1
|
Melhorn P, Mazal P, Wolff L, Kretschmer-Chott E, Raderer M, Kiesewetter B. From biology to clinical practice: antiproliferative effects of somatostatin analogs in neuroendocrine neoplasms. Ther Adv Med Oncol 2024; 16:17588359241240316. [PMID: 38529270 PMCID: PMC10962050 DOI: 10.1177/17588359241240316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 02/27/2024] [Indexed: 03/27/2024] Open
Abstract
Somatostatin analogs (SSA), specifically octreotide and lanreotide, have demonstrated antiproliferative effects in patients with neuroendocrine tumors (NET), a group of rare malignancies of diverse origin and presentation. A prominent feature of NET cells is the expression of G protein-coupled receptors called somatostatin receptors (SSTR). Although these SSTR are not uniformly present in NET, they can be instrumental in the diagnosis and treatment of NET. Apart from their application in nuclear imaging and radionuclide therapy, SSA have proven invaluable in the treatment of hormonal syndromes associated with certain NET (antisecretory effects of SSA), but it took more than two decades to convincingly demonstrate the antiproliferative effects of SSA in metastatic NET with the two pivotal studies PROMID and CLARINET. The current review summarizes three decades of SSA treatment and provides an overview of the clinical trial landscape for SSA monotherapy and combination therapy, including clinical implications and quality of life aspects, as well as ongoing fields of research.
Collapse
Affiliation(s)
- Philipp Melhorn
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Peter Mazal
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Ladislaia Wolff
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Elisabeth Kretschmer-Chott
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Markus Raderer
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Waehringer Guertel 18-20, Vienna A-1090, Austria
| | - Barbara Kiesewetter
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
2
|
Una Cidon E. Vasoactive intestinal peptide secreting tumour: An overview. World J Gastrointest Oncol 2022; 14:808-819. [PMID: 35582098 PMCID: PMC9048535 DOI: 10.4251/wjgo.v14.i4.808] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/15/2021] [Accepted: 03/27/2022] [Indexed: 02/06/2023] Open
Abstract
Vasoactive intestinal peptide (VIP) secreting tumour (VIPoma) is a rare functional neuroendocrine tumour that typically arises from pancreatic islet cells. These present as sporadic, solitary pancreatic neoplasias with an estimated incidence of one in ten million individuals per year. Only around 5% of VIPomas are associated with multiple endocrine neoplasia type I syndrome. Excessive VIP secretion produces a clinical syndrome characterized by refractory watery diarrhoea, hypokalemia and metabolic acidosis. These coupled with elevated plasma levels of VIP are diagnostic. The majority of VIPomas are malignant and have already metastasized at the time of diagnosis (60%). Metastases occur most frequently in the liver, or regional lymph nodes, lungs, kidneys and bones. Some reports of skin metastases have been documented. Complete surgical resection continues to be the only potentially curative treatment. However, when the neoplasia cannot be excised completely, surgical debulking may provide palliative benefit. Other palliative options have included recently the peptide receptor radionuclide therapy which has shown to be effective and well-tolerated. This article will review all aspects of pancreatic VIPomas highlighting aspects such as clinical presentation, diagnosis and management.
Collapse
Affiliation(s)
- Esther Una Cidon
- Department of Medical Oncology, University Hospitals Dorset, Bournemouth BH7 7DW, Dorset, United Kingdom
| |
Collapse
|
3
|
Caplin ME, Pavel M, Phan AT, Ćwikła JB, Sedláčková E, Thanh XMT, Wolin EM, Ruszniewski P. Lanreotide autogel/depot in advanced enteropancreatic neuroendocrine tumours: final results of the CLARINET open-label extension study. Endocrine 2021; 71:502-513. [PMID: 33052555 PMCID: PMC7881960 DOI: 10.1007/s12020-020-02475-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 08/23/2020] [Indexed: 12/18/2022]
Abstract
PURPOSE In the phase III CLARINET study (NCT00353496), lanreotide autogel/depot (lanreotide) significantly improved progression-free survival (PFS) vs placebo in patients with non-functioning intestinal or pancreatic neuroendocrine tumours (NETs). The aim of CLARINET open-label extension (OLE) (NCT00842348) was to evaluate long-term safety and efficacy of lanreotide in these patients. METHODS Patients from the CLARINET study were eligible for the OLE if they had stable disease (irrespective of treatment group) or progressive disease (PD) (placebo-treated patients only). All patients in the OLE received lanreotide 120 mg every 28 days. Computed tomography or magnetic resonance imaging scans were conducted every 6 months and assessed locally for PD (the final scan was also assessed centrally). RESULTS Overall, 89 patients took part in the OLE (lanreotide, n = 42; placebo, n = 47). Median (range) exposure to lanreotide in patients who received lanreotide in the core study and OLE (LAN-LAN group) was 59.0 (26.0-102.3) months. In this group, the overall incidences of adverse events (AEs) and treatment-related AEs were lower in the OLE than in the core study. Median [95% CI] PFS in the LAN-LAN group was 38.5 [30.9; 59.4] months. In placebo-treated patients with PD at the end of the core study, time to death or subsequent PD during the OLE was 19 [10.1; 26.7] months. CONCLUSIONS This study provides new evidence on the long-term safety profile and sustained anti-tumour effects of lanreotide autogel/depot in indolent and progressive metastatic intestinal or pancreatic NETs.
Collapse
Affiliation(s)
- Martyn E Caplin
- Department of Gastroenterology and Tumour Neuroendocrinology, Royal Free Hospital, London, UK.
| | - Marianne Pavel
- Department of Medicine, Division of Endocrinology and Diabetology, Universitätsklinikum Erlangen, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Alexandria T Phan
- Department of Hematology-Oncology, University of Texas Health Science Center at Tyler, Tyler, TX, USA
- Cancer Treatment Centers of America at South Eastern Regional Center, Atlanta, GA, USA
| | - Jarosław B Ćwikła
- Department of Cardiology and Cardiac Surgery, School of Medicine, University of Warmia and Mazury, Olsztyn, Poland
- Diagnostic and Therapeutic Center - Gammed, Warsaw, Poland
| | - Eva Sedláčková
- Department of Oncology, First Faculty of Medicine and General Teaching Hospital, Prague, Czech Republic
| | | | - Edward M Wolin
- Tisch Cancer Institute at Mount Sinai and Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Carcinoid and Neuroendocrine Tumors, New York, NY, USA
| | - Philippe Ruszniewski
- Division of Gastroenterology and Pancreatology, Beaujon Hospital, Clichy, France
- Université de Paris, Paris, France
| |
Collapse
|
4
|
Abstract
INTRODUCTION Knowledge of the molecular subtypes of bladder cancer enables powerful generalizations involving the distinctive biology and pathways driving different disease subsets. Areas covered: In this review, we summarize the findings of a number of published studies exploring the molecular landscape of bladder cancer by analysis of genomic data from The Cancer Genome Atlas (TCGA). TCGA project has provided a comprehensive data resource of 412 muscle-invasive bladder cancers as characterized by multiple molecular analytical platforms. These data have been and will continue to be utilized in numerous subsequent studies aimed at better understanding the molecular basis of bladder cancer. The catalog of DNA-level alterations can greatly inform personalized and precision medicine approaches. Molecular subtypes of bladder cancer include distinct 'basal/squamous' and 'luminal' subtypes, cancers with papillary histology, disease subsets with prominent leukocyte infiltration and immune checkpoint marker expression, and a 'neuronal' subtype lacking small cell or neuroendocrine histology. The gene-level alterations and subtypes as revealed by TCGA data are relevant from the standpoint of both basic biology and clinical trial studies. Expert commentary: Multiple studies analyzing TCGA muscle-invasive bladder cancer cases point to the existence of five major expression-based molecular subtypes of the disease, with these subtypes having therapeutic implications.
Collapse
Affiliation(s)
- Chad J. Creighton
- Dan L. Duncan Comprehensive Cancer Center Division of Biostatistics, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
5
|
Barrows SM, Cai B, Copley-Merriman C, Wright KR, Castro CV, Soufi-Mahjoubi R. Systematic literature review of the antitumor effect of octreotide in neuroendocrine tumors. World J Meta-Anal 2018; 6:9-20. [DOI: 10.13105/wjma.v6.i2.9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 04/26/2018] [Accepted: 05/15/2018] [Indexed: 02/06/2023] Open
Abstract
AIM To provide a comprehensive examination of the existing evidence of the antitumor effect of long-acting octreotide in neuroendocrine tumors (NETs).
METHODS A systematic literature review of clinical trials and observational studies was conducted in PubMed, EMBASE, and Cochrane through January 18, 2017. Conference abstracts for 2015 and 2016 from 5 scientific meetings were also searched.
RESULTS Of 41 articles/abstracts identified, 13 unique studies compared octreotide with active or no treatment. Two of the 13 studies were clinical trials; the remaining were observational studies. The phase 3 Placebo-Controlled, Double-Blind, Prospective, Randomized Study of the Effect of Octreotide long-acting repeatable (LAR) in the Control of Tumor Growth in Patients with Metastatic Neuroendocrine Midgut Tumors clinical trial showed that long-acting octreotide significantly prolonged time to tumor progression compared with placebo in patients with functionally active and inactive metastatic midgut NETs; no statistically significant difference in overall survival (OS) was observed, possibly due to the crossover of placebo patients to octreotide. Retrospective observational studies found that long-acting octreotide use was associated with significantly longer OS than no octreotide use for patients with distant metastases although not for those with local/regional disease.
CONCLUSION The clinical trial and observational studies with informative evidence support long-acting octreotide’s antitumor effect on time to tumor progression and OS. This review showed the rarity of existing studies assessing octreotide’s antitumor effect and recommends that future research is warranted.
Collapse
Affiliation(s)
| | - Beilei Cai
- Novartis Pharmaceuticals, East Hanover, NJ 07936, United States
| | | | - Kelly R Wright
- RTI Health Solutions, Ann Arbor, MI 48108, United States
| | - Colleen V Castro
- RTI Health Solutions, Research Triangle Park, NC 27709, United States
| | | |
Collapse
|
6
|
Lee L, Ito T, Jensen RT. Everolimus in the treatment of neuroendocrine tumors: efficacy, side-effects, resistance, and factors affecting its place in the treatment sequence. Expert Opin Pharmacother 2018; 19:909-928. [PMID: 29757017 PMCID: PMC6064188 DOI: 10.1080/14656566.2018.1476492] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Since the initial approval of everolimus in 2011, there have been a number of important changes in therapeutic/diagnostic modalities as well as classification/staging systems of neuroendocrine tumors (NETs), which can significantly impact the use of everolimus in patients with advanced NETs. Areas covered: The efficacy of everolimus monotherapy and combination therapy demonstrated in clinical studies involving patients with advanced NETs are reviewed. Several factors affecting everolimus use are described including: the development and routine use of NET classification/staging systems; widespread use of molecular imaging modalities; side effects; drug resistance; and the availability of other treatment options. Furthermore, the current position of everolimus in the treatment approach is discussed, taking into account the recommendations from the recent guidelines. Expert opinion: Although everolimus demonstrated its high efficacy and tolerability in the RADIANT trials and other clinical studies, there still remain a number of controversies related to everolimus treatment in the management of NETs. The synergistic anti-growth effect of other agents in combination with everolimus or its effect on overall survival have not been established. The appropriate order of the use of everolimus in the treatment of advanced NETs still remains unclear, which needs to be defined in further studies and will be addressed in the new guidelines.
Collapse
Affiliation(s)
- Lingaku Lee
- a Digestive Diseases Branch , National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health , Bethesda , MD , USA
| | - Tetsuhide Ito
- b Neuroendocrine Tumor Centre , Fukuoka Sanno Hospital, International University of Health and Welfare , Fukuoka , Japan
| | - Robert T Jensen
- a Digestive Diseases Branch , National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health , Bethesda , MD , USA
| |
Collapse
|
7
|
Pusceddu S, Prinzi N, Raimondi A, Corti F, Buzzoni R, Di Bartolomeo M, Seregni E, Maccauro M, Coppa J, Milione M, Mazzaferro V, de Braud F. Entering the third decade of experience with octreotide LAR in neuroendocrine tumors: A review of current knowledge. TUMORI JOURNAL 2018; 105:113-120. [DOI: 10.1177/0300891618765362] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Gastroenteropancreatic neuroendocrine tumors (NETs) are a relatively rare group of heterogeneous neoplasms. The most significant advance in therapy of NETs has been the advent of the somatostatin analog octreotide, which represents a cornerstone in their management and dramatically changed the therapeutic landscape. Octreotide long-acting release (LAR) was developed to overcome some of the limitations of octreotide. Several clinical studies, including PROMID and RADIANT-2, have validated the clinical benefits of octreotide LAR in NETs, with tumor shrinkage in about 10% of patients and tumor stabilization in roughly half of cases. While the use of octreotide LAR is well-consolidated in NETs, some open questions remain. These include the use of high-dose octreotide LAR, as there is evidence that higher dose may provide longer disease control, and nonstandard treatment schedules, with administration every 21 days instead of 28 days, as well as their use in combination with targeted agents or peptide receptor radiotherapy in clinical practice. After 3 decades of clinical experience with octreotide LAR, the drug has a well-established safety profile. It is well-tolerated and treatment discontinuations due to adverse events are uncommon. One exception is cholelithiasis, which may increase with longer duration of treatment. According to the literature data, octreotide LAR is currently recommended in both functioning and nonfunctioning advanced NETs. This review summarizes the available clinical data with octreotide LAR and also provides future perspectives on its possible uses in patients with NETs.
Collapse
Affiliation(s)
- Sara Pusceddu
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Milan, Italy
| | - Natalie Prinzi
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Milan, Italy
| | - Alessandra Raimondi
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Milan, Italy
| | - Francesca Corti
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Milan, Italy
| | - Roberto Buzzoni
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Milan, Italy
| | - Maria Di Bartolomeo
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Milan, Italy
| | - Ettore Seregni
- Department of Nuclear Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Milan, Italy
| | - Marco Maccauro
- Department of Nuclear Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Milan, Italy
| | - Jorgelina Coppa
- Department of Surgery and Liver Transplantation, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Milan, Italy
| | - Massimo Milione
- Department of Pathology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Milan, Italy
| | - Vincenzo Mazzaferro
- Department of Surgery and Liver Transplantation, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Milan, Italy
- University of Milan, Milan, Italy
| | - Filippo de Braud
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Milan, Italy
- University of Milan, Milan, Italy
| |
Collapse
|
8
|
Abstract
PURPOSE OF REVIEW To review recent advances and controversies in all aspects of carcinoid-syndrome. RECENT FINDINGS Over the last few years there have been a number of advances in all aspects of carcinoid syndrome as well as new therapies. These include new studies on its epidemiology which demonstrate it is increasing in frequency; increasing insights into the pathogenesis of its various clinical manifestations and into its natural history: definition of prognostic factors; new methods to verify its presence; the development of new drugs to treat its various manifestations, both initially and in somatostatin-refractory cases; and an increased understanding of the pathogenesis, natural history and management of carcinoid heart disease. These advances have generated several controversies and these are also reviewed. SUMMARY There have been numerous advances in all aspects of the carcinoid-syndrome, which is the most common functional syndrome neuroendocrine tumors produce. These advances are leading to new approaches to the management of these patients and in some cases to new controversies.
Collapse
Affiliation(s)
- Tetsuhide Ito
- Neuroendocrine Tumor Centre, Fukuoka Sanno Hospital, International University of Health and Welfare
| | - Lingaku Lee
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Digestive Diseases Branch, NIDDK, NIH, Bethesda, Maryland, USA
| | - Robert T Jensen
- Digestive Diseases Branch, NIDDK, NIH, Bethesda, Maryland, USA
| |
Collapse
|
9
|
Koenig A, Krug S, Mueller D, Barth PJ, Koenig U, Scharf M, Ellenrieder V, Michl P, Moll R, Homayunfar K, Kann PH, Stroebel P, Gress TM, Rinke A. Clinicopathological hallmarks and biomarkers of colorectal neuroendocrine neoplasms. PLoS One 2017; 12:e0188876. [PMID: 29232390 PMCID: PMC5726657 DOI: 10.1371/journal.pone.0188876] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Accepted: 11/14/2017] [Indexed: 12/29/2022] Open
Abstract
Chromogranin A (CgA) is a well-established marker for diagnosis and follow up of patients with gastroenteropancreatic neuroendocrine neoplasms (GEP-NEN). Recently, it has been shown that plasma levels of CgA correlate with tumor load and predict survival of patients with NEN of the small bowel. It is assumed that this is as well valid for NEN of the colon and rectum, however, this is not supported by data. To evaluate this assumption, we analyzed 62 patients with NEN of the colon and rectum listed in the Marburg GEP-NEN registry for clinicopathological characteristics, expression and plasma levels of CgA. The present study demonstrates that immunohistochemical CgA and synaptophysin are good markers for histological diagnosis in patients with NEN of the colon and rectum. However, plasma CgA is a poor marker to follow-up these patients because only a minority exhibited increased levels which did not increase significantly during tumor progression. In contrast to NEN of the small bowel, there is no correlation of CgA plasma levels with tumor burden or survival. Patients with NEN of the colon and rectum displayed a relatively good prognosis resulting in a median survival of 8.5 years. However, a subset of patients affected by G3 neoplasms, exhibited a poorer prognosis with a median survival of 2.5 years. Taken together, CgA is a valuable marker for immunohistochemical diagnosis, but CgA plasma concentration is not suitable to mirror tumor burden or prognosis in patients with NEN of the colon and rectum.
Collapse
Affiliation(s)
- Alexander Koenig
- Department of Gastroenterology and Endocrinology, Philipps-University of Marburg, Marburg, Germany
- Department of Gastroenterology and Gastrointestinal Oncology, University Medical Center Goettingen, Goettingen, Germany
- * E-mail: (AK); (AR)
| | - Sebastian Krug
- Department of Gastroenterology and Endocrinology, Philipps-University of Marburg, Marburg, Germany
- Department of Internal Medicine I, University Halle, Halle, Germany
| | - Daniela Mueller
- Department of Gastroenterology and Endocrinology, Philipps-University of Marburg, Marburg, Germany
| | - Peter J. Barth
- Gerhard-Domagk-Institute of Pathology, University of Muenster, Muenster, Germany
- Department of Pathology, Philipps-University of Marburg, Marburg, Germany
| | - Ute Koenig
- Department of Gastroenterology and Gastrointestinal Oncology, University Medical Center Goettingen, Goettingen, Germany
| | - Michael Scharf
- Department of Gastroenterology and Endocrinology, Philipps-University of Marburg, Marburg, Germany
| | - Volker Ellenrieder
- Department of Gastroenterology and Gastrointestinal Oncology, University Medical Center Goettingen, Goettingen, Germany
| | - Patrick Michl
- Department of Internal Medicine I, University Halle, Halle, Germany
| | - Roland Moll
- Department of Pathology, Philipps-University of Marburg, Marburg, Germany
| | - Kia Homayunfar
- Department of General-, Visceral- and Pediatric Surgery University Medical Center Goettingen, Goettingen, Germany
| | - Peter Herbert Kann
- Department of Gastroenterology and Endocrinology, Philipps-University of Marburg, Marburg, Germany
| | - Philipp Stroebel
- Department of Pathology, University Medical Center Goettingen, Goettingen, Germany
| | - Thomas M. Gress
- Department of Gastroenterology and Endocrinology, Philipps-University of Marburg, Marburg, Germany
| | - Anja Rinke
- Department of Gastroenterology and Endocrinology, Philipps-University of Marburg, Marburg, Germany
- * E-mail: (AK); (AR)
| |
Collapse
|
10
|
Neuzillet C, de Mestier L, Rousseau B, Mir O, Hebbar M, Kocher HM, Ruszniewski P, Tournigand C. Unravelling the pharmacologic opportunities and future directions for targeted therapies in gastro-intestinal cancers part 2: Neuroendocrine tumours, hepatocellular carcinoma, and gastro-intestinal stromal tumours. Pharmacol Ther 2017; 181:49-75. [PMID: 28723416 DOI: 10.1016/j.pharmthera.2017.07.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Until the 1990s, cytotoxic chemotherapy has been the cornerstone of medical therapy for gastrointestinal (GI) cancers. Better understanding of the cancer cell molecular biology has led to the therapeutic revolution of targeted therapies, i.e. monoclonal antibodies or small molecule inhibitors directed against proteins that are specifically overexpressed or mutated in cancer cells. These agents, being more specific to cancer cells, were expected to be less toxic than conventional cytotoxic agents. However, their effects have sometimes been disappointing, due to intrinsic or acquired resistance mechanisms, or to an activity restricted to some tumour settings, illustrating the importance of patient selection and early identification of predictive biomarkers of response to these therapies. Targeted agents have provided clinical benefit in many GI cancer types. Particularly, some GI tumours are considered chemoresistant and targeted therapies have offered a new therapeutic base for their management. Hence, somatostatin receptor-directed strategies, sorafenib, and imatinib have revolutioned the management of neuroendocrine tumours (NET), hepatocellular carcinoma (HCC), and gastrointestinal stromal tumours (GIST), respectively, and are now used as first-line treatment in many patients affected by these tumours. However, these agents face problems of resistances and identification of predictive biomarkers from imaging and/or biology. We propose a comprehensive two-part review providing a panoramic approach of the successes and failures of targeted agents in GI cancers to unravel the pharmacologic opportunities and future directions for these agents in GI oncology. In this second part, we will focus on NET, HCC, and GIST, whose treatment relies primarily on targeted therapies.
Collapse
Affiliation(s)
- Cindy Neuzillet
- INSERM UMR1149, Beaujon University Hospital (Assistance Publique-Hôpitaux de Paris, AP-HP), Paris 7 Diderot University, 100 Boulevard du Général Leclerc, 92110 Clichy, France; Department of Medical Oncology, Henri Mondor University Hospital (AP-HP), Paris Est Créteil University (UPEC), 51 Avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France; Tumour Biology Laboratory, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom; Barts and The London HPB Centre, The Royal London Hospital, Whitechapel, London E1 1BB, United Kingdom.
| | - Louis de Mestier
- INSERM UMR1149, Beaujon University Hospital (Assistance Publique-Hôpitaux de Paris, AP-HP), Paris 7 Diderot University, 100 Boulevard du Général Leclerc, 92110 Clichy, France; Department of Gastroenterology and Pancreatology, Beaujon University Hospital (AP-HP), Paris 7 Diderot University, 100 Boulevard du Général Leclerc, 92110 Clichy, France
| | - Benoît Rousseau
- Department of Medical Oncology, Henri Mondor University Hospital (AP-HP), Paris Est Créteil University (UPEC), 51 Avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France; Institut Mondor de Recherche Biomédicale, INSERM UMR955 Team 18, Paris Est Créteil University (UPEC), 51 Avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France
| | - Olivier Mir
- Department of Cancer Medicine - Sarcoma Group, Department of Early Drug Development (DITEP) - Phase 1 Unit, Gustave Roussy Cancer Campus, University of Paris Sud, 114, Rue Edouard Vaillant, 94800 Villejuif, France
| | - Mohamed Hebbar
- Department of Medical Oncology, Lille University Hospital, 1, Rue Polonovski, 59037 Lille, France
| | - Hemant M Kocher
- Tumour Biology Laboratory, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom; Barts and The London HPB Centre, The Royal London Hospital, Whitechapel, London E1 1BB, United Kingdom
| | - Philippe Ruszniewski
- INSERM UMR1149, Beaujon University Hospital (Assistance Publique-Hôpitaux de Paris, AP-HP), Paris 7 Diderot University, 100 Boulevard du Général Leclerc, 92110 Clichy, France
| | - Christophe Tournigand
- Department of Medical Oncology, Henri Mondor University Hospital (AP-HP), Paris Est Créteil University (UPEC), 51 Avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France
| |
Collapse
|
11
|
Sullivan I, Le Teuff G, Guigay J, Caramella C, Berdelou A, Leboulleux S, Déandréis D, Hadoux J, Ducreux M, Duvillard P, Adam J, Scoazec JY, Baudin E, Planchard D. Antitumour activity of somatostatin analogues in sporadic, progressive, metastatic pulmonary carcinoids. Eur J Cancer 2017; 75:259-267. [DOI: 10.1016/j.ejca.2016.11.034] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 10/17/2016] [Accepted: 11/11/2016] [Indexed: 11/27/2022]
|
12
|
Merola E, Panzuto F, Fave GD. Antiproliferative effect of somatostatin analogs in advanced gastro-entero-pancreatic neuroendocrine tumors: a systematic review and meta-analysis. Oncotarget 2017; 8:46624-46634. [PMID: 28402955 PMCID: PMC5542298 DOI: 10.18632/oncotarget.16686] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 03/17/2017] [Indexed: 01/12/2023] Open
Abstract
A meta-analysis has systematically investigated the antineoplastic efficacy and safety of somatostatin analogs (SSAs) in advanced gastro-entero-pancreatic neuroendocrine tumors (GEP-NETs). Randomized controlled trials (RCTs) reporting the hazard ratio (HR) for disease progression (DP) were evaluated. Response rate and risk ratio (RR) for adverse events were also analyzed. A total of 289 patients (143 receiving SSAs vs. 146 placebo) were evaluated from two RCTs. A significant benefit from SSAs in terms of disease control was observed (HR 0.41, 95% CI: 0.29 to 0.58, P < 0.01; I20%), response rate being 58.0% vs. 32.2%, respectively. The occurrence of adverse events significantly differed from the placebo arm only in terms of biliary stones (RR 3.79, 95% CI: 1.28 to 11.17, P = 0.02; I20%). In conclusion, SSAs showed an antiproliferative effect in advanced GEP-NETs, with a good safety profile.
Collapse
Affiliation(s)
- Elettra Merola
- Department of Digestive and Liver Diseases, Sapienza University, Sant'Andrea Hospital, Rome, Italy
| | - Francesco Panzuto
- Department of Digestive and Liver Diseases, Sapienza University, Sant'Andrea Hospital, Rome, Italy
| | - Gianfranco Delle Fave
- Department of Digestive and Liver Diseases, Sapienza University, Sant'Andrea Hospital, Rome, Italy
| |
Collapse
|
13
|
Öberg K, Lamberts SWJ. Somatostatin analogues in acromegaly and gastroenteropancreatic neuroendocrine tumours: past, present and future. Endocr Relat Cancer 2016; 23:R551-R566. [PMID: 27697899 DOI: 10.1530/erc-16-0151] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 10/03/2016] [Indexed: 11/08/2022]
Abstract
Acromegaly is a hormonal disorder that arises when the pituitary gland secretes excess growth hormone (GH), which in turn stimulates a concomitant increase in serum insulin-like growth factor 1 (IGF-1) levels. Gastroenteropancreatic neuroendocrine tumours (GEP-NET) constitute a heterogeneous group of tumours that can secrete serotonin and a variety of peptide hormones that may cause characteristic symptoms known as carcinoid syndrome or other symptoms and hormonal hypersecretion syndromes depending on the tumour's site of origin. Current medical therapy for the treatment of acromegaly and GEP-NET involves the administration of somatostatin analogues that effectively suppress excess hormone secretion. After its discovery in 1979, octreotide became the first synthetic biologically stable somatostatin analogue with a short-acting formulation of octreotide introduced into clinical practice in the late 1980s. Lanreotide, another somatostatin analogue, became available in the mid-1990s initially as a prolonged-release formulation administered every 10 or 14 days. Long-acting release formulations of both octreotide (Sandostatin LAR and Novartis) and lanreotide (Somatuline Autogel, Ipsen), based on microparticle and nanoparticle drug-delivery technologies, respectively, were later developed, which allowed for once-monthly administration and improved convenience. First-generation somatostatin analogues remain one of the cornerstones of medical therapy in the management of pituitary and GEP-NET hormone hypersecretion, with octreotide having the longest established efficacy and safety profile of the somatostatin analogue class. More recently, pasireotide (Signifor), a next-generation multireceptor-targeted somatostatin analogue, has emerged as an alternative therapeutic option for the treatment of acromegaly. This review summarizes the development and clinical success of somatostatin analogues.
Collapse
|
14
|
Walls GV, Stevenson M, Soukup BS, Lines KE, Grossman AB, Schmid HA, Thakker RV. Pasireotide Therapy of Multiple Endocrine Neoplasia Type 1-Associated Neuroendocrine Tumors in Female Mice Deleted for an Men1 Allele Improves Survival and Reduces Tumor Progression. Endocrinology 2016; 157:1789-98. [PMID: 26990064 PMCID: PMC4870877 DOI: 10.1210/en.2015-1965] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Pasireotide, a somatostatin analog, is reported to have anti-proliferative effects in neuroendocrine tumors (NETs). We therefore assessed the efficacy of pasireotide for treating pancreatic and pituitary NETs that develop in a mouse model of multiple endocrine neoplasia type 1 (MEN1). Men1(+/-) mice were treated from age 12 mo with 40 mg/kg pasireotide long-acting release formulation, or PBS, intramuscularly monthly for 9 mo. The Men1(+/-) mice had magnetic resonance imaging at 12 and 21 mo, and from 20 mo oral 5-bromo-2-deoxyuridine for 1 mo, to assess tumor development and proliferation, respectively. NETs were collected at age 21 mo, and proliferation and apoptosis assessed by immunohistochemistry and TUNEL assays, respectively. Pasireotide-treated Men1(+/-) mice had increased survival (pasireotide, 80.9% vs PBS, 65.2%; P < .05), with fewer mice developing pancreatic NETs (pasireotide, 86.9% vs PBS, 96.9%; P < .05) and smaller increases in pituitary NET volumes (pre-treated vs post-treated, 0.803 ± 0.058 mm(3) vs 2.872 ± 0.728 mm(3) [pasireotide] compared with 0.844 ± 0.066 mm(3) vs 8.847 ±1.948 mm(3) [PBS]; P < .01). In addition, pasireotide-treated mice had fewer pancreatic NETs compared with PBS-treated mice (2.36 ± 0.25 vs 3.72 ± 0.32, respectively; P < .001), with decreased proliferation in pancreatic NETs (pasireotide, 0.35 ± 0.03% vs PBS, 0.78 ± 0.08%; P < .0001) and pituitary NETs (pasireotide, 0.73 ±0.07% vs PBS, 1.81 ± 0.15%; P < .0001), but increased apoptosis in pancreatic NETs (pasireotide, 0.42 ± 0.05% vs PBS, 0.19 ± 0.03%; P < .001) and pituitary NETs (pasireotide, 14.75 ± 1.58% vs PBS, 2.35 ± 0.44%; P < .001). Thus, pasireotide increased survival and inhibited pancreatic and pituitary NET growth, thereby indicating its potential as an anti-proliferative and pro-apoptotic therapy.
Collapse
Affiliation(s)
- Gerard V Walls
- Academic Endocrine Unit (G.V.W., M.S., B.S.S., K.E.L., R.V.T.), Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), University of Oxford, Churchill Hospital, Oxford, OX3 7LJ, United Kingdom; Nuffield Department of Surgical Sciences (G.V.W., B.S.S.), University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom; Department of Endocrinology (A.B.G.), OCDEM, Churchill Hospital, Oxford, OX3 7LJ, United Kingdom; and Novartis Pharma AG (H.A.S.), Novartis Institutes for Biomedical Research, Oncology, CH-4057 Basel, Switzerland
| | - Mark Stevenson
- Academic Endocrine Unit (G.V.W., M.S., B.S.S., K.E.L., R.V.T.), Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), University of Oxford, Churchill Hospital, Oxford, OX3 7LJ, United Kingdom; Nuffield Department of Surgical Sciences (G.V.W., B.S.S.), University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom; Department of Endocrinology (A.B.G.), OCDEM, Churchill Hospital, Oxford, OX3 7LJ, United Kingdom; and Novartis Pharma AG (H.A.S.), Novartis Institutes for Biomedical Research, Oncology, CH-4057 Basel, Switzerland
| | - Benjamin S Soukup
- Academic Endocrine Unit (G.V.W., M.S., B.S.S., K.E.L., R.V.T.), Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), University of Oxford, Churchill Hospital, Oxford, OX3 7LJ, United Kingdom; Nuffield Department of Surgical Sciences (G.V.W., B.S.S.), University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom; Department of Endocrinology (A.B.G.), OCDEM, Churchill Hospital, Oxford, OX3 7LJ, United Kingdom; and Novartis Pharma AG (H.A.S.), Novartis Institutes for Biomedical Research, Oncology, CH-4057 Basel, Switzerland
| | - Kate E Lines
- Academic Endocrine Unit (G.V.W., M.S., B.S.S., K.E.L., R.V.T.), Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), University of Oxford, Churchill Hospital, Oxford, OX3 7LJ, United Kingdom; Nuffield Department of Surgical Sciences (G.V.W., B.S.S.), University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom; Department of Endocrinology (A.B.G.), OCDEM, Churchill Hospital, Oxford, OX3 7LJ, United Kingdom; and Novartis Pharma AG (H.A.S.), Novartis Institutes for Biomedical Research, Oncology, CH-4057 Basel, Switzerland
| | - Ashley B Grossman
- Academic Endocrine Unit (G.V.W., M.S., B.S.S., K.E.L., R.V.T.), Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), University of Oxford, Churchill Hospital, Oxford, OX3 7LJ, United Kingdom; Nuffield Department of Surgical Sciences (G.V.W., B.S.S.), University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom; Department of Endocrinology (A.B.G.), OCDEM, Churchill Hospital, Oxford, OX3 7LJ, United Kingdom; and Novartis Pharma AG (H.A.S.), Novartis Institutes for Biomedical Research, Oncology, CH-4057 Basel, Switzerland
| | - Herbert A Schmid
- Academic Endocrine Unit (G.V.W., M.S., B.S.S., K.E.L., R.V.T.), Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), University of Oxford, Churchill Hospital, Oxford, OX3 7LJ, United Kingdom; Nuffield Department of Surgical Sciences (G.V.W., B.S.S.), University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom; Department of Endocrinology (A.B.G.), OCDEM, Churchill Hospital, Oxford, OX3 7LJ, United Kingdom; and Novartis Pharma AG (H.A.S.), Novartis Institutes for Biomedical Research, Oncology, CH-4057 Basel, Switzerland
| | - Rajesh V Thakker
- Academic Endocrine Unit (G.V.W., M.S., B.S.S., K.E.L., R.V.T.), Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), University of Oxford, Churchill Hospital, Oxford, OX3 7LJ, United Kingdom; Nuffield Department of Surgical Sciences (G.V.W., B.S.S.), University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom; Department of Endocrinology (A.B.G.), OCDEM, Churchill Hospital, Oxford, OX3 7LJ, United Kingdom; and Novartis Pharma AG (H.A.S.), Novartis Institutes for Biomedical Research, Oncology, CH-4057 Basel, Switzerland
| |
Collapse
|
15
|
Sbardella E, Grossman A. New Developments in the Treatment of Neuroendocrine Tumours - RADIANT-4, NETTER-1 and Telotristat Etiprate. EUROPEAN ENDOCRINOLOGY 2016; 12:44-46. [PMID: 29632589 PMCID: PMC5813460 DOI: 10.17925/ee.2016.12.01.44] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 02/29/2016] [Indexed: 11/24/2022]
Abstract
Neuroendocrine tumours (NETs) are a heterogeneous group of neoplasms whose incidence has increased significantly in recent years, and whose optimal management remains controversial. We report the latest innovations in their management, in particular the results of three trials concerning the use of the mammalian target of rapamycin (mTOR) inhibitor, everolimus, in non-functional NETs of lung/ gastrointestinal (GI) origin, the first randomised trial of radiolabelled 177Lu-DOTATATE in patients with mid-gut NETs, and the use of the 5-HT synthesis inhibitor, telotristat etiprate, in patients with the carcinoid syndrome.
Collapse
Affiliation(s)
- Emilia Sbardella
- Department of Endocrinology, Oxford Centre for Diabetes, Endocrinology and Metabolism, Churchill Hospital, University of Oxford, Oxford, UK
| | - Ashley Grossman
- Department of Endocrinology, Oxford Centre for Diabetes, Endocrinology and Metabolism, Churchill Hospital, University of Oxford, Oxford, UK
| |
Collapse
|