łukasz Kowalski
- Department of Endocrinology and Isotope Therapy, Military Institute of Medicine, Warsaw, Mazowieckie, Poland
| | - Elżbieta Deptuła-Krawczyk
- Department of Endocrinology and Isotope Therapy, Military Institute of Medicine, Warsaw, Mazowieckie, Poland
| | - Waldemar Kapusta
- Department of Endocrinology and Isotope Therapy, Military Institute of Medicine, Warsaw, Mazowieckie, Poland
| | - Grzegorz Kamiński
- Department of Endocrinology and Isotope Therapy, Military Institute of Medicine, Warsaw, Mazowieckie, Poland
| | - Olga Mozenska
- Department of Internal Medicine, Hypertension and Vascular Diseases, Warsaw Medical University, Warsaw, Poland
| | - Jacek Bil
- Department of Invasive Cardiology, Centre of Postgraduate Medical Education, Warsaw, Poland
| |
Collapse
|
10
|
Ahmed M. Gastrointestinal neuroendocrine tumors in 2020. World J Gastrointest Oncol 2020; 12:791-807. [PMID: 32879660 PMCID: PMC7443843 DOI: 10.4251/wjgo.v12.i8.791] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 05/26/2020] [Accepted: 07/18/2020] [Indexed: 02/05/2023] Open
Abstract
Gastrointestinal neuroendocrine tumors are rare slow-growing tumors with distinct histological, biological, and clinical characteristics that have increased in incidence and prevalence within the last few decades. They contain chromogranin A, synaptophysin and neuron-specific enolase which are necessary for making a diagnosis of neuroendocrine tumor. Ki-67 index and mitotic index correlate with cellular proliferation. Serum chromogranin A is the most commonly used biomarker to assess the bulk of disease and monitor treatment and is raised in both functioning and non-functioning neuroendocrine tumors. Most of the gastrointestinal neuroendocrine tumors are non-functional. World Health Organization updated the classification of neuroendocrine tumors in 2017 and renamed mixed adenoneuroendocrine carcinoma into mixed neuroendocrine neoplasm. Gastric neuroendocrine tumors arise from enterochromaffin like cells. They are classified into 4 types. Only type I and type II are gastrin dependent. Small intestinal neuroendocrine tumor is the most common small bowel malignancy. More than two-third of them occur in the terminal ileum within 60 cm of ileocecal valve. Patients with small intestinal neuroendrocrine tumors frequently show clinical symptoms and develop distant metastases more often than those with neuroendocrine tumors of other organs. Duodenal and jejuno-ileal neuroendocrine tumors are distinct biologically and clinically. Carcinoid syndrome generally occurs when jejuno-ileal neuroendocrine tumors metastasize to the liver. Appendiceal neuroendocrine tumors are generally detected after appendectomy. Colonic neuroendocrine tumors generally present as a large tumor with local or distant metastasis at the time of diagnosis. Rectal neuroendocrine tumors are increasingly being diagnosed since the implementation of screening colonoscopy in 2000. Gastrointestinal neuroendocrine tumors are diagnosed and staged by endoscopy with biopsy, endoscopic ultrasound, serology of biomarkers, imaging studies and functional somatostatin scans. Various treatment options are available for curative and palliative treatment of gastrointestinal neuroendocrine tumors.
Collapse
Affiliation(s)
- Monjur Ahmed
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Thomas Jefferson University, Philadelphia, PA 19107, United States
| |
Collapse
|
11
|
Larouche V, Akirov A, Alshehri S, Ezzat S. Management of Small Bowel Neuroendocrine Tumors. Cancers (Basel) 2019; 11:cancers11091395. [PMID: 31540509 PMCID: PMC6770692 DOI: 10.3390/cancers11091395] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 09/08/2019] [Accepted: 09/15/2019] [Indexed: 12/20/2022] Open
Abstract
Several important landmark trials have reshaped the landscape of non-surgical management of small bowel neuroendocrine tumors over the last few years, with the confirmation of the antitumor effect of somatostatin analogue therapy in PROMID and CLARINET trials as well as the advent of therapies with significant potential such as mammalian target of rapamycin inhibitor (mTor) everolimus (RADIANT trials) and peptide receptor radionuclide therapy (PRRT) with 177-Lutetium (NETTER-1 trial). This narrative summarizes the recommended management strategies of small bowel neuroendocrine tumors. We review the main evidence behind each recommendation as well as compare and contrast four major guidelines, namely the 2016 Canadian Consensus guidelines, the 2017 North American Neuroendocrine Tumor Society guidelines, the 2018 National Comprehensive Cancer Network guidelines, and the 2016 European Neuroendocrine Tumor Society guidelines. Different clinical situations will be addressed, from loco-regional therapy to metastatic unresectable disease. Carcinoid syndrome, which is mostly managed by somatostatin analogue therapy and the serotonin antagonist telotristat etiprate for refractory diarrhea, as well as neuroendocrine carcinoma will be reviewed. However, several questions remain unanswered, such as the optimal management of neuroendocrine carcinomas or the effect of combining and sequencing of the aforementioned modalities where more randomized controlled trials are needed.
Collapse
Affiliation(s)
- Vincent Larouche
- Endocrine Oncology Site Group, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON M5G2C1, Canada; (A.A.); (S.A.); (S.E.)
- Division of Endocrinology and Metabolism, Department of Medicine, Jewish General Hospital, McGill University, Montreal, QC H3T1E2, Canada
- Correspondence: ; Tel.: +1-(514)-340-8222 (ext. 28521)
| | - Amit Akirov
- Endocrine Oncology Site Group, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON M5G2C1, Canada; (A.A.); (S.A.); (S.E.)
- Department of Medicine, Institute of Endocrinology, Beilinson Hospital, Petach Tikva 4941492, Israel
- Department of Medicine, Sackler School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Sameerah Alshehri
- Endocrine Oncology Site Group, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON M5G2C1, Canada; (A.A.); (S.A.); (S.E.)
| | - Shereen Ezzat
- Endocrine Oncology Site Group, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON M5G2C1, Canada; (A.A.); (S.A.); (S.E.)
| |
Collapse
|
12
|
Torniai M, Scortichini L, Tronconi F, Rubini C, Morgese F, Rinaldi S, Mazzanti P, Berardi R. Systemic treatment for lung carcinoids: from bench to bedside. Clin Transl Med 2019; 8:22. [PMID: 31273555 PMCID: PMC6609661 DOI: 10.1186/s40169-019-0238-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Accepted: 06/21/2019] [Indexed: 12/13/2022] Open
Abstract
In the huge spectrum of lung neuroendocrine neoplasms, typical and atypical carcinoids should be considered as a separate biological entity from poorly differentiated forms, harboring peculiar molecular alterations. Despite their indolent behavior, lung carcinoids correlate with a worse survival. To date, only limited therapeutic options are available and novel drugs are strongly needed. In this work, we extensively reviewed scientific literature exploring available therapeutic options, new molecular targets and future perspectives in the management of well differentiated neoplasms of bronchopulmonary tree. Systemic therapy represents the main option in advanced and unresectable disease; accepted choices are somatostatin analogs, peptide receptor radionuclide therapy, everolimus and chemotherapy. To date, an univocal treatment strategy has not been identified yet, thus tailored therapeutic algorithms should consider treatment efficacy as well as safety profiles. Several molecular alterations found in carcinoid tumors might act as molecular targets leading to development of new therapeutic options. Further studies are necessary to identify new potential "druggable" molecular targets in the selected subset of low-grade lung carcinoids. Furthermore, evaluating the available therapies in more homogeneous population might improve their efficacy through a perfect tailoring of treatment options.
Collapse
Affiliation(s)
- Mariangela Torniai
- Clinica Oncologica, Università Politecnica delle Marche, AOU Ospedali Riuniti di Ancona, Via Conca 71, 60126 Ancona, Italy
| | - Laura Scortichini
- Clinica Oncologica, Università Politecnica delle Marche, AOU Ospedali Riuniti di Ancona, Via Conca 71, 60126 Ancona, Italy
| | - Francesca Tronconi
- Clinica Oncologica, Università Politecnica delle Marche, AOU Ospedali Riuniti di Ancona, Via Conca 71, 60126 Ancona, Italy
| | - Corrado Rubini
- Section of Pathological Anatomy and Histopathology, Department of Neuroscience, Università Politecnica delle Marche, AOU Ospedali Riuniti di Ancona, Ancona, Italy
| | - Francesca Morgese
- Clinica Oncologica, Università Politecnica delle Marche, AOU Ospedali Riuniti di Ancona, Via Conca 71, 60126 Ancona, Italy
| | - Silvia Rinaldi
- Clinica Oncologica, Università Politecnica delle Marche, AOU Ospedali Riuniti di Ancona, Via Conca 71, 60126 Ancona, Italy
| | - Paola Mazzanti
- Clinica Oncologica, Università Politecnica delle Marche, AOU Ospedali Riuniti di Ancona, Via Conca 71, 60126 Ancona, Italy
| | - Rossana Berardi
- Clinica Oncologica, Università Politecnica delle Marche, AOU Ospedali Riuniti di Ancona, Via Conca 71, 60126 Ancona, Italy
| |
Collapse
|
13
|
Wolin E, Benson III A. Systemic Treatment Options for Carcinoid Syndrome: A Systematic Review. Oncology 2019; 96:273-289. [DOI: 10.1159/000499049] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 02/07/2019] [Indexed: 11/19/2022]
|
14
|
Fagan R, Bokhari SSN, Inayat F. Vitamin D and vitamin B 12 deficiencies in patients with small intestinal carcinoid tumour: is opioid use disorder a confounding factor in the diagnosis? BMJ Case Rep 2019; 12:12/3/e227430. [PMID: 30878964 DOI: 10.1136/bcr-2018-227430] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Carcinoid tumours have the ability to secrete various peptides and bioamines that lead to carcinoid syndrome manifested as cutaneous flushing, diarrhoea, bronchial constriction and cardiac involvement. The deficiencies of vitamins D and B12 have previously been reported in patients with carcinoid tumours presumably due to chronic diarrhoea associated with the carcinoid syndrome. Herein, we chronicle the case of a patient with opioid use disorder who presented with small bowel obstruction that was found to be caused by a midgut carcinoid tumour. Laboratory studies revealed deficiencies of vitamins D and B12 even though he denied diarrhoea and had no other aetiology of deficiencies of these vitamins. Additionally, this paper presents a review of the published medical literature pertaining to clinical features, diagnostic investigations and treatment of intestinal carcinoid tumours and explores possible explanations for the observed deficiencies in these patients.
Collapse
Affiliation(s)
- Richard Fagan
- West Suburban Medical Center, Oak Park, Illinois, USA
| | | | | |
Collapse
|
15
|
Melosky B. Advanced typical and atypical carcinoid tumours of the lung: management recommendations. Curr Oncol 2018; 25:S86-S93. [PMID: 29910651 PMCID: PMC6001761 DOI: 10.3747/co.25.3808] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Background Neuroendocrine tumours (nets) are classified by site of origin, with lung being the second most common primary site after the gastrointestinal tract. Lung nets are rare and heterogeneous, with varied pathologic and clinical features. Typical and atypical carcinoid tumours are low-grade lung nets which, compared with the more common high-grade nets, are associated with a more favourable prognosis. Still, optimal treatment strategies are lacking. Methods This review concentrates on classification and treatment strategies for metastatic low-grade lung nets, considering both typical and atypical carcinoids. The terminology can be confusing, and an attempt is made to simplify it. Promising results from recent trials that included lung nets are presented and discussed. Finally, guidelines from Europe and North America are discussed, and differences are noted. Results Even within the group of patients with low-grade nets, the presentation, the locations of metastasis, and the speed of progression can be very different. The initial work-up and an understanding of the tumour's biology are key in making management decisions. Various treatment options-including somatostatin analogs, peptide receptor radioligand therapy, and biologic systemic therapy, specifically with the mtor (mechanistic target of rapamycin) inhibitor everolimus-are now available and are presented in a treatment algorithm. Summary Although lung nets are rare and evidence supporting optimal treatment strategies is lacking, the recent publication of trials that have included patients with lung nets advances evidence-based therapy for these tumours. Many variables have to be considered in managing these tumours that have received little attention. Education for treating physicians is needed.
Collapse
Affiliation(s)
- B. Melosky
- Medical Oncology, BC Cancer–Vancouver Centre, BC
| |
Collapse
|
16
|
Della Torre S, Procopio G, Fusi A, Catena L, Ferrari L, Nova P, Denaro A, Bichisao E, Bajetta E. Current Treatments of Neuroendocrine Tumors Role of Biotherapy and Chemotherapy. TUMORI JOURNAL 2018; 89:111-6. [PMID: 12841654 DOI: 10.1177/030089160308900201] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Neuroendocrine tumors are rare neoplasms originating from cells belonging to a diffuse or confined neuroendocrine system and characterized by a significant histopatologic and biologic heterogeneity. Timely diagnosis is delayed because they are often clinically silent for their low differentiation grade and the absence of any symptom due to abnormal hormone release. For these reasons, many neuroendocrine tumor patients are not treated medically for metastatic or inoperable disease. Medical treatments include biotherapy, with interferon-α and somatostatin analogues, and chemotherapy. Somastostatin analogues are widely used in patients with symptoms and with carcinoids of low differentiation grade. Interferon-α is used alone or in combination with somatostatin analogues. Chemotherapy is active in patients with poorly differentiated neuroendocrine tumors. The therapeutic regimen commonly used is the combination of cisplatinum and etoposide. In conclusion, no standard treatment for NET has yet been identified, and the response criteria suggested by ITMO remain a reference point. The clinical aspect of the disease and biologic features suggest the identification of neuroendocrine tumors patients suitable for the appropriate therapies. On these bases, it is recommended that diagnosis and treatment of neuroendocrine tumors be carried out at specialized oncological centers involved in clinical trials.
Collapse
|
17
|
Cuyle PJ, Prenen H. Practical management of toxicities associated with targeted therapies for advanced gastroenteropancreatic neuroendocrine tumors. Ann Gastroenterol 2018; 31:140-150. [PMID: 29507461 PMCID: PMC5825944 DOI: 10.20524/aog.2018.0224] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Accepted: 11/13/2017] [Indexed: 02/06/2023] Open
Abstract
Neuroendocrine tumors are heterogeneous, rare malignancies that arise most frequently in the gastroenteropancreatic tract (GEPNET). The therapeutic armamentarium for the treatment of GEPNETs has expanded significantly over the last two decades, however the ideal sequencing strategy remains controversial. As this disease may be relatively slow-growing, patients are expected to be treated for longer periods, so that even mild toxicities can influence quality of life, compliance and outcome in the long run. Prospective data on optimal adverse event management are lacking and recommendations are largely based on expert opinion and drug prescribing information. This review summarizes practical recommendations for toxicity management associated with the most commonly used GEPNET treatment options and stresses important focus points for future clinical trials.
Collapse
Affiliation(s)
- Pieter-Jan Cuyle
- Department of Gastroenterology/Digestive Oncology, Imelda General Hospital, Bonheiden (Pieter-Jan Cuyle), Belgium
| | - Hans Prenen
- Department of Gastroenterology/Digestive Oncology, University Hospitals Gasthuisberg Leuven, Leuven (Hans Prenen), Belgium
| |
Collapse
|
18
|
Carmona-Bayonas A, Jiménez-Fonseca P, Custodio A, Grande E, Capdevila J, López C, Teule A, Garcia-Carbonero R. Optimizing Somatostatin Analog Use in Well or Moderately Differentiated Gastroenteropancreatic Neuroendocrine Tumors. Curr Oncol Rep 2017; 19:72. [PMID: 28920153 DOI: 10.1007/s11912-017-0633-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Somatostatin analogues, aiming to control tumor secretion or growth, constitute the most attractive therapeutic option for patients with well-differentiated gastroenteropancreatic neuroendocrine tumors (GEP-NETs). The objective of this article is to provide a comprehensive review of the current state-of-the-art knowledge gaps and potential opportunities for future development and optimization of this therapeutic modality. METHOD A contextualized systematic review with a narrative component was conducted using PubMed, The Cochrane Library, EMBASE, and Google Scholar. Titles were screened, and non-English, duplicate, or irrelevant entries were excluded. Selection criteria for articles included the following: publication in English between 1995 and 2016, patients with GEP-NETs, analysis of efficacy, safety, practical management considerations, predictive factors, and/or strategies for overcoming resistance, concerning somatostatin analogs. RESULTS Ninety-seven studies out of 2771 screened publications met the inclusion criteria (16 randomized clinical trials, 27 phase II trials, 3 phase I trials, 3 subgroup analyses of clinical trials, 1 open-label extension of a randomized trial, 1 phase IV trial, 32 observational studies, and 14 basic research articles). The nature and scope of literature was diverse with most articles dedicated to drug efficacy or indications of use (n = 49), pharmacological issues (n = 8), assessment or predictors of response (n = 4), practical management (n = 11), combination therapy or other means to overcome resistance (n = 19), receptors and signaling pathways (n = 3), and subgroup analyses (n = 3). CONCLUSION In this appraisal, we have found some practical aspects that can help to the optimization of somatostatin analog (SSA) therapy in patients with well-differentiated GEP-NETs. We have also identified areas of uncertainty in an effort to guide clinical research in the coming years.
Collapse
Affiliation(s)
- Alberto Carmona-Bayonas
- Department of Hematology and Medical Oncology, Morales Meseguer University Hospital, Calle Marqués de los Vélez, s/n, CP 30008, Murcia, Spain.
| | | | - Ana Custodio
- Department of Medical Oncology, La Paz University Hospital, Madrid, Spain
| | - Enrique Grande
- Department of Medical Oncology, Ramón y Cajal University Hospital, Madrid, Spain
| | - Jaume Capdevila
- Department of Medical Oncology, Vall D'Hebrón University Hospital, Vall D'Hebrón Institute of Oncology (VHIO), Universitat Autònoma de Barcelona, center affiliated with the Red Temática de Investigación Cooperativa en Cáncer (RTICC), Instituto Carlos III, Spanish Ministry of Science and Innovation, Barcelona, Spain
| | - Carlos López
- Department of Medical Oncology, Marqués de Valdecilla University Hospital, Santander, Spain
| | - Alex Teule
- Department of Medical Oncology, Institut Català d'Oncologia, L'Hospitalet de Llobregat, center affiliated with the Red Temática de Investigación Cooperativa en Cáncer (RTICC), Instituto Carlos III, Spanish Ministry of Science and Innovation, Barcelona, Spain
| | - Rocío Garcia-Carbonero
- Department of Medical Oncology, Doce de Octubre University Hospital, center affiliated with the Red Temática de Investigación Cooperativa en Cáncer (RTICC), Instituto Carlos III, Spanish Ministry of Science and Innovation, Madrid, Spain
| | | |
Collapse
|
19
|
The North American Neuroendocrine Tumor Society Consensus Guidelines for Surveillance and Medical Management of Midgut Neuroendocrine Tumors. Pancreas 2017; 46:707-714. [PMID: 28609356 PMCID: PMC5642985 DOI: 10.1097/mpa.0000000000000850] [Citation(s) in RCA: 218] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
There have been significant developments in diagnostic and therapeutic options for patients with neuroendocrine tumors (NETs). Key phase 3 studies include the CLARINET trial, which evaluated lanreotide in patients with nonfunctioning enteropancreatic NETs; the RADIANT-2 and RADIANT-4 studies, which evaluated everolimus in functioning and nonfunctioning NETs of the gastrointestinal tract and lungs; the TELESTAR study, which evaluated telotristat ethyl in patients with refractory carcinoid syndrome; and the NETTER-1 trial, which evaluated Lu-DOTATATE in NETs of the small intestine and proximal colon (midgut). Based on these and other advances, the North American Neuroendocrine Tumor Society convened a multidisciplinary panel of experts with the goal of updating consensus-based guidelines for evaluation and treatment of midgut NETs. The medical aspects of these guidelines (focusing on systemic treatment, nonsurgical liver-directed therapy, and postoperative surveillance) are summarized in this article. Surgical guidelines are described in a companion article.
Collapse
|
20
|
Abstract
The lung is the second most common site of neuroendocrine tumors (NETs). Typical and atypical carcinoids are low-grade NETs of the lung. They present a favorable prognosis comported to the more common high-grade NETs. The low- and high-grade NETs require different treatment strategies; effective management of these tumors is essential to prolong survival and to manage the symptoms in patients with secretory or functional tumors. These rare tumors have received little attention and education is needed for treating physicians. This mini-review will concentrate mainly on advanced low-grade lung NETs. The article describes the classification of lung NETs and the diagnostic work-up. Different treatment methods including somatostatin analogs, peptide receptor radioligand therapy, and biologic systemic therapy are discussed. Promising results from recent trials are presented and discussed in the context of the lung primary site.
Collapse
Affiliation(s)
- Barbara Melosky
- Medical Oncology, British Columbia Cancer Agency – Vancouver Centre, Vancouver, BC, Canada
| |
Collapse
|
21
|
Ducreux M. Carcinoid syndrome in neuroendocrine tumors: a prognostic effect? Lancet Oncol 2017; 18:426-428. [DOI: 10.1016/s1470-2045(17)30159-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 02/09/2017] [Accepted: 02/13/2017] [Indexed: 11/28/2022]
|
22
|
Gut P, Waligórska-Stachura J, Czarnywojtek A, Sawicka-Gutaj N, Bączyk M, Ziemnicka K, Fischbach J, Woliński K, Kaznowski J, Wrotkowska E, Ruchała M. Management of the hormonal syndrome of neuroendocrine tumors. Arch Med Sci 2017; 13:515-524. [PMID: 28507564 PMCID: PMC5420621 DOI: 10.5114/aoms.2016.60311] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Accepted: 04/07/2016] [Indexed: 12/17/2022] Open
Abstract
Gastroenteropancreatic neuroendocrine tumors (GEP/NET) are unusual and rare neoplasms that present many clinical challenges. They characteristically synthesize store and secrete a variety of peptides and neuroamines which can lead to the development of distinct clinical syndrome, however many are clinically silent until late presentation with mass effects. Management strategies include surgery cure and cytoreduction with the use of somatostatin analogues. Somatostatin have a broad range of biological actions that include inhibition of exocrine and endocrine secretions, gut motility, cell proliferation, cell survival and angiogenesis. Five somatostatin receptors (SSTR1-SSTR5) have been cloned and characterized. Somatostatin analogues include octreotide and lanreotide are effective medical tools in the treatment and present selectivity for SSTR2 and SSTR5. During treatment is seen disapperance of flushing, normalization of bowel movements and reduction of serotonin and 5-hydroxyindole acetic acid (5-HIAA) secretion. Telotristat represents a novel approach by specifically inhibiting serotonin synthesis and as such, is a promising potential new treatment for patients with carcinoid syndrome. To pancreatic functionig neuroendocrine tumors belongs insulinoma, gastrinoma, glucagonoma and VIP-oma. Medical management in patients with insulinoma include diazoxide which suppresses insulin release. Also mTOR inhibitors may inhibit insulin secretion. Treatment of gastrinoma include both proton pump inhibitors (PPIs) and histamine H2 - receptor antagonists. In patients with glucagonomas hyperglycaemia can be controlled using insulin and oral blood glucose lowering drugs. In malignant glucagonomas smatostatin analogues are effective in controlling necrolytic migratory erythemia. Severe cases of the VIP-oma syndrome require supplementation of fluid losses. Octreotide reduce tumoral VIP secretion and control secretory diarrhoea.
Collapse
Affiliation(s)
- Paweł Gut
- Department of Endocrinology, Poznan University of Medical Sciences, Poznan, Poland
| | | | - Agata Czarnywojtek
- Department of Endocrinology, Poznan University of Medical Sciences, Poznan, Poland
| | - Nadia Sawicka-Gutaj
- Department of Endocrinology, Poznan University of Medical Sciences, Poznan, Poland
| | - Maciej Bączyk
- Department of Endocrinology, Poznan University of Medical Sciences, Poznan, Poland
| | - Katarzyna Ziemnicka
- Department of Endocrinology, Poznan University of Medical Sciences, Poznan, Poland
| | - Jakub Fischbach
- Department of Endocrinology, Poznan University of Medical Sciences, Poznan, Poland
| | - Kosma Woliński
- Department of Endocrinology, Poznan University of Medical Sciences, Poznan, Poland
| | - Jarosław Kaznowski
- Department of Endocrinology, Poznan University of Medical Sciences, Poznan, Poland
| | - Elżbieta Wrotkowska
- Department of Endocrinology, Poznan University of Medical Sciences, Poznan, Poland
| | - Marek Ruchała
- Department of Endocrinology, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
23
|
Michael M, Garcia-Carbonero R, Weber MM, Lombard-Bohas C, Toumpanakis C, Hicks RJ. The Antiproliferative Role of Lanreotide in Controlling Growth of Neuroendocrine Tumors: A Systematic Review. Oncologist 2017; 22:272-285. [PMID: 28220021 PMCID: PMC5344642 DOI: 10.1634/theoncologist.2016-0305] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 09/27/2016] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Neuroendocrine tumors (NETs) are a heterogeneous group of tumors, with >50% of cases involving the gastrointestinal system or pancreas. Somatostatin analogs (SSAs) are used for treating NET-related secretory syndromes and, more recently, for their antiproliferative effects. We conducted a systematic review of published literature on the antiproliferative efficacy and safety of the SSA lanreotide Autogel in the management of NETs to gain a fuller understanding of the evidence and identify future areas of research. METHODS Searches were conducted in PubMed up to March 16, 2016, and in the proceedings of four congresses from 2013 to 2016. RESULTS Screening of 1,132 publications identified in the searches found 40 relevant publications, including 27 full-length publications and 13 congress abstracts. Twenty-four of these publications reported antiproliferative efficacy data for lanreotide Autogel. The CLARINET study showed that 120 mg lanreotide Autogel every 4 weeks improves progression-free survival (PFS) in patients with gastroenteropancreatic (GEP)-NETs, with grade 1 or grade 2 (Ki-67 <10%) disease, providing class I evidence of its antiproliferative effects. The CLARINET open-label extension study reported a median PFS of 32.8 months with lanreotide Autogel. Other smaller studies generally support CLARINET. CONCLUSION Current clinical evidence shows that lanreotide Autogel has good antiproliferative activity with favorable safety and tolerability in patients with GEP-NETs, suggesting it should be considered as an early first-line treatment in this population. Further studies are needed to assess the potential benefits of higher doses and the use of lanreotide Autogel in combination therapy and as maintenance therapy in the absence of disease progression following other therapies. The Oncologist 2017;22:272-285 IMPLICATIONS FOR PRACTICE: This review presents the current clinical evidence for the antiproliferative activity of lanreotide Autogel in patients with midgut or pancreatic neuroendocrine tumors (NETs) and shows its effectiveness, safety, and tolerability in these patient populations. By systematically presenting all the clinical evidence, the review adds to existing publications by discussing results in a broad range of settings. The review also indicates future directions for investigation of the use of lanreotide Autogel in NETs originating in other locations, in combination therapy, or as maintenance therapy in progressive disease.
Collapse
Affiliation(s)
- Michael Michael
- Neuorendocrine Service & Division of Cancer Medicine, Peter MacCallum Cancer Centre, St. Andrews Place, East Melbourne, Australia
| | | | | | | | | | - Rodney J Hicks
- Cancer Imaging & Neuroendocrine Service & Molecular Imaging and Targeted Therapeutics Laboratory, Peter MacCallum Cancer Centre, St. Andrews Place, East Melbourne, Australia
| |
Collapse
|
24
|
Kulke MH, Hörsch D, Caplin ME, Anthony LB, Bergsland E, Öberg K, Welin S, Warner RR, Lombard-Bohas C, Kunz PL, Grande E, Valle JW, Fleming D, Lapuerta P, Banks P, Jackson S, Zambrowicz B, Sands AT, Pavel M. Telotristat Ethyl, a Tryptophan Hydroxylase Inhibitor for the Treatment of Carcinoid Syndrome. J Clin Oncol 2017; 35:14-23. [DOI: 10.1200/jco.2016.69.2780] [Citation(s) in RCA: 209] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Purpose Preliminary studies suggested that telotristat ethyl, a tryptophan hydroxylase inhibitor, reduces bowel movement (BM) frequency in patients with carcinoid syndrome. This placebo-controlled phase III study evaluated telotristat ethyl in this setting. Patients and Methods Patients (N = 135) experiencing four or more BMs per day despite stable-dose somatostatin analog therapy received (1:1:1) placebo, telotristat ethyl 250 mg, or telotristat ethyl 500 mg three times per day orally during a 12-week double-blind treatment period. The primary end point was change from baseline in BM frequency. In an open-label extension, 115 patients subsequently received telotristat ethyl 500 mg. Results Estimated differences in BM frequency per day versus placebo averaged over 12 weeks were –0.81 for telotristat ethyl 250 mg ( P < .001) and ‒0.69 for telotristat ethyl 500 mg ( P < .001). At week 12, mean BM frequency reductions per day for placebo, telotristat ethyl 250 mg, and telotristat ethyl 500 mg were –0.9, –1.7, and –2.1, respectively. Responses, predefined as a BM frequency reduction ≥ 30% from baseline for ≥ 50% of the double-blind treatment period, were observed in 20%, 44%, and 42% of patients given placebo, telotristat ethyl 250 mg, and telotristat ethyl 500 mg, respectively. Both telotristat ethyl dosages significantly reduced mean urinary 5-hydroxyindole acetic acid versus placebo at week 12 ( P < .001). Mild nausea and asymptomatic increases in gamma-glutamyl transferase were observed in some patients receiving telotristat ethyl. Follow-up of patients during the open-label extension revealed no new safety signals and suggested sustained BM responses to treatment. Conclusion Among patients with carcinoid syndrome not adequately controlled by somatostatin analogs, treatment with telotristat ethyl was generally safe and well tolerated and resulted in significant reductions in BM frequency and urinary 5-hydroxyindole acetic acid.
Collapse
Affiliation(s)
- Matthew H. Kulke
- Matthew H. Kulke, Dana-Farber Cancer Institute, Boston; Douglas Fleming, Ipsen Bioscience, Cambridge, MA; Dieter Hörsch, Zentralklinik Bad Berka, Bad Berka; Marianne Pavel, Charité-Universitätsmedizin, Berlin, Germany; Martyn E. Caplin, Royal Free Hospital, London; Juan W. Valle, The University of Manchester-The Christie National Health Service Foundation Trust, Manchester, United Kingdom; Lowell B. Anthony, University of Kentucky, Lexington, KY; Emily Bergsland, University of California at San Francisco
| | - Dieter Hörsch
- Matthew H. Kulke, Dana-Farber Cancer Institute, Boston; Douglas Fleming, Ipsen Bioscience, Cambridge, MA; Dieter Hörsch, Zentralklinik Bad Berka, Bad Berka; Marianne Pavel, Charité-Universitätsmedizin, Berlin, Germany; Martyn E. Caplin, Royal Free Hospital, London; Juan W. Valle, The University of Manchester-The Christie National Health Service Foundation Trust, Manchester, United Kingdom; Lowell B. Anthony, University of Kentucky, Lexington, KY; Emily Bergsland, University of California at San Francisco
| | - Martyn E. Caplin
- Matthew H. Kulke, Dana-Farber Cancer Institute, Boston; Douglas Fleming, Ipsen Bioscience, Cambridge, MA; Dieter Hörsch, Zentralklinik Bad Berka, Bad Berka; Marianne Pavel, Charité-Universitätsmedizin, Berlin, Germany; Martyn E. Caplin, Royal Free Hospital, London; Juan W. Valle, The University of Manchester-The Christie National Health Service Foundation Trust, Manchester, United Kingdom; Lowell B. Anthony, University of Kentucky, Lexington, KY; Emily Bergsland, University of California at San Francisco
| | - Lowell B. Anthony
- Matthew H. Kulke, Dana-Farber Cancer Institute, Boston; Douglas Fleming, Ipsen Bioscience, Cambridge, MA; Dieter Hörsch, Zentralklinik Bad Berka, Bad Berka; Marianne Pavel, Charité-Universitätsmedizin, Berlin, Germany; Martyn E. Caplin, Royal Free Hospital, London; Juan W. Valle, The University of Manchester-The Christie National Health Service Foundation Trust, Manchester, United Kingdom; Lowell B. Anthony, University of Kentucky, Lexington, KY; Emily Bergsland, University of California at San Francisco
| | - Emily Bergsland
- Matthew H. Kulke, Dana-Farber Cancer Institute, Boston; Douglas Fleming, Ipsen Bioscience, Cambridge, MA; Dieter Hörsch, Zentralklinik Bad Berka, Bad Berka; Marianne Pavel, Charité-Universitätsmedizin, Berlin, Germany; Martyn E. Caplin, Royal Free Hospital, London; Juan W. Valle, The University of Manchester-The Christie National Health Service Foundation Trust, Manchester, United Kingdom; Lowell B. Anthony, University of Kentucky, Lexington, KY; Emily Bergsland, University of California at San Francisco
| | - Kjell Öberg
- Matthew H. Kulke, Dana-Farber Cancer Institute, Boston; Douglas Fleming, Ipsen Bioscience, Cambridge, MA; Dieter Hörsch, Zentralklinik Bad Berka, Bad Berka; Marianne Pavel, Charité-Universitätsmedizin, Berlin, Germany; Martyn E. Caplin, Royal Free Hospital, London; Juan W. Valle, The University of Manchester-The Christie National Health Service Foundation Trust, Manchester, United Kingdom; Lowell B. Anthony, University of Kentucky, Lexington, KY; Emily Bergsland, University of California at San Francisco
| | - Staffan Welin
- Matthew H. Kulke, Dana-Farber Cancer Institute, Boston; Douglas Fleming, Ipsen Bioscience, Cambridge, MA; Dieter Hörsch, Zentralklinik Bad Berka, Bad Berka; Marianne Pavel, Charité-Universitätsmedizin, Berlin, Germany; Martyn E. Caplin, Royal Free Hospital, London; Juan W. Valle, The University of Manchester-The Christie National Health Service Foundation Trust, Manchester, United Kingdom; Lowell B. Anthony, University of Kentucky, Lexington, KY; Emily Bergsland, University of California at San Francisco
| | - Richard R.P. Warner
- Matthew H. Kulke, Dana-Farber Cancer Institute, Boston; Douglas Fleming, Ipsen Bioscience, Cambridge, MA; Dieter Hörsch, Zentralklinik Bad Berka, Bad Berka; Marianne Pavel, Charité-Universitätsmedizin, Berlin, Germany; Martyn E. Caplin, Royal Free Hospital, London; Juan W. Valle, The University of Manchester-The Christie National Health Service Foundation Trust, Manchester, United Kingdom; Lowell B. Anthony, University of Kentucky, Lexington, KY; Emily Bergsland, University of California at San Francisco
| | - Catherine Lombard-Bohas
- Matthew H. Kulke, Dana-Farber Cancer Institute, Boston; Douglas Fleming, Ipsen Bioscience, Cambridge, MA; Dieter Hörsch, Zentralklinik Bad Berka, Bad Berka; Marianne Pavel, Charité-Universitätsmedizin, Berlin, Germany; Martyn E. Caplin, Royal Free Hospital, London; Juan W. Valle, The University of Manchester-The Christie National Health Service Foundation Trust, Manchester, United Kingdom; Lowell B. Anthony, University of Kentucky, Lexington, KY; Emily Bergsland, University of California at San Francisco
| | - Pamela L. Kunz
- Matthew H. Kulke, Dana-Farber Cancer Institute, Boston; Douglas Fleming, Ipsen Bioscience, Cambridge, MA; Dieter Hörsch, Zentralklinik Bad Berka, Bad Berka; Marianne Pavel, Charité-Universitätsmedizin, Berlin, Germany; Martyn E. Caplin, Royal Free Hospital, London; Juan W. Valle, The University of Manchester-The Christie National Health Service Foundation Trust, Manchester, United Kingdom; Lowell B. Anthony, University of Kentucky, Lexington, KY; Emily Bergsland, University of California at San Francisco
| | - Enrique Grande
- Matthew H. Kulke, Dana-Farber Cancer Institute, Boston; Douglas Fleming, Ipsen Bioscience, Cambridge, MA; Dieter Hörsch, Zentralklinik Bad Berka, Bad Berka; Marianne Pavel, Charité-Universitätsmedizin, Berlin, Germany; Martyn E. Caplin, Royal Free Hospital, London; Juan W. Valle, The University of Manchester-The Christie National Health Service Foundation Trust, Manchester, United Kingdom; Lowell B. Anthony, University of Kentucky, Lexington, KY; Emily Bergsland, University of California at San Francisco
| | - Juan W. Valle
- Matthew H. Kulke, Dana-Farber Cancer Institute, Boston; Douglas Fleming, Ipsen Bioscience, Cambridge, MA; Dieter Hörsch, Zentralklinik Bad Berka, Bad Berka; Marianne Pavel, Charité-Universitätsmedizin, Berlin, Germany; Martyn E. Caplin, Royal Free Hospital, London; Juan W. Valle, The University of Manchester-The Christie National Health Service Foundation Trust, Manchester, United Kingdom; Lowell B. Anthony, University of Kentucky, Lexington, KY; Emily Bergsland, University of California at San Francisco
| | - Douglas Fleming
- Matthew H. Kulke, Dana-Farber Cancer Institute, Boston; Douglas Fleming, Ipsen Bioscience, Cambridge, MA; Dieter Hörsch, Zentralklinik Bad Berka, Bad Berka; Marianne Pavel, Charité-Universitätsmedizin, Berlin, Germany; Martyn E. Caplin, Royal Free Hospital, London; Juan W. Valle, The University of Manchester-The Christie National Health Service Foundation Trust, Manchester, United Kingdom; Lowell B. Anthony, University of Kentucky, Lexington, KY; Emily Bergsland, University of California at San Francisco
| | - Pablo Lapuerta
- Matthew H. Kulke, Dana-Farber Cancer Institute, Boston; Douglas Fleming, Ipsen Bioscience, Cambridge, MA; Dieter Hörsch, Zentralklinik Bad Berka, Bad Berka; Marianne Pavel, Charité-Universitätsmedizin, Berlin, Germany; Martyn E. Caplin, Royal Free Hospital, London; Juan W. Valle, The University of Manchester-The Christie National Health Service Foundation Trust, Manchester, United Kingdom; Lowell B. Anthony, University of Kentucky, Lexington, KY; Emily Bergsland, University of California at San Francisco
| | - Phillip Banks
- Matthew H. Kulke, Dana-Farber Cancer Institute, Boston; Douglas Fleming, Ipsen Bioscience, Cambridge, MA; Dieter Hörsch, Zentralklinik Bad Berka, Bad Berka; Marianne Pavel, Charité-Universitätsmedizin, Berlin, Germany; Martyn E. Caplin, Royal Free Hospital, London; Juan W. Valle, The University of Manchester-The Christie National Health Service Foundation Trust, Manchester, United Kingdom; Lowell B. Anthony, University of Kentucky, Lexington, KY; Emily Bergsland, University of California at San Francisco
| | - Shanna Jackson
- Matthew H. Kulke, Dana-Farber Cancer Institute, Boston; Douglas Fleming, Ipsen Bioscience, Cambridge, MA; Dieter Hörsch, Zentralklinik Bad Berka, Bad Berka; Marianne Pavel, Charité-Universitätsmedizin, Berlin, Germany; Martyn E. Caplin, Royal Free Hospital, London; Juan W. Valle, The University of Manchester-The Christie National Health Service Foundation Trust, Manchester, United Kingdom; Lowell B. Anthony, University of Kentucky, Lexington, KY; Emily Bergsland, University of California at San Francisco
| | - Brian Zambrowicz
- Matthew H. Kulke, Dana-Farber Cancer Institute, Boston; Douglas Fleming, Ipsen Bioscience, Cambridge, MA; Dieter Hörsch, Zentralklinik Bad Berka, Bad Berka; Marianne Pavel, Charité-Universitätsmedizin, Berlin, Germany; Martyn E. Caplin, Royal Free Hospital, London; Juan W. Valle, The University of Manchester-The Christie National Health Service Foundation Trust, Manchester, United Kingdom; Lowell B. Anthony, University of Kentucky, Lexington, KY; Emily Bergsland, University of California at San Francisco
| | - Arthur T. Sands
- Matthew H. Kulke, Dana-Farber Cancer Institute, Boston; Douglas Fleming, Ipsen Bioscience, Cambridge, MA; Dieter Hörsch, Zentralklinik Bad Berka, Bad Berka; Marianne Pavel, Charité-Universitätsmedizin, Berlin, Germany; Martyn E. Caplin, Royal Free Hospital, London; Juan W. Valle, The University of Manchester-The Christie National Health Service Foundation Trust, Manchester, United Kingdom; Lowell B. Anthony, University of Kentucky, Lexington, KY; Emily Bergsland, University of California at San Francisco
| | - Marianne Pavel
- Matthew H. Kulke, Dana-Farber Cancer Institute, Boston; Douglas Fleming, Ipsen Bioscience, Cambridge, MA; Dieter Hörsch, Zentralklinik Bad Berka, Bad Berka; Marianne Pavel, Charité-Universitätsmedizin, Berlin, Germany; Martyn E. Caplin, Royal Free Hospital, London; Juan W. Valle, The University of Manchester-The Christie National Health Service Foundation Trust, Manchester, United Kingdom; Lowell B. Anthony, University of Kentucky, Lexington, KY; Emily Bergsland, University of California at San Francisco
| |
Collapse
|
25
|
Lamarca A, Barriuso J, McNamara MG, Hubner RA, Valle JW. Telotristat ethyl: a new option for the management of carcinoid syndrome. Expert Opin Pharmacother 2016; 17:2487-2498. [PMID: 27817224 DOI: 10.1080/14656566.2016.1254191] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Many patients with neuroendocrine tumour-related carcinoid syndrome treated with somatostatin analogues (SSA) won't achieve adequate symptom relief with the SSA alone; new treatment options are required. Telotristat ethyl is a tryptophan hydroxylase inhibitor, developed for the treatment of carcinoid syndrome. Areas covered: This review summarises the evidence supporting the role of telotristat ethyl in the management of carcinoid syndrome. Rationale, pharmacodynamics, pharmacokinetics, metabolism, clinical experience, efficacy and toxicity profiles are covered. Expert opinion: The efficacy of telotristat ethyl in producing a statistically-significant and clinically-meaningful reduction in daily bowel movements has been confirmed in phase III clinical trials. Two pivotal trials, TELESTAR and TELECAST, explored the role of telotristat ethyl in the management of patients with carcinoid syndrome refractory to SSAs focusing on patients with ≥4 and <4 daily bowel movements, respectively. In addition, benefit was confirmed in patient-reported outcomes. Based on activity and safe toxicity profile, telotristat ethyl is pending regulatory agencies evaluation and is likely to add to the armamentarium used to treat carcinoid syndrome. Long-term safety and efficacy data will be available from the ongoing TELEPATH study. The impact on carcinoid heart disease, mesenteric fibrosis and other long-term complications of carcinoid syndrome as well as its role earlier in patients' pathways remain investigational.
Collapse
Affiliation(s)
- Angela Lamarca
- a Department of Medical Oncology , The Christie NHS Foundation Trust , Manchester , UK
| | - Jorge Barriuso
- a Department of Medical Oncology , The Christie NHS Foundation Trust , Manchester , UK.,b Faculty of Medical, Biological and Human Sciences , University of Manchester , Manchester , UK
| | - Mairéad G McNamara
- a Department of Medical Oncology , The Christie NHS Foundation Trust , Manchester , UK.,c Institute of Cancer Sciences , University of Manchester , Manchester , UK
| | - Richard A Hubner
- a Department of Medical Oncology , The Christie NHS Foundation Trust , Manchester , UK
| | - Juan W Valle
- a Department of Medical Oncology , The Christie NHS Foundation Trust , Manchester , UK.,c Institute of Cancer Sciences , University of Manchester , Manchester , UK
| |
Collapse
|
26
|
Pokuri VK, Fong MK, Iyer R. Octreotide and Lanreotide in Gastroenteropancreatic Neuroendocrine Tumors. Curr Oncol Rep 2016; 18:7. [PMID: 26743514 DOI: 10.1007/s11912-015-0492-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Neuroendocrine tumors are heterogeneous, rare malignancies that arise most commonly in the gastrointestinal tract and pancreas. They often secrete vasoactive substances resulting in carcinoid syndrome and the tumor cells exclusively express somatostatin receptors. Octreotide and lanreotide are the two synthetic somatostatin analogs used for the control of carcinoid symptoms and tumor progression in advanced inoperable disease. Recent pivotal trials (PROMID and CLARINET studies) established their antitumor activity. We discuss the available data to support their use as symptom controlling and antiproliferative agents. This article also reviews the guidelines (National Comprehensive Cancer Network and North American Neuro Endocrine Tumor Society), cost-analysis (suggesting the cost-effectiveness of lanreotide autogel compared to higher doses of octreotide long acting release formulation in refractory patients), and future directions of somatostatin analogs in the management of patients refractory to conventional doses of octreotide and lanreotide.
Collapse
Affiliation(s)
- Venkata K Pokuri
- Department of Medical Oncology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Mei Ka Fong
- Department of Pharmacy, Carolinas Healthcare System, Levine Cancer Institute, Charlotte, NC, USA
| | - Renuka Iyer
- Department of Medical Oncology, Roswell Park Cancer Institute, Buffalo, NY, USA.
| |
Collapse
|
27
|
Fotouhi O, Kjellin H, Larsson C, Hashemi J, Barriuso J, Juhlin CC, Lu M, Höög A, Pastrián LG, Lamarca A, Soto VH, Zedenius J, Mendiola M, Lehtiö J, Kjellman M. Proteomics Suggests a Role for APC-Survivin in Response to Somatostatin Analog Treatment of Neuroendocrine Tumors. J Clin Endocrinol Metab 2016; 101:3616-3627. [PMID: 27459532 PMCID: PMC5052342 DOI: 10.1210/jc.2016-2028] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
CONTEXT Somatostatin analogs are established in the treatment of neuroendocrine tumors (NETs) including small intestinal NET; however, the molecular mechanisms are not well known. Here, we examined the direct effects of lanreotide in NET cell line models. SETTING AND DESIGN The cell lines HC45 and H727 were treated with 10nM lanreotide for different time periods and alterations of the proteome were analyzed by in-depth high-resolution isoelectric focusing tandem liquid chromatography-mass spectrometry. We next investigated whether the observed suppression of survivin was mediated by adenomatous polyposis coli (APC) and possible effects on tumor proliferation in vitro. Expression of survivin was assessed by immunohistochemistry in 112 NET cases and compared with patient outcome. RESULTS We quantified 6451 and 7801 proteins in HC45 and H727, respectively. After short time lanreotide treatment APC was increased and survivin reduced. Overexpression of APC in H727 cells decreased, and APC knock-down elevated the survivin level. The lanreotide regulation of APC-survivin could be suppressed by small interfering RNA against somatostatin receptor 2. Although lanreotide only gave slight inhibition of proliferation, targeting of survivin with the small molecule YM155 dramatically reduced proliferation. Moderate or high as compared with low or absent total survivin expression was associated with shorter progression-free survival, independent of tumor stage, grade, and localization. CONCLUSIONS We report a proteome-wide analysis of changes in response to lanreotide in NET cell lines. This analysis suggests a connection between somatostatin analog, APC, and survivin levels. Survivin is a possible prognostic factor and a new potential therapeutic target in NETs.
Collapse
Affiliation(s)
- Omid Fotouhi
- Departments of Oncology-Pathology (O.F., C.L., J.H., C.C.J., M.L., A.H., J.L.) and Molecular Medicine and Surgery (H.K., J.Z., M.K.), Karolinska Institutet, Stockholm, Sweden; Cancer Center Karolinska (O.F., C.L., J.H., C.C.J., M.L., A.H.), Karolinska University Hospital, Stockholm, Sweden; Cancer Proteomics Mass Spectrometry (H.K., J.L.), Science for Life Laboratory, Stockholm, Sweden SE-171 76; Faculty of Biology (J.B.), Medicine and Health, University of Manchester, M13 9PT, Manchester, United Kingdom; Laboratory of Molecular Pathology and Therapeutic Targets, and Translational Oncology Research Group (J.B., A.L., V.H.S., M.M.), Instituto de Investigación; Department of Pathology (L.G.P.); and Molecular Pathology Section (M.M.), Instituto de Genética Médica, Hospital Universitario La Paz 28046, Madrid, Spain; and Department of Medical Oncology (A.L.), The Christie NHS Trust, M20 4BX, Manchester, United Kingdom
| | - Hanna Kjellin
- Departments of Oncology-Pathology (O.F., C.L., J.H., C.C.J., M.L., A.H., J.L.) and Molecular Medicine and Surgery (H.K., J.Z., M.K.), Karolinska Institutet, Stockholm, Sweden; Cancer Center Karolinska (O.F., C.L., J.H., C.C.J., M.L., A.H.), Karolinska University Hospital, Stockholm, Sweden; Cancer Proteomics Mass Spectrometry (H.K., J.L.), Science for Life Laboratory, Stockholm, Sweden SE-171 76; Faculty of Biology (J.B.), Medicine and Health, University of Manchester, M13 9PT, Manchester, United Kingdom; Laboratory of Molecular Pathology and Therapeutic Targets, and Translational Oncology Research Group (J.B., A.L., V.H.S., M.M.), Instituto de Investigación; Department of Pathology (L.G.P.); and Molecular Pathology Section (M.M.), Instituto de Genética Médica, Hospital Universitario La Paz 28046, Madrid, Spain; and Department of Medical Oncology (A.L.), The Christie NHS Trust, M20 4BX, Manchester, United Kingdom
| | - Catharina Larsson
- Departments of Oncology-Pathology (O.F., C.L., J.H., C.C.J., M.L., A.H., J.L.) and Molecular Medicine and Surgery (H.K., J.Z., M.K.), Karolinska Institutet, Stockholm, Sweden; Cancer Center Karolinska (O.F., C.L., J.H., C.C.J., M.L., A.H.), Karolinska University Hospital, Stockholm, Sweden; Cancer Proteomics Mass Spectrometry (H.K., J.L.), Science for Life Laboratory, Stockholm, Sweden SE-171 76; Faculty of Biology (J.B.), Medicine and Health, University of Manchester, M13 9PT, Manchester, United Kingdom; Laboratory of Molecular Pathology and Therapeutic Targets, and Translational Oncology Research Group (J.B., A.L., V.H.S., M.M.), Instituto de Investigación; Department of Pathology (L.G.P.); and Molecular Pathology Section (M.M.), Instituto de Genética Médica, Hospital Universitario La Paz 28046, Madrid, Spain; and Department of Medical Oncology (A.L.), The Christie NHS Trust, M20 4BX, Manchester, United Kingdom
| | - Jamileh Hashemi
- Departments of Oncology-Pathology (O.F., C.L., J.H., C.C.J., M.L., A.H., J.L.) and Molecular Medicine and Surgery (H.K., J.Z., M.K.), Karolinska Institutet, Stockholm, Sweden; Cancer Center Karolinska (O.F., C.L., J.H., C.C.J., M.L., A.H.), Karolinska University Hospital, Stockholm, Sweden; Cancer Proteomics Mass Spectrometry (H.K., J.L.), Science for Life Laboratory, Stockholm, Sweden SE-171 76; Faculty of Biology (J.B.), Medicine and Health, University of Manchester, M13 9PT, Manchester, United Kingdom; Laboratory of Molecular Pathology and Therapeutic Targets, and Translational Oncology Research Group (J.B., A.L., V.H.S., M.M.), Instituto de Investigación; Department of Pathology (L.G.P.); and Molecular Pathology Section (M.M.), Instituto de Genética Médica, Hospital Universitario La Paz 28046, Madrid, Spain; and Department of Medical Oncology (A.L.), The Christie NHS Trust, M20 4BX, Manchester, United Kingdom
| | - Jorge Barriuso
- Departments of Oncology-Pathology (O.F., C.L., J.H., C.C.J., M.L., A.H., J.L.) and Molecular Medicine and Surgery (H.K., J.Z., M.K.), Karolinska Institutet, Stockholm, Sweden; Cancer Center Karolinska (O.F., C.L., J.H., C.C.J., M.L., A.H.), Karolinska University Hospital, Stockholm, Sweden; Cancer Proteomics Mass Spectrometry (H.K., J.L.), Science for Life Laboratory, Stockholm, Sweden SE-171 76; Faculty of Biology (J.B.), Medicine and Health, University of Manchester, M13 9PT, Manchester, United Kingdom; Laboratory of Molecular Pathology and Therapeutic Targets, and Translational Oncology Research Group (J.B., A.L., V.H.S., M.M.), Instituto de Investigación; Department of Pathology (L.G.P.); and Molecular Pathology Section (M.M.), Instituto de Genética Médica, Hospital Universitario La Paz 28046, Madrid, Spain; and Department of Medical Oncology (A.L.), The Christie NHS Trust, M20 4BX, Manchester, United Kingdom
| | - C Christofer Juhlin
- Departments of Oncology-Pathology (O.F., C.L., J.H., C.C.J., M.L., A.H., J.L.) and Molecular Medicine and Surgery (H.K., J.Z., M.K.), Karolinska Institutet, Stockholm, Sweden; Cancer Center Karolinska (O.F., C.L., J.H., C.C.J., M.L., A.H.), Karolinska University Hospital, Stockholm, Sweden; Cancer Proteomics Mass Spectrometry (H.K., J.L.), Science for Life Laboratory, Stockholm, Sweden SE-171 76; Faculty of Biology (J.B.), Medicine and Health, University of Manchester, M13 9PT, Manchester, United Kingdom; Laboratory of Molecular Pathology and Therapeutic Targets, and Translational Oncology Research Group (J.B., A.L., V.H.S., M.M.), Instituto de Investigación; Department of Pathology (L.G.P.); and Molecular Pathology Section (M.M.), Instituto de Genética Médica, Hospital Universitario La Paz 28046, Madrid, Spain; and Department of Medical Oncology (A.L.), The Christie NHS Trust, M20 4BX, Manchester, United Kingdom
| | - Ming Lu
- Departments of Oncology-Pathology (O.F., C.L., J.H., C.C.J., M.L., A.H., J.L.) and Molecular Medicine and Surgery (H.K., J.Z., M.K.), Karolinska Institutet, Stockholm, Sweden; Cancer Center Karolinska (O.F., C.L., J.H., C.C.J., M.L., A.H.), Karolinska University Hospital, Stockholm, Sweden; Cancer Proteomics Mass Spectrometry (H.K., J.L.), Science for Life Laboratory, Stockholm, Sweden SE-171 76; Faculty of Biology (J.B.), Medicine and Health, University of Manchester, M13 9PT, Manchester, United Kingdom; Laboratory of Molecular Pathology and Therapeutic Targets, and Translational Oncology Research Group (J.B., A.L., V.H.S., M.M.), Instituto de Investigación; Department of Pathology (L.G.P.); and Molecular Pathology Section (M.M.), Instituto de Genética Médica, Hospital Universitario La Paz 28046, Madrid, Spain; and Department of Medical Oncology (A.L.), The Christie NHS Trust, M20 4BX, Manchester, United Kingdom
| | - Anders Höög
- Departments of Oncology-Pathology (O.F., C.L., J.H., C.C.J., M.L., A.H., J.L.) and Molecular Medicine and Surgery (H.K., J.Z., M.K.), Karolinska Institutet, Stockholm, Sweden; Cancer Center Karolinska (O.F., C.L., J.H., C.C.J., M.L., A.H.), Karolinska University Hospital, Stockholm, Sweden; Cancer Proteomics Mass Spectrometry (H.K., J.L.), Science for Life Laboratory, Stockholm, Sweden SE-171 76; Faculty of Biology (J.B.), Medicine and Health, University of Manchester, M13 9PT, Manchester, United Kingdom; Laboratory of Molecular Pathology and Therapeutic Targets, and Translational Oncology Research Group (J.B., A.L., V.H.S., M.M.), Instituto de Investigación; Department of Pathology (L.G.P.); and Molecular Pathology Section (M.M.), Instituto de Genética Médica, Hospital Universitario La Paz 28046, Madrid, Spain; and Department of Medical Oncology (A.L.), The Christie NHS Trust, M20 4BX, Manchester, United Kingdom
| | - Laura G Pastrián
- Departments of Oncology-Pathology (O.F., C.L., J.H., C.C.J., M.L., A.H., J.L.) and Molecular Medicine and Surgery (H.K., J.Z., M.K.), Karolinska Institutet, Stockholm, Sweden; Cancer Center Karolinska (O.F., C.L., J.H., C.C.J., M.L., A.H.), Karolinska University Hospital, Stockholm, Sweden; Cancer Proteomics Mass Spectrometry (H.K., J.L.), Science for Life Laboratory, Stockholm, Sweden SE-171 76; Faculty of Biology (J.B.), Medicine and Health, University of Manchester, M13 9PT, Manchester, United Kingdom; Laboratory of Molecular Pathology and Therapeutic Targets, and Translational Oncology Research Group (J.B., A.L., V.H.S., M.M.), Instituto de Investigación; Department of Pathology (L.G.P.); and Molecular Pathology Section (M.M.), Instituto de Genética Médica, Hospital Universitario La Paz 28046, Madrid, Spain; and Department of Medical Oncology (A.L.), The Christie NHS Trust, M20 4BX, Manchester, United Kingdom
| | - Angela Lamarca
- Departments of Oncology-Pathology (O.F., C.L., J.H., C.C.J., M.L., A.H., J.L.) and Molecular Medicine and Surgery (H.K., J.Z., M.K.), Karolinska Institutet, Stockholm, Sweden; Cancer Center Karolinska (O.F., C.L., J.H., C.C.J., M.L., A.H.), Karolinska University Hospital, Stockholm, Sweden; Cancer Proteomics Mass Spectrometry (H.K., J.L.), Science for Life Laboratory, Stockholm, Sweden SE-171 76; Faculty of Biology (J.B.), Medicine and Health, University of Manchester, M13 9PT, Manchester, United Kingdom; Laboratory of Molecular Pathology and Therapeutic Targets, and Translational Oncology Research Group (J.B., A.L., V.H.S., M.M.), Instituto de Investigación; Department of Pathology (L.G.P.); and Molecular Pathology Section (M.M.), Instituto de Genética Médica, Hospital Universitario La Paz 28046, Madrid, Spain; and Department of Medical Oncology (A.L.), The Christie NHS Trust, M20 4BX, Manchester, United Kingdom
| | - Victoria Heredia Soto
- Departments of Oncology-Pathology (O.F., C.L., J.H., C.C.J., M.L., A.H., J.L.) and Molecular Medicine and Surgery (H.K., J.Z., M.K.), Karolinska Institutet, Stockholm, Sweden; Cancer Center Karolinska (O.F., C.L., J.H., C.C.J., M.L., A.H.), Karolinska University Hospital, Stockholm, Sweden; Cancer Proteomics Mass Spectrometry (H.K., J.L.), Science for Life Laboratory, Stockholm, Sweden SE-171 76; Faculty of Biology (J.B.), Medicine and Health, University of Manchester, M13 9PT, Manchester, United Kingdom; Laboratory of Molecular Pathology and Therapeutic Targets, and Translational Oncology Research Group (J.B., A.L., V.H.S., M.M.), Instituto de Investigación; Department of Pathology (L.G.P.); and Molecular Pathology Section (M.M.), Instituto de Genética Médica, Hospital Universitario La Paz 28046, Madrid, Spain; and Department of Medical Oncology (A.L.), The Christie NHS Trust, M20 4BX, Manchester, United Kingdom
| | - Jan Zedenius
- Departments of Oncology-Pathology (O.F., C.L., J.H., C.C.J., M.L., A.H., J.L.) and Molecular Medicine and Surgery (H.K., J.Z., M.K.), Karolinska Institutet, Stockholm, Sweden; Cancer Center Karolinska (O.F., C.L., J.H., C.C.J., M.L., A.H.), Karolinska University Hospital, Stockholm, Sweden; Cancer Proteomics Mass Spectrometry (H.K., J.L.), Science for Life Laboratory, Stockholm, Sweden SE-171 76; Faculty of Biology (J.B.), Medicine and Health, University of Manchester, M13 9PT, Manchester, United Kingdom; Laboratory of Molecular Pathology and Therapeutic Targets, and Translational Oncology Research Group (J.B., A.L., V.H.S., M.M.), Instituto de Investigación; Department of Pathology (L.G.P.); and Molecular Pathology Section (M.M.), Instituto de Genética Médica, Hospital Universitario La Paz 28046, Madrid, Spain; and Department of Medical Oncology (A.L.), The Christie NHS Trust, M20 4BX, Manchester, United Kingdom
| | - Marta Mendiola
- Departments of Oncology-Pathology (O.F., C.L., J.H., C.C.J., M.L., A.H., J.L.) and Molecular Medicine and Surgery (H.K., J.Z., M.K.), Karolinska Institutet, Stockholm, Sweden; Cancer Center Karolinska (O.F., C.L., J.H., C.C.J., M.L., A.H.), Karolinska University Hospital, Stockholm, Sweden; Cancer Proteomics Mass Spectrometry (H.K., J.L.), Science for Life Laboratory, Stockholm, Sweden SE-171 76; Faculty of Biology (J.B.), Medicine and Health, University of Manchester, M13 9PT, Manchester, United Kingdom; Laboratory of Molecular Pathology and Therapeutic Targets, and Translational Oncology Research Group (J.B., A.L., V.H.S., M.M.), Instituto de Investigación; Department of Pathology (L.G.P.); and Molecular Pathology Section (M.M.), Instituto de Genética Médica, Hospital Universitario La Paz 28046, Madrid, Spain; and Department of Medical Oncology (A.L.), The Christie NHS Trust, M20 4BX, Manchester, United Kingdom
| | - Janne Lehtiö
- Departments of Oncology-Pathology (O.F., C.L., J.H., C.C.J., M.L., A.H., J.L.) and Molecular Medicine and Surgery (H.K., J.Z., M.K.), Karolinska Institutet, Stockholm, Sweden; Cancer Center Karolinska (O.F., C.L., J.H., C.C.J., M.L., A.H.), Karolinska University Hospital, Stockholm, Sweden; Cancer Proteomics Mass Spectrometry (H.K., J.L.), Science for Life Laboratory, Stockholm, Sweden SE-171 76; Faculty of Biology (J.B.), Medicine and Health, University of Manchester, M13 9PT, Manchester, United Kingdom; Laboratory of Molecular Pathology and Therapeutic Targets, and Translational Oncology Research Group (J.B., A.L., V.H.S., M.M.), Instituto de Investigación; Department of Pathology (L.G.P.); and Molecular Pathology Section (M.M.), Instituto de Genética Médica, Hospital Universitario La Paz 28046, Madrid, Spain; and Department of Medical Oncology (A.L.), The Christie NHS Trust, M20 4BX, Manchester, United Kingdom
| | - Magnus Kjellman
- Departments of Oncology-Pathology (O.F., C.L., J.H., C.C.J., M.L., A.H., J.L.) and Molecular Medicine and Surgery (H.K., J.Z., M.K.), Karolinska Institutet, Stockholm, Sweden; Cancer Center Karolinska (O.F., C.L., J.H., C.C.J., M.L., A.H.), Karolinska University Hospital, Stockholm, Sweden; Cancer Proteomics Mass Spectrometry (H.K., J.L.), Science for Life Laboratory, Stockholm, Sweden SE-171 76; Faculty of Biology (J.B.), Medicine and Health, University of Manchester, M13 9PT, Manchester, United Kingdom; Laboratory of Molecular Pathology and Therapeutic Targets, and Translational Oncology Research Group (J.B., A.L., V.H.S., M.M.), Instituto de Investigación; Department of Pathology (L.G.P.); and Molecular Pathology Section (M.M.), Instituto de Genética Médica, Hospital Universitario La Paz 28046, Madrid, Spain; and Department of Medical Oncology (A.L.), The Christie NHS Trust, M20 4BX, Manchester, United Kingdom
| |
Collapse
|
28
|
Mulvey CK, Bergsland EK. Systemic Therapies for Advanced Gastrointestinal Carcinoid Tumors. Hematol Oncol Clin North Am 2016; 30:63-82. [PMID: 26614369 DOI: 10.1016/j.hoc.2015.09.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Well-differentiated gastrointestinal neuroendocrine tumors (GINETs) tend to be slow growing, but treatment of advanced disease remains a challenge. Somatostatin analogues (SSAs) are considered standard therapy for carcinoid syndrome. SSAs delay tumor progression in advanced well-differentiated gastroenteropancreatic NETs. Cytotoxic chemotherapy and interferon play a limited role in the treatment of nonpancreatic GINETs. There is no standard approach to treatment of patients with disease progression. Identification of systemic agents with antitumor activity in advanced disease remains an unmet medical need. Enrollment to clinical trials is encouraged; potential therapeutic targets include the vascular endothelial growth factor and mammalian target of rapamycin signaling pathways.
Collapse
Affiliation(s)
- Claire K Mulvey
- Department of Medicine, University of California, San Francisco, 505 Parnassus Avenue, Box 0119, San Francisco, CA 94143, USA
| | - Emily K Bergsland
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, 1600 Divisadero Street, A727, San Francisco, CA 94115, USA.
| |
Collapse
|
29
|
Kwekkeboom DJ, Krenning EP. Peptide Receptor Radionuclide Therapy in the Treatment of Neuroendocrine Tumors. Hematol Oncol Clin North Am 2016; 30:179-91. [PMID: 26614376 DOI: 10.1016/j.hoc.2015.09.009] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Peptide receptor radionuclide therapy (PRRT) is a promising new treatment modality for inoperable or metastasized gastroenteropancreatic neuroendocrine tumors patients. Most studies report objective response rates in 15% to 35% of patients. Progression-free (PFS) and overall survival (OS) compare favorably with that for somatostatin analogues, chemotherapy, or newer, "targeted" therapies. Prospective, randomized data regarding the potential PFS and OS benefit of PRRT compared with standard therapies is anticipated.
Collapse
Affiliation(s)
- Dik J Kwekkeboom
- Department of Nuclear Medicine, Erasmus MC, University Medical Center, s-Gravendijkwal 230, Rotterdam 3015CE, The Netherlands.
| | - Eric P Krenning
- Department of Nuclear Medicine, Erasmus MC, University Medical Center, s-Gravendijkwal 230, Rotterdam 3015CE, The Netherlands
| |
Collapse
|
30
|
Strosberg JR. Systemic treatment of gastroenteropancreatic neuroendocrine tumors (GEP-NETS): current approaches and future options. Endocr Pract 2016; 20:167-75. [PMID: 24014009 DOI: 10.4158/ep13262.ra] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE To describe recent advances in the treatment of gastroenteropancreatic neuroendocrine tumors (GEP-NETs). METHODS A review of the published English language literature on GEP-NET therapy with a focus on practice-changing clinical trials. RESULTS Somatostatin analog (SSA) treatment remains a cornerstone of GEP-NET therapy, primarily for patients with hormonally functional tumors and midgut carcinoids. The biologic agents everolimus and sunitinib have similar tumor-stabilizing effects in pancreatic NETs and are both approved to treat progressive low-intermediate-grade tumors. Their role in nonpancreatic NETs remains controversial. Cytotoxic chemotherapy is effective against pancreatic NETs, but modern prospective data is lacking. Radiolabeled SSAs will likely become more widely available once phase III randomized studies are completed. CONCLUSIONS New treatment options for GEP-NETs have become available and highlight the necessity of developing predictive biomarkers that will allow for appropriate and individualized therapy selection.
Collapse
|
31
|
Mehrvarz Sarshekeh A, Halperin DM, Dasari A. Update on management of midgut neuroendocrine tumors. INTERNATIONAL JOURNAL OF ENDOCRINE ONCOLOGY 2016; 3:175-189. [PMID: 27347369 PMCID: PMC4915384 DOI: 10.2217/ije-2015-0004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Midgut neuroendocrine tumors are typically indolent but can be fatal when advanced. They can also cause significant morbidity due to the characteristic carcinoid syndrome. Somatostatin analogs continue to be the mainstay of treatment given their antiproliferative properties, as well as inhibitory effects on hormones that cause carcinoid syndrome. There have been several recent advances in the systemic therapy of these tumors including consolidation of somatostatin analogs as the cornerstone of therapy, completion of pivotal trials with mTOR inhibitors, and the establishment of novel approaches including peptide receptor radionuclide therapy and oral inhibitors of peripheral tryptophan hydroxylase in tumor and symptom control, respectively. In this review article, the recent advances are summarized and an updated approach to management is proposed.
Collapse
Affiliation(s)
- Amir Mehrvarz Sarshekeh
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Daniel M Halperin
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Arvind Dasari
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| |
Collapse
|
32
|
Ruszniewski P, Valle JW, Lombard-Bohas C, Cuthbertson DJ, Perros P, Holubec L, Delle Fave G, Smith D, Niccoli P, Maisonobe P, Atlan P, Caplin ME. Patient-reported outcomes with lanreotide Autogel/Depot for carcinoid syndrome: An international observational study. Dig Liver Dis 2016; 48:552-558. [PMID: 26917486 DOI: 10.1016/j.dld.2015.12.013] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 12/18/2015] [Accepted: 12/22/2015] [Indexed: 12/11/2022]
Abstract
BACKGROUND Lanreotide Autogel/Depot effectively controls symptoms in patients with carcinoid syndrome associated with neuroendocrine tumours. Data on patient-reported outcomes are sparse. AIM To evaluate the effect of lanreotide on patient-reported outcomes (PROs) with carcinoid syndrome. METHODS This was an international, open-label, observational study of adults with neuroendocrine tumours and history of diarrhoea, receiving lanreotide for >3 months for relief of carcinoid syndrome symptoms. The primary PRO measure was satisfaction with diarrhoea control. Secondary PRO measures included severity, change in symptoms and impact on daily life of diarrhoea; and patient satisfaction with flushing control. RESULTS Of 273 patients enrolled, 76% were 'completely' or 'rather' satisfied with diarrhoea control; 79% reported improvement in diarrhoea with lanreotide. The proportion of patients with 'mild', 'minimal', or 'no diarrhoea' increased from 33% before treatment to 75% during treatment; 75% were unconcerned about the impact of diarrhoea on daily life. Satisfaction with flushing control amongst patients with significant flushing at treatment initiation was 73%. CONCLUSIONS Lanreotide treatment was associated with improvements in symptoms as well as a range of PROs in patients with neuroendocrine tumours and carcinoid syndrome (ClinicalTrials.gov: NCT01234168).
Collapse
Affiliation(s)
| | - Juan W Valle
- Institute of Cancer Studies, University of Manchester/The Christie NHS Foundation Trust, Manchester, UK
| | | | - Daniel J Cuthbertson
- Department of Obesity and Endocrinology and Liverpool ENETS Centre of Excellence, University Hospital Aintree and University of Liverpool, Liverpool, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Massironi S, Conte D, Rossi RE. Somatostatin analogues in functioning gastroenteropancreatic neuroendocrine tumours: literature review, clinical recommendations and schedules. Scand J Gastroenterol 2015; 51:513-523. [PMID: 26605828 DOI: 10.3109/00365521.2015.1115117] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 10/27/2015] [Accepted: 10/27/2015] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Neuroendocrine tumours (NETs) represent a heterogeneous group of neoplasms, which include functioning and non-functioning forms. Somatostatin analogues (SSAs) play a key role in the management of these tumours. Herein, we aimed at reviewing the current evidence about the role of SSAs in the treatment of gastro-entero-pancreatic (GEP)-NETs. MATERIAL AND METHODS An extensive bibliographical search was performed in PubMed using the following keywords: gastro-entero-pancreatic neuroendocrine tumours, somatostatin analogues, octreotide, lanreotide, in order to identify all the pertinent English-written articles published between 1990 and 2015. RESULTS SSAs have shown to help the symptomatic and biochemical improvement of patients with NETs and to exhibit a good safety profile. Recent studies have also reported a role for SSAs in tumour growth control, although the results are less impressive and the underlying mechanisms are not fully understood. CONCLUSIONS SSAs are well known as a symptomatic and, to lesser extent, anti-proliferative treatment in GEP-NETs. However, some issues, including optimal dosage, benefits and adverse events of combination with other molecules, and the role of new analogues, remain to be elucidated in further randomised studies.
Collapse
Affiliation(s)
- Sara Massironi
- a Gastroenterology and Endoscopy Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico , Milan , Italy
| | - Dario Conte
- a Gastroenterology and Endoscopy Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico , Milan , Italy
- b Department of Pathophysiology and Transplantation , Università Degli Studi Di Milano , Milan , Italy
| | - Roberta Elisa Rossi
- a Gastroenterology and Endoscopy Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico , Milan , Italy
- b Department of Pathophysiology and Transplantation , Università Degli Studi Di Milano , Milan , Italy
| |
Collapse
|
34
|
Al-Efraij K, Aljama MA, Kennecke HF. Association of dose escalation of octreotide long-acting release on clinical symptoms and tumor markers and response among patients with neuroendocrine tumors. Cancer Med 2015; 4:864-70. [PMID: 25727756 PMCID: PMC4472209 DOI: 10.1002/cam4.435] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 01/21/2015] [Accepted: 01/23/2015] [Indexed: 12/13/2022] Open
Abstract
Patients with nonresectable metastatic neuroendocrine tumors (NETs) experience symptoms of hormone hypersecretion including diarrhea, flushing, and bronchoconstriction, which can interfere with quality of life [Anthony and Vinik (2011) Pancreas, 40:987]. Treatment with a long-acting release formulation of octreotide, a somatostatin analog, can help to alleviate these symptoms. Although high doses of octreotide are often required for adequate symptom control, the relationship between octreotide dose escalation and symptom control in the NET context is not well quantified in the literature. A retrospective chart review was conducted of nonresectable metastatic NET patients who received a dose greater than 30 mg intramuscular octreotide long-acting formulation (O-LAR) at any time between January 2005 and December 2011 at the British Columbia Cancer Agency (BCCA). The association between dose escalation of O-LAR, chromogranin A (CGA), 24-h urine 5-hydoxyindoacetate (5-HIAA), symptom control, and radiological progression was explored. Dose escalation of O-LAR was associated with improved symptom control in NET patients who were refractory to the standard dose levels. Reduction of serum CGA & 5-HIAA levels by at least 10% was observed in 31% and 23% respectively. Retrospective review of imaging did not document any reductions in tumor volume. Higher doses of O-LAR are associated with improved symptom control in NET patients. The variability in tumor marker levels in response to O-LAR dose escalation may indicate that tumor marker levels may not be an accurate assessment of therapeutic efficacy.
Collapse
Affiliation(s)
- Khalid Al-Efraij
- Division of Respirology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Mohammed A Aljama
- Division of Hematology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Hagen Fritz Kennecke
- Division of Medical Oncology, British Columbia Cancer Agency, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
35
|
Kamp K, Feelders RA, van Adrichem RCS, de Rijke YB, van Nederveen FH, Kwekkeboom DJ, de Herder WW. Parathyroid hormone-related peptide (PTHrP) secretion by gastroenteropancreatic neuroendocrine tumors (GEP-NETs): clinical features, diagnosis, management, and follow-up. J Clin Endocrinol Metab 2014; 99:3060-9. [PMID: 24905065 DOI: 10.1210/jc.2014-1315] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
CONTEXT Only a small number of case reports has been published on patients with PTHrP-hypersecreting metastatic gastroenteropancreatic (GEP) neuroendocrine tumors (NETs). OBJECTIVE The objective of this study was to evaluate the clinical, biochemical, and radiological features, management, and treatment outcome of patients with PTHrP-hypersecreting GEP-NETs. DESIGN Retrospective case series. SETTING Tertiary referral hospital. MAIN OUTCOME MEASURES Clinical, biochemical, and radiological features were measured, as well as response to therapy and survival. PATIENTS Ten patients with PTHrP-secreting GEP-NETs (nine pancreatic and one unknown primary) with a median age of 50.4 years (range, 38.3-61.1) were studied. Multiple endocrine neoplasia type 1 patients were excluded. RESULTS The median follow-up was 57.2 months (range, 11.6-204.5 mo). Median overall survival was 86.0 months. In total, 51 different treatment interventions and combinations were applied. In seven of the 10 patients, somatostatin analog (SSA) treatment resulted in a temporary normalization of serum calcium levels with a long-term response observed in two patients (up to 35.2 mo). Peptide receptor radiotherapy (PRRT) with radiolabeled SSAs induced long-term responses ranging from 9.0-49.0 months in four of six patients treated with PRRT. CONCLUSIONS Hypersecretion of PTHrP by metastatic GEP-NETs is very rare and seems to be exclusively associated with metastatic pancreatic NETs. PTHrP production has major clinical impact because poorly controllable hypercalcemia is associated with increased morbidity and mortality. The most successful treatment options for PTHrP-producing GEP-NETs are SSAs and PRRT using radiolabeled SSAs. Isotonic saline and bisphosphonates can be considered as supportive therapies.
Collapse
Affiliation(s)
- Kimberly Kamp
- Department of Internal Medicine, Sector of Endocrinology (K.K., R.A.F., R.C.S.v.A., W.W.d.H.), Department of Clinical Chemistry (Y.B.d.R.), Department of Pathology (F.H.v.N.), and Department of Nuclear Medicine (D.J.K.), ENETS Centre of Excellence, Erasmus Medical Center, 3015 CE Rotterdam, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
36
|
Martín-Richard M, Massutí B, Pineda E, Alonso V, Marmol M, Castellano D, Fonseca E, Galán A, Llanos M, Sala MA, Pericay C, Rivera F, Sastre J, Segura A, Quindós M, Maisonobe P. Antiproliferative effects of lanreotide autogel in patients with progressive, well-differentiated neuroendocrine tumours: a Spanish, multicentre, open-label, single arm phase II study. BMC Cancer 2013; 13:427. [PMID: 24053191 PMCID: PMC3853091 DOI: 10.1186/1471-2407-13-427] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Accepted: 09/11/2013] [Indexed: 01/17/2023] Open
Abstract
Background Somatostatin analogues (SSAs) are indicated to relieve carcinoid syndrome but seem to have antiproliferative effects on neuroendocrine tumours (NETs). This is the first prospective study investigating tumour stabilisation with the long-acting SSA lanreotide Autogel in patients with progressive NETs. Methods This was a multicentre, open-label, phase II trial conducted in 17 Spanish specialist centres. Patients with well-differentiated NETs and radiologically confirmed progression within the previous 6 months received lanreotide Autogel, 120 mg every 28 days over ≤92 weeks. The primary endpoint was progression-free survival (PFS). Secondary endpoints were response rate, tumour biomarkers, symptom control, quality of life (QoL), and safety. Radiographic imaging was assessed by a blinded central radiologist. Results Of 30 patients included in the efficacy and safety analyses, 40% had midgut tumours and 27% pancreatic tumours; 63% of tumours were functioning. Median PFS time was 12.9 (95% CI: 7.9, 16.5) months, and most patients achieved disease stabilisation (89%) or partial response (4%). No deterioration in QoL was observed. Nineteen patients (63%) experienced treatment-related adverse events, most frequently diarrhoea and asthenia; only one treatment-related adverse event (aerophagia) was severe. Conclusion Lanreotide Autogel provided effective tumour stabilisation and PFS >12 months in patients with progressive NETs ineligible for surgery or chemotherapy, with a safety profile consistent with the pharmacology of the class. Trial registration ClinicalTrials.gov Identifier NCT00326469; EU Clinical Trial Register EudraCT no 2004-002871-18.
Collapse
Affiliation(s)
- Marta Martín-Richard
- Medical Oncology Department, Hospital de la Santa Creu y Sant Pau, Av, Sant Antoni Mª Claret, 167, Barcelona 08025, Spain.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Theodoropoulou M, Stalla GK. Somatostatin receptors: from signaling to clinical practice. Front Neuroendocrinol 2013; 34:228-52. [PMID: 23872332 DOI: 10.1016/j.yfrne.2013.07.005] [Citation(s) in RCA: 280] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 06/13/2013] [Accepted: 07/12/2013] [Indexed: 02/08/2023]
Abstract
Somatostatin is a peptide with a potent and broad antisecretory action, which makes it an invaluable drug target for the pharmacological management of pituitary adenomas and neuroendocrine tumors. Somatostatin receptors (SSTR1, 2A and B, 3, 4 and 5) belong to the G protein coupled receptor family and have a wide expression pattern in both normal tissues and solid tumors. Investigating the function of each SSTR in several tumor types has provided a wealth of information about the common but also distinct signaling cascades that suppress tumor cell proliferation, survival and angiogenesis. This provided the rationale for developing multireceptor-targeted somatostatin analogs and combination therapies with signaling-targeted agents such as inhibitors of the mammalian (or mechanistic) target of rapamycin (mTOR). The ability of SSTR to internalize and the development of rabiolabeled somatostatin analogs have improved the diagnosis and treatment of neuroendocrine tumors.
Collapse
Affiliation(s)
- Marily Theodoropoulou
- Department of Endocrinology, Max Planck Institute of Psychiatry, Kraepelinstrasse 10, 80804 Munich, Germany.
| | | |
Collapse
|
38
|
|
39
|
Keskin O, Yalcin S. A review of the use of somatostatin analogs in oncology. Onco Targets Ther 2013; 6:471-83. [PMID: 23667314 PMCID: PMC3650572 DOI: 10.2147/ott.s39987] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Indexed: 12/12/2022] Open
Abstract
Somatostatin is a neuropeptide produced by paracrine cells that are located throughout the gastrointestinal tract, lung, and pancreas, and is also found in various locations of the nervous system. It exerts neural control over many physiological functions including inhibition of gastrointestinal endocrine secretion through its receptors. Potent and biologically stable analogs of somatostatin have been developed. These somatostatin analogs show different efficacy on different receptors, and receptors are varyingly concentrated in specific tissues. Antitumor and antisecretory effects of somatostatin analogs in cancer have been shown in several in vivo and in vitro studies. However, these activities have not always yielded into clinically relevant patient outcome benefit. Somatostatin analogs are of clinical benefit in treating symptoms of ectopic hormone secretion (adrenocorticotropic hormone, growth hormone-releasing hormone) in lung cancer, without inducing a significant tumor response. They have also been shown to induce a statistically significant decrease in bone pain and increase in Karnofsky performance status in patients with metastatic prostate cancer. Somatostatin analogs alone or in combination with other agents have only limited antitumoral effect in breast cancer. In gastrointestinal cancers, studies have not shown an objective tumor response to somatostatin analogs except in endocrine tumors of the liver with symptomatic and biochemical improvement. In neuroendocrine tumors of the gastrointestinal system and pancreas, very high symptomatic and biochemical response rates have been achieved with somatostatin analogs. Antiproliferative activity has been clearly shown in metastatic midgut neuroendocrine tumors.
Collapse
Affiliation(s)
- Ozge Keskin
- Department of Medical Oncology, Hacettepe University Institute of Cancer, Ankara, Turkey
| | - Suayib Yalcin
- Department of Medical Oncology, Hacettepe University Institute of Cancer, Ankara, Turkey
| |
Collapse
|
40
|
Toumpanakis C, Caplin ME. Update on the role of somatostatin analogs for the treatment of patients with gastroenteropancreatic neuroendocrine tumors. Semin Oncol 2013; 40:56-68. [PMID: 23391113 DOI: 10.1053/j.seminoncol.2012.11.006] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Somatostatin analogs (SA) are the standard of care for controlling symptoms of patients with functional gastro-entero-pancreatic neuroendocrine tumors (GEP-NETs). SA control symptoms in more than 70% of patients with carcinoid syndrome. Similar results are obtained in patients with functional, hormone-secreting, pancreatic NETs. The use of SA as antiproliferative agents has been established only recently. Retrospective studies have shown stabilization of tumor growth in >50% of patients with progressive disease. The results of a recent randomized phase III trial (PROMID) demonstrated that the median time to progression in patients with midgut carcinoid tumors treated with octreotide LAR (Long-Acting-Repeatable, Novartis, Basel, Switzerland) was more than twice as long compared to that of patients treated with placebo. The results of a phase III study of lanreotide versus placebo in nonfunctional NETs are not yet available. More studies are needed to determine whether combining SA with novel targeted treatments will result in enhanced antiproliferative activity compared to treatment with a SA alone. Studies are ongoing using pan-receptor agonists (eg, pasireotide) and chimeric dimers, which possess features of somatostatin and dopamine agonists (dopastatins) and are thought to enhance symptom control by binding multiple receptors (somatostatin and dopamine receptors). Somatostatin receptor antagonists are also currently being developed for clinical use. Peptide receptor radionuclide therapy (PRRT), consisting of yttrium-90 and lutetium-177 isotopes conjugated with SA appear to be efficacious in advanced NETs. Randomized studies are needed to definitively establish the safety and efficacy of this strategy compared to other available treatments, and to determine which radiolabeled isotopes or combinations are most effective.
Collapse
Affiliation(s)
- Christos Toumpanakis
- Neuroendocrine Tumour Unit, ENETS Centre of Excellence, Royal Free Hospital, London, UK
| | | |
Collapse
|
41
|
|
42
|
Raju N, Pai R, Welton ML. Miscellaneous Neoplasms. COLORECTAL SURGERY 2013:326-336. [DOI: 10.1016/b978-1-4377-1724-2.00020-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
43
|
Strosberg J. Neuroendocrine tumours of the small intestine. Best Pract Res Clin Gastroenterol 2012; 26:755-73. [PMID: 23582917 DOI: 10.1016/j.bpg.2012.12.002] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Accepted: 12/27/2012] [Indexed: 01/31/2023]
Abstract
The prevalence of intestinal neuroendocrine tumours, also known as carcinoid tumours, has increased significantly over the past three decades. Tumours of the distal small intestine (midgut) are often indolent, but are characterized by a high potential to metastasize to the small-bowel mesentery and liver. Patients with distant metastases are prone to development of the carcinoid syndrome, a constellation of symptoms which includes flushing, diarrhoea, and valvular heart disease. The carcinoid syndrome is caused by secretion of serotonin and other vasoactive substances into the systemic circulation. Treatment options for metastatic intestinal NETs have expanded in recent years. Of particular importance has been the development of somatostatin-analogue therapies. Somatostatin analogues were originally introduced for palliation of the carcinoid syndrome; however recent clinical trials have demonstrated that they can exert an inhibitory effect on tumour growth. Other novel agents targeting the VEGF and mTOR pathways have recently been evaluated in phase III trials, however their role in the management of small-intestinal NETs remains controversial. This article examines the biological characteristics of small intestinal NETs, summarizes current guidelines on classification, staging and grading, and reviews developments in locoregional and systemic therapy.
Collapse
Affiliation(s)
- Jonathan Strosberg
- H. Lee Moffitt Cancer Center and Research Institute, Dept. of GI Oncology, 12902 Magnolia Dr., Tampa, FL 33612, USA.
| |
Collapse
|
44
|
Mizutani G, Nakanishi Y, Watanabe N, Honma T, Obana Y, Seki T, Ohni S, Nemoto N. Expression of Somatostatin Receptor (SSTR) Subtypes (SSTR-1, 2A, 3, 4 and 5) in Neuroendocrine Tumors Using Real-time RT-PCR Method and Immunohistochemistry. Acta Histochem Cytochem 2012; 45:167-76. [PMID: 22829710 PMCID: PMC3395302 DOI: 10.1267/ahc.12006] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Accepted: 02/21/2012] [Indexed: 01/21/2023] Open
Abstract
Molecule targeting therapy using somatostatin (SS) analogues has become a widely accepted modality to treat neuroendocrine tumors (NETs), particularly gastrointestinal (GI) and pancreatic endocrine tumors. On the other hand, little is known about the expression of somatostatin receptor (SSTR) subtypes in neuroendocrine carcinomas (NECs). We investigated the expression of SSTR subtypes (SSTR-1, 2A, 3, 4 and 5) using real-time reverse transcription polymerase chain reaction (RT-PCR) method and immunohistochemistry in 32 neuroendocrine neoplasms (9 NET G1, 2 NET G2, 18 NECs G3 and 3 mixed NEC G3) of various primary sites. Expression of more than two SSTR subtypes was detected in all neuroendocrine neoplasms examined. Expression of SSTR-2A mRNA was significantly higher than other subtypes. In addition, mRNA expression of SSTR-3 and SSTR-5 was significantly low or below the detection level except for gastroduodenal NET G1. No significant difference of the expression of SSTR subtypes was observed between the NET and NEC groups. The expression of protein and mRNA was generally well correlated. In conclusion, NECs would be a good candidate for molecule targeting therapy using SS analogues, and the expression of SSTR-2A can be useful as a biomarker of neuroendocrine differentiation. We have demonstrated that NEC G3 small cell type shows a different expression profile of SSTR subtypes compared with NET and NEC non-small cell type.
Collapse
Affiliation(s)
- Gou Mizutani
- Department of Pathology, Nihon University School of Medicine
- Department of Pathology, Nihon University School of Medicine
| | - Yoko Nakanishi
- Department of Pathology, Nihon University School of Medicine
- Department of Pathology, Nihon University School of Medicine
| | - Noriko Watanabe
- Department of Pathology, Nihon University School of Medicine
- Pathology Division, Nihon University Itabashi Hospital
- Department of Pathology, Nihon University School of Medicine
- Pathology Division, Nihon University Itabashi Hospital
| | - Taku Honma
- Department of Pathology, Nihon University School of Medicine
- Pathology Division, Nihon University Itabashi Hospital
- Department of Pathology, Nihon University School of Medicine
- Pathology Division, Nihon University Itabashi Hospital
| | - Yukari Obana
- Department of Pathology, Nihon University School of Medicine
- Department of Pathology, Nihon University School of Medicine
| | - Toshimi Seki
- Pathology Division, Nihon University Itabashi Hospital
- Pathology Division, Nihon University Itabashi Hospital
| | - Sumie Ohni
- Department of Pathology, Nihon University School of Medicine
- Pathology Division, Nihon University Itabashi Hospital
- Department of Pathology, Nihon University School of Medicine
- Pathology Division, Nihon University Itabashi Hospital
| | - Norimichi Nemoto
- Department of Pathology, Nihon University School of Medicine
- Pathology Division, Nihon University Itabashi Hospital
- Department of Pathology, Nihon University School of Medicine
- Pathology Division, Nihon University Itabashi Hospital
| |
Collapse
|
45
|
Karpathakis A, Caplin M, Thirlwell C. Hitting the target: where do molecularly targeted therapies fit in the treatment scheduling of neuroendocrine tumours? Endocr Relat Cancer 2012; 19:R73-92. [PMID: 22474226 DOI: 10.1530/erc-12-0050] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Neuroendocrine tumours (NETs) are a rare and heterogeneous group of tumours whose incidence is increasing and their prevalence is now greater than that of any other upper gastrointestinal tumour. Diagnosis can be challenging, and up to 25% of patients present with metastatic disease. Following the recent FDA approval of two new molecularly targeted therapies for the treatment of advanced pancreatic NETs (pNETs), the first in 25 years, we review all systemic therapies and suggest where these newer targeted therapies fit in the treatment schedule for these challenging tumours. Clinical trial data relating to the routine use of sunitinib and everolimus in low-intermediate-grade pNETs are summarised alongside newer molecularly targeted agents undergoing clinical assessment in NETs. We particularly focus on the challenge of optimal scheduling of molecularly targeted treatments around existing systemic and localised treatment such as chemotherapy or radiotargeted therapy. We also discuss application of current evidence to subgroups of patients who have not so far been directly addressed such as those with poorer performance status or patients receiving radical surgery who may benefit from adjuvant treatment.
Collapse
Affiliation(s)
- Anna Karpathakis
- University College London Cancer Institute, Paul O'Gorman Building, Huntley Street, London WC1E 6BT, UK
| | | | | |
Collapse
|
46
|
Ramage JK, Ahmed A, Ardill J, Bax N, Breen DJ, Caplin ME, Corrie P, Davar J, Davies AH, Lewington V, Meyer T, Newell-Price J, Poston G, Reed N, Rockall A, Steward W, Thakker RV, Toubanakis C, Valle J, Verbeke C, Grossman AB. Guidelines for the management of gastroenteropancreatic neuroendocrine (including carcinoid) tumours (NETs). Gut 2012; 61:6-32. [PMID: 22052063 PMCID: PMC3280861 DOI: 10.1136/gutjnl-2011-300831] [Citation(s) in RCA: 388] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
These guidelines update previous guidance published in 2005. They have been revised by a group who are members of the UK and Ireland Neuroendocrine Tumour Society with endorsement from the clinical committees of the British Society of Gastroenterology, the Society for Endocrinology, the Association of Surgeons of Great Britain and Ireland (and its Surgical Specialty Associations), the British Society of Gastrointestinal and Abdominal Radiology and others. The authorship represents leaders of the various groups in the UK and Ireland Neuroendocrine Tumour Society, but a large amount of work has been carried out by other specialists, many of whom attended a guidelines conference in May 2009. We have attempted to represent this work in the acknowledgements section. Over the past few years, there have been advances in the management of neuroendocrine tumours, which have included clearer characterisation, more specific and therapeutically relevant diagnosis, and improved treatments. However, there remain few randomised trials in the field and the disease is uncommon, hence all evidence must be considered weak in comparison with other more common cancers.
Collapse
Affiliation(s)
- John K Ramage
- Basingstoke and North Hampshire Hospital, Aldermaston Road, Basingstoke RG24 9NA, UK.
| | - A Ahmed
- Department of Gastroenterology, County Durham and Darlington Foundation Trust, Darlington, UK
| | - J Ardill
- Peptide Laboratory, Royal Victoria Hospital, Belfast, UK
| | - N Bax
- Department of Clinical Pharmacology, University of Sheffield, Sheffield, UK
| | - D J Breen
- Department of Radiology, Southampton General Hospital, Southampton, UK
| | - M E Caplin
- Department of Gastroenterology, University College London, London, UK
| | - P Corrie
- Department of Oncology, Addenbrookes Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - J Davar
- Department of Cardiology, Royal Free Hospital, London, UK
| | - A H Davies
- Department of Gastroenterology, University Hospitals of Morecambe Bay Foundation Trust, Barrow-in-Furness, UK
| | - V Lewington
- Nuclear Medicine, Kings College London, London, UK
| | - T Meyer
- Department of Oncology, University College London, London, UK
| | - J Newell-Price
- Department of Endocrinology, University of Sheffield, Sheffield, UK
| | - G Poston
- Department of Surgery, University of Liverpool, Liverpool, UK
| | - N Reed
- Department of Oncology, Beatson Centre, Glasgow, UK
| | - A Rockall
- Department of Radiology, Queen Mary's University, London, UK
| | - W Steward
- Department of Oncology, University of Leicester, Leicester, UK
| | - R V Thakker
- Academic Department of Endocrinology, Diabetes and Metabolism, Oxford University, Oxford, UK
| | - C Toubanakis
- Department of Gastroenterology, Royal Free Hospital, London, UK
| | - J Valle
- Department of Medical Oncology, The Christie Hospital NHS Foundation Trust, Manchester, UK
| | - C Verbeke
- Department of Histopathology, St James's University Hospital, Leeds, UK
| | - A B Grossman
- Academic Department of Endocrinology, Diabetes and Metabolism, Oxford University, Oxford, UK
| | | |
Collapse
|
47
|
Khan MS, Caplin ME. Therapeutic management of patients with gastroenteropancreatic neuroendocrine tumours. Endocr Relat Cancer 2011; 18 Suppl 1:S53-74. [PMID: 22005115 DOI: 10.1530/erc-10-0271] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Patients with neuroendocrine tumours (NETs) are best managed in a specialist centre as part of a multidisciplinary team comprising gastroenterologists, oncologists, endocrinologists, gastrointestinal and hepatopancreaticobiliary surgeons, pathologists, nuclear medicine physicians and technicians, radiologists, specialist nurses, pharmacists, biochemists and dieticians. This should ideally be led by a clinician with experience and interest in NETs. Although the number of medical treatments and clinical trials has increased in the decade, there is still a lack of prospective randomised trials; thus, management is mainly based on limited often single-centre studies, although there are now formal guidelines based on consensus expert opinion. We have outlined the current optimal management of patients with NETs. We have reviewed therapeutic options including surgery, somatostatin analogues and other biotherapies and peptide receptor-targeted therapy. We have discussed the challenge in managing hepatic metastases including hepatic artery embolisation, ablation and orthotopic liver transplant. In addition, we have briefly reviewed the emerging therapies such as the mammalian target of rapamycin and angiogenic inhibitors and the newer somatostatin analogues.
Collapse
Affiliation(s)
- Mohid S Khan
- Neuroendocrine Tumour Unit, Centre for Gastroenterology, Royal Free Hospital, London NW3 2QG, UK
| | | |
Collapse
|
48
|
|
49
|
Khan MS, El-Khouly F, Davies P, Toumpanakis C, Caplin ME. Long-term results of treatment of malignant carcinoid syndrome with prolonged release Lanreotide (Somatuline Autogel). Aliment Pharmacol Ther 2011; 34:235-42. [PMID: 21585408 DOI: 10.1111/j.1365-2036.2011.04693.x] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Somatostatin analogues are the mainstay of therapy for malignant carcinoid syndrome. There is clear evidence that the once monthly intramuscular formulation, Octreotide LAR, controls symptoms of carcinoid syndrome, and recent data also suggests an antitumour effect. There is limited data on prolonged release Lanreotide (Somatuline Autogel, Ipsen Pharma Biotech, Signes, France) and no long-term data to date. AIM To present long-term results of prolonged release Lanreotide in a large cohort of patients with malignant carcinoid syndrome, assessing clinical and objective response and tolerance. METHODS Seventy six patients with metastatic midgut neuroendocrine tumours and carcinoid syndrome were included in this 9-year retrospective study. Clinical response was based on symptom score with radiological assessment based on RECIST (Response Evaluation Criteria In Solid Tumours). RESULTS Data were available in 69 patients. Ninety four percent achieved symptomatic response at first follow-up visit. Forty six percent had loss of symptomatic response, but 44% of these achieved control with an increase in dose of prolonged release Lanreotide. Overall, symptoms were well controlled throughout the study period with prolonged release Lanreotide alone in 74% of patients. Twenty six percent required additional treatment despite good initial response. Only 30% demonstrated radiological progression. Eleven patients who were switched from Octreotide LAR had return of symptomatic control. No significant adverse effects were experienced. CONCLUSIONS Prolonged release Lanreotide provides good symptomatic control of diarrhoea and flushing as well as tumour stability in patients with malignant carcinoid syndrome.
Collapse
Affiliation(s)
- M S Khan
- Neuroendocrine Tumour Unit, Centre for Gastroenterology, Royal Free Hospital, London, UK
| | | | | | | | | |
Collapse
|
50
|
Strosberg JR, Cheema A, Kvols LK. A Review of Systemic and Liver-Directed Therapies for Metastatic Neuroendocrine Tumors of the Gastroenteropancreatic Tract. Cancer Control 2011; 18:127-137. [DOI: 10.1177/107327481101800207] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
Affiliation(s)
- Jonathan R. Strosberg
- Department of Gastrointestinal Oncology at the H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Asima Cheema
- Department of Gastrointestinal Oncology at the H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Larry K. Kvols
- Department of Gastrointestinal Oncology at the H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| |
Collapse
|