201
|
Panzuto F, Cicchese N, Partelli S, Rinzivillo M, Capurso G, Merola E, Manzoni M, Pucci E, Iannicelli E, Pilozzi E, Rossi M, Doglioni C, Falconi M, Delle Fave G. Impact of Ki67 re-assessment at time of disease progression in patients with pancreatic neuroendocrine neoplasms. PLoS One 2017; 12:e0179445. [PMID: 28644861 PMCID: PMC5482443 DOI: 10.1371/journal.pone.0179445] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 05/29/2017] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Although re-assessment of proliferative activity by K67 evaluation during the course of neuroendocrine neoplasms (NENs) is recommended in selected patients, its impact on patients' management is not clear due to the lack of data supporting this practice. AIM To investigate Ki67 change at time of progressive disease (PD) in entero-pancreatic NENs (EP-NENs). PATIENTS AND METHODS Retrospective analysis of sporadic EP-NENs which received histological re-assessment after PD once radiologically documented. RESULTS Forty-three patients were evaluated, including 24 pancreatic NENs (PNENs), and 19 small intestine NENs (SI-NENs). At time of initial histological evaluation, 19 patients had grade 1 (G1) NETs (44.2%), and 24 grade 2 (G2) NETs (55.8%), overall median Ki67 being 3% (range 1%-20%). At time of PD, 13 patients had G1 NETs (30.2%), 26 G2 NETs (60.5%), and 4 had grade 3 (G3) NECs (9.3%), thus resulting in a significant median Ki67 increase (8%, range 1%-70%; p = 0.0006), and a G upgrading in 12 patients (27.9%). A statistically significant Ki67 increase and G grading change at time of PD was observed in PNENs (p = 0.0005 and p = 0.028, respectively). Conversely, no statistically significant change occurred in non-PNENs. CONCLUSIONS In PNENs with documented PD, Ki67 increase occurs in a significant proportion of patients, providing useful information necessary to choose appropriate therapeutic options.
Collapse
Affiliation(s)
- Francesco Panzuto
- Digestive and Liver Disease Unit, Sant’Andrea Hospital Sapienza University of Rome, Roma, Italy
| | - Noemi Cicchese
- Digestive and Liver Disease Unit, Sant’Andrea Hospital Sapienza University of Rome, Roma, Italy
| | - Stefano Partelli
- Pancreatic Surgery Unit, Pancreas Translational & Clinical Research Center, San Raffaele Scientific Institute, “Vita-Salute” University, Milan, Italy
| | - Maria Rinzivillo
- Digestive and Liver Disease Unit, Sant’Andrea Hospital Sapienza University of Rome, Roma, Italy
| | - Gabriele Capurso
- Digestive and Liver Disease Unit, Sant’Andrea Hospital Sapienza University of Rome, Roma, Italy
| | - Elettra Merola
- Digestive and Liver Disease Unit, Sant’Andrea Hospital Sapienza University of Rome, Roma, Italy
| | - Marco Manzoni
- Department of Endocrinology and Internal Medicine, San Raffaele Hospital Scientific Institute, Milan, Italy
| | - Eugenio Pucci
- Department of Experimental Medicine and Pathology, Sant’Andrea Hospital Sapienza University of Rome, Roma, Italy
| | - Elsa Iannicelli
- Department of Radiology, Sant’Andrea Hospital Sapienza University of Rome, Roma, Italy
| | - Emanuela Pilozzi
- Department of Experimental Medicine and Pathology, Sant’Andrea Hospital Sapienza University of Rome, Roma, Italy
| | - Michele Rossi
- Department of Radiology, Sant’Andrea Hospital Sapienza University of Rome, Roma, Italy
| | - Claudio Doglioni
- Department of Pathology, San Raffaele Scientific Institute, “Vita-Salute” University, Milan, Italy
| | - Massimo Falconi
- Pancreatic Surgery Unit, Pancreas Translational & Clinical Research Center, San Raffaele Scientific Institute, “Vita-Salute” University, Milan, Italy
| | - Gianfranco Delle Fave
- Digestive and Liver Disease Unit, Sant’Andrea Hospital Sapienza University of Rome, Roma, Italy
| |
Collapse
|
202
|
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
|
203
|
Favorable response of colonic mixed adenoneuroendocrine carcinoma to streptozocin monotherapy. Int Cancer Conf J 2017; 6:175-179. [PMID: 31149497 DOI: 10.1007/s13691-017-0301-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 05/31/2017] [Indexed: 10/19/2022] Open
Abstract
Mixed adenoneuroendocrine carcinoma (MANEC) of the colon is rare and has a poor prognosis. Here, we report a case of MANEC in the ascending colon, in which streptozocin monotherapy achieved a partial response. A 36-year-old woman underwent right hemicolectomy for colonic polyposis, which included ascending colon cancer. Pathological examination revealed that some mucosal polyps were adenocarcinoma while one submucosal polyp was neuroendocrine carcinoma. Adjuvant chemotherapy was not administered, and 5 months after the operation, multiple liver metastases were identified. She was started on modified (5-FU, leucovorin, oxaliplatin) followed by XELOX (capecitabine, oxaliplatin) plus bevacizumab. Although these regimens helped achieve stable disease, computed tomography showed that the hepatic metastatic lesions had enlarged 4 months later. Subsequently, the regimen was changed to streptozocin monotherapy (1000 mg/m2, weekly). After 5 cycles, the regimen achieved partial response and was continued for a total of 17 courses without significant adverse events until progressive disease. As a third-line chemotherapy regimen, cisplatin plus etoposide (EP) was administered. The EP regimen reduced the size of the hepatic and ovarian metastatic lesions but severe neutropenia and anemia was observed. Amrubicin monotherapy was also administered as fourth-line chemotherapy but a good clinical response was not detected, and the patient died 20 months after the operation. Streptozocin monotherapy has the potential to be a therapeutic option for mixed adenoneuroendocrine carcinoma of the colon.
Collapse
|
204
|
Bijlsma MF, Sadanandam A, Tan P, Vermeulen L. Molecular subtypes in cancers of the gastrointestinal tract. Nat Rev Gastroenterol Hepatol 2017; 14:333-342. [PMID: 28400627 DOI: 10.1038/nrgastro.2017.33] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Malignancies of the gastrointestinal tract are among the most common human cancers. The distinct tissues of origin give rise to a diverse set of diseases, such as colorectal cancer, pancreatic carcinoma and gastric cancers, with each associating with specific clinical features. Genomic and transcriptomic analyses have further defined the heterogeneity that occurs within these cancers by identifying so-called molecular subtypes. These subtypes are characterized by specific genetic aberrations and expression signatures that suggest important biological differences. Although at first sight this subdivision of organ-specific cancers might increase the complexity of classification, closer analysis suggests that the subtypes detected in the various malignancies are recurring. For example, nearly all gastrointestinal cancers appear to present with subtypes that are either characterized by a mesenchymal gene expression signatures, extensive immune infiltration or metabolic dysregulation. Additionally, in each of the gastrointestinal malignancies, a 'canonical' subtype is recognized that retains characteristic features of the epithelial tissue of origin. These common themes can enhance our collective understanding of these malignancies, and could perhaps be therapeutically exploited. In this Review, the identification of subtypes in the various gastrointestinal cancer types are discussed along with how they could be incorporated into clinical practice.
Collapse
Affiliation(s)
- Maarten F Bijlsma
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Anguraj Sadanandam
- Division of Molecular Pathology, The Institute of Cancer Research, 15 Cotswold Road, London SM2 5NG, UK
| | - Patrick Tan
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | - Louis Vermeulen
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
205
|
Lappano R, Maggiolini M. Pharmacotherapeutic Targeting of G Protein-Coupled Receptors in Oncology: Examples of Approved Therapies and Emerging Concepts. Drugs 2017; 77:951-965. [PMID: 28401445 DOI: 10.1007/s40265-017-0738-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
G protein-coupled receptors (GPCRs) are involved in numerous physio-pathological processes, including the stimulation of cancer progression. In this regard, it should be mentioned that although GPCRs may represent major pharmaceutical targets, only a few drugs acting as GPCR inhibitors are currently used in anti-tumor therapies. For instance, certain pro-malignancy effects mediated by GPCRs are actually counteracted by the use of small molecules and peptides that function as receptor antagonists or inverse agonists. Recently, humanized monoclonal antibodies targeting GPCRs have also been developed. Here, we review the current GPCR-targeted therapies for cancer treatment, summarizing the clinical studies that led to their official approval. We provide a broad overview of the mechanisms of action of the available anti-cancer drugs targeting gonadotropin-releasing hormone, somatostatin, chemokine, and Smoothened receptors. In addition, we discuss the anti-tumor potential of novel non-approved molecules and antibodies able to target some of the aforementioned GPCRs in different experimental models and clinical trials. Likewise, we focus on the repurposing in cancer patients of non-oncological GPCR-based drugs, elucidating the rationale behind this approach and providing clinical evidence on their safety and efficacy.
Collapse
Affiliation(s)
- Rosamaria Lappano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy.
| | - Marcello Maggiolini
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy.
| |
Collapse
|
206
|
Abstract
Pancreatic neuroendocrine tumors (PanNETs) manifest with a range of symptoms and pose a therapeutic challenge. A team approach, in which many specialists come together, is necessary in the quest for the best patient-tailored treatment. Disciplines such as oncology, surgery, basic science, endocrinology, radiology, and nuclear medicine need to work side by side, equally contributing to patient care and to advancing our better understanding of this fascinating disease.
Collapse
Affiliation(s)
| | - Christos Dervenis
- Department of Surgical Oncology and HPB Surgery, Metropolitan Hospital, Athens, Greece
| |
Collapse
|
207
|
Bar-Moshe Y, Mazeh H, Grozinsky-Glasberg S. Non-functioning pancreatic neuroendocrine tumors: Surgery or observation? World J Gastrointest Endosc 2017; 9:153-161. [PMID: 28465781 PMCID: PMC5394721 DOI: 10.4253/wjge.v9.i4.153] [Citation(s) in RCA: 16] [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: 08/28/2016] [Revised: 10/23/2016] [Accepted: 01/14/2017] [Indexed: 02/05/2023] Open
Abstract
Incidentally detected, sporadic, nonfunctional pancreatic neuroendocrine tumors are increasingly diagnosed on imaging studies performed for unrelated purposes. Although their resection is usually recommended, controversy still exists regarding their optimal management, due to their highly variable and difficult to predict biologic behavior. Recently, several studies and guidelines advocated an expectant management approach in small size, low grade, incidentally diagnosed nonfunctional pancreatic neuroendocrine tumors. The aim of this study is to review and summarize the available literature addressing nonfunctional pancreatic neuroendocrine tumors, with an emphasis on surgical management controversies.
Collapse
|
208
|
Translational research in neuroendocrine tumors: pitfalls and opportunities. Oncogene 2017; 36:1899-1907. [PMID: 27641330 DOI: 10.1038/onc.2016.316] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 07/19/2016] [Accepted: 07/22/2016] [Indexed: 12/16/2022]
Abstract
Interest in research on neuroendocrine tumors (NETs) has grown in the past 10 years, coinciding with improvements in our understanding of the molecular pathogenesis of NETs. In addition, NETs have become one of the most exciting settings for drug development. Two targeted agents for the management of advanced pancreatic NETs have been approved, but the development of targeted agents for NETs is limited by problems with both patient selection and demonstration of activity. In this review, we analyze these limitations and discuss ways to increase the predictive value of preclinical models for target discovery and drug development. The role of translational research and 'omics' methodologies is emphasized, with the final aim of developing personalized medicine. Because NETs usually grow slowly and metastatic tumors are found at easily accessible locations, and owing to improvements in techniques for liquid biopsies, NETs provide a unique opportunity to obtain tumor samples at all stages of the evolution of the disease and to adapt treatment to changes in tumor biology. Combining clinical and translational research is essential to achieve progress in the NET field. Slow growth and genetic stability limit and challenge both the availability and further development of preclinical models of NETs, one of the most crucial unmet research needs in the field. Finally, we suggest some useful approaches for improving clinical drug development for NETs: moving from classical RECIST-based response end points to survival parameters; searching for different criteria to define response rates (for example, antiangiogenic effects and metabolic responses); implementing randomized phase II studies to avoid single-arm phase II studies that produce limited data on drug efficacy; and using predictive biomarkers for patient selection.
Collapse
|
209
|
Dias AR, Azevedo BC, Alban LBV, Yagi OK, Ramos MFKP, Jacob CE, Barchi LC, Cecconello I, Ribeiro U, Zilberstein B. GASTRIC NEUROENDOCRINE TUMOR: REVIEW AND UPDATE. ARQUIVOS BRASILEIROS DE CIRURGIA DIGESTIVA : ABCD = BRAZILIAN ARCHIVES OF DIGESTIVE SURGERY 2017; 30:150-154. [PMID: 29257854 PMCID: PMC5543797 DOI: 10.1590/0102-6720201700020016] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 09/21/2016] [Indexed: 02/08/2023]
Abstract
INTRODUCTION The frequency of gastric neuroendocrine tumors is increasing. Reasons are the popularization of endoscopy and its technical refinements. Despite this, they are still poorly understood and have complex management. AIM Update the knowledge on gastric neuroendocrine tumor and expose the future perspectives on the diagnosis and treatment of this disease. METHOD Literature review using the following databases: Medline/PubMed, Cochrane Library and SciELO. Search terms were: gastric carcinoid, gastric neuroendocrine tumor, treatment. From the selected articles, 38 were included in this review. RESULTS Gastric neuroendocrine tumors are classified in four clinical types. Correct identification of the clinical type and histological grade is fundamental, since treatment varies accordingly and defines survival. CONCLUSION Gastric neuroendocrine tumors comprise different subtypes with distinct management and prognosis. Correct identification allows for a tailored therapy. Further studies will clarify the diseases biology and improve its treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ivan Cecconello
- School of Medicine, University of São Paulo, São Paulo, SP, Brazil
| | | | | |
Collapse
|
210
|
Horiguchi S, Kato H, Shiraha H, Tsutsumi K, Yamamoto N, Matsumoto K, Tomoda T, Uchida D, Akimoto Y, Mizukawa S, Tanaka T, Ichimura K, Takaki A, Yagi T, Okada H. Dynamic computed tomography is useful for prediction of pathological grade in pancreatic neuroendocrine neoplasm. J Gastroenterol Hepatol 2017; 32:925-931. [PMID: 27637470 DOI: 10.1111/jgh.13594] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/05/2016] [Indexed: 01/25/2023]
Abstract
BACKGROUND AND AIM Pathological grading is important in defining the therapeutic strategy in pancreatic neuroendocrine neoplasm (PNEN) but is difficult for unresectable cases. Endoscopic ultrasound (EUS)-guided fine needle aspiration (FNA) is useful in the diagnosis of PNEN, but its usefulness for pathological grading is not well established. No studies have examined the diagnostic ability of dynamic computed tomography (CT) for pathological grading of PNEN. We investigated the usefulness of EUS-FNA and dynamic CT in the diagnosis and pathological grading of PNEN. METHODS In this retrospective study, 39 PNEN patients finally diagnosed via EUS-FNA and/or surgical resection underwent dynamic CT. Pathological samples were diagnosed based on WHO2010; staging was based on the European Neuroendocrine Tumor Society classification. The proportion of the quantification value in the tumor to the pancreatic parenchyma in arterial phase was defined as the CT ratio. Immunohistochemical staining with CD31 was performed to evaluate microvessel density (MVD). We evaluated the relationship between pathological grade, CT ratio, and MVD. RESULTS By using EUS-FNA, 35 of 39 (90%) cases were diagnosed as PNEN. As for pathological grade, 15 of 35 (43%) cases could be identified correctly. CT ratio could predict pathological Grade 3 disease. The sensitivity, specificity, and diagnostic accuracy were 100%, 94%, and 95%. MVD was significantly correlated with CT ratio (r = 0.83, P < 0.0001) and pathological grade (P = 0.0074). CONCLUSIONS Computed tomography ratio has a relationship with pathological grade in PNEN, which would help decide therapeutic strategy in unresectable cases and cases in which pathological grading is difficult.
Collapse
Affiliation(s)
- Shigeru Horiguchi
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Science, Okayama, Japan
| | - Hironari Kato
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Science, Okayama, Japan
| | - Hidenori Shiraha
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Science, Okayama, Japan
| | - Koichiro Tsutsumi
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Science, Okayama, Japan
| | - Naoki Yamamoto
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Science, Okayama, Japan
| | - Kazuyuki Matsumoto
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Science, Okayama, Japan
| | - Takeshi Tomoda
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Science, Okayama, Japan
| | - Daisuke Uchida
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Science, Okayama, Japan
| | - Yutaka Akimoto
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Science, Okayama, Japan
| | - Syou Mizukawa
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Science, Okayama, Japan
| | - Takehiro Tanaka
- Department of Pathology and Oncology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Koichi Ichimura
- Department of Pathology and Oncology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Akinobu Takaki
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Science, Okayama, Japan
| | - Takahito Yagi
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hiroyuki Okada
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Science, Okayama, Japan
| |
Collapse
|
211
|
Surveillance strategy for small asymptomatic non-functional pancreatic neuroendocrine tumors - a systematic review and meta-analysis. HPB (Oxford) 2017; 19:310-320. [PMID: 28254159 DOI: 10.1016/j.hpb.2016.12.010] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 12/11/2016] [Accepted: 12/22/2016] [Indexed: 02/06/2023]
Abstract
BACKGROUND Non-functional pancreatic neuroendocrine tumors (NF-PNET) are rare neoplasms being increasingly diagnosed. Surgical treatment or expectant management are both suggested for small NF-PNETs. The aim of this study was to evaluate the outcome of surveillance strategy for small NF-PNETs. METHODS A systematic search was performed up to March 2016 in MEDLINE, EMBASE and the Cochrane Library according to the PRISMA guidelines. Data was pooled using the random-effects model. RESULTS Nine articles including 344 patients with sporadic and 64 patients with MEN1 related NF-PNET were selected. Tumor growth was observed in 22% and 52%, development of metastases were reported on 0% and 9%, and rate of secondary surgical resection was 12% and 25% in patients with sporadic or MEN1 related NF-PNETs, respectively. All metastases (1 distant, 4 nodal) were reported by a single study in patients with MEN1. Reason for secondary surgery was tumor growth in half of patients undergoing surgery. DISCUSSION Expectant management of small asymptomatic, sporadic, NF-PNETs could be a reasonable option in highly selected patients. However, the level of evidence is low and longer follow-up is needed to identify patients could benefit from upfront surgery instead of expectant treatment.
Collapse
|
212
|
Yalcin S, Bayram F, Erdamar S, Kucuk O, Oruc N, Coker A. Gastroenteropancreatic neuroendocrine tumors: recommendations of Turkish multidisciplinary neuroendocrine tumor study group on diagnosis, treatment and follow-up. Arch Med Sci 2017; 13:271-282. [PMID: 28261279 PMCID: PMC5332464 DOI: 10.5114/aoms.2017.65449] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 05/20/2015] [Indexed: 02/06/2023] Open
Abstract
Gastroenteropancreatic neuroendocrine tumors (GEPNETs) are a relatively rare, heterogeneous group of diseases in which important advances have been observed in the diagnosis and treatment as well as in our understanding of the biology and genetics of the disease in recent years. Given the insufficient scientific data available on evidence-based management of GEPNETs and the differences in circumstances in individual countries, a multidisciplinary study group was established to provide guidelines for the management of GEPNETS. This study group consisted of a medical oncologist, endocrinologist, surgeon, pathologist, gastroenterologist, and a nuclear medicine specialist, who aimed to prepare a practical guide in the light of existing scientific data and international guidelines, to be used in common clinical practice.
Collapse
Affiliation(s)
- Suayib Yalcin
- Department of Medical Oncology, Institute of Cancer, Hacettepe University, Ankara, Turkey
| | - Fahri Bayram
- Department of Endocrinology, Erciyes University, Kayseri, Turkey
| | - Sibel Erdamar
- Department of Pathology, Cerrahpasa Medical School, Istanbul, Turkey
| | - Ozlem Kucuk
- Department of Nuclear Medicine, Ankara University, Ankara, Turkey
| | - Nevin Oruc
- Department of Gastroenterology, Ege University, Izmir, Turkey
| | - Ahmet Coker
- Department of Gastroenterology, Ege University, Izmir, Turkey
| |
Collapse
|
213
|
Yordanova A, Mayer K, Brossart P, Gonzalez-Carmona MA, Strassburg CP, Essler M, Ahmadzadehfar H. Safety of multiple repeated cycles of 177Lu-octreotate in patients with recurrent neuroendocrine tumour. Eur J Nucl Med Mol Imaging 2017; 44:1207-1214. [DOI: 10.1007/s00259-017-3652-1] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Accepted: 02/08/2017] [Indexed: 12/28/2022]
|
214
|
Khan M, Dirweesh A, Alvarez C, Conaway H, Moser R. Anal Neuroendocrine Tumor Masquerading as External Hemorrhoids: A Case Report. Gastroenterology Res 2017; 10:56-58. [PMID: 28270879 PMCID: PMC5330695 DOI: 10.14740/gr751w] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/23/2017] [Indexed: 12/24/2022] Open
Abstract
Neuroendocrine tumors in gastrointestinal (GI) tract are a rare source of GI malignancy with an estimated incidence of 2.5 - 5 per 100,000 people per year and the prevalence of 35 per 100,000. In the GI tract, they are located in decreasing order of frequency in appendix, ileum, rectum, stomach, and colon. Those found in the anal region represent just 1% of all malignancies of the anal canal. Their clinical presentation can be widely varying, sometimes being found incidentally with metastatic disease and an unknown primary source. We report a case of a 60-year-old male who presented with a 2-week history of intermittent bright red blood per rectum and anal pain. He was found to have a lesion in the perianal area which was subsequently diagnosed has a poorly differentiated large cell type neuroendocrine carcinoma (NEC) with hepatic metastasis.
Collapse
Affiliation(s)
- Muhammad Khan
- Department of Internal Medicine, Saint Francis Medical Center, Seton Hall University, Trenton, NJ, USA
| | - Ahmed Dirweesh
- Department of Internal Medicine, Saint Francis Medical Center, Seton Hall University, Trenton, NJ, USA
| | - Chikezie Alvarez
- Department of Internal Medicine, Saint Francis Medical Center, Seton Hall University, Trenton, NJ, USA
| | - Herbert Conaway
- Department of Internal Medicine, Saint Francis Medical Center, Seton Hall University, Trenton, NJ, USA
| | - Robert Moser
- Department of Pathology, Saint Francis Medical Center, Trenton, NJ, USA
| |
Collapse
|
215
|
Gantenbein N, Haybaeck J. Neuroendocrine Tumorigenesis. MECHANISMS OF MOLECULAR CARCINOGENESIS – VOLUME 2 2017:141-146. [DOI: 10.1007/978-3-319-53661-3_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
216
|
Spychalski M, Koptas W, Zelga P, Dziki A. Role of endoscopic submucosal dissection in treatment of rectal gastroenteropancreatic neuroendocrine neoplasms. PRZEGLAD GASTROENTEROLOGICZNY 2017; 12:17-21. [PMID: 28337231 PMCID: PMC5360662 DOI: 10.5114/pg.2016.64635] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 12/24/2015] [Indexed: 02/07/2023]
Abstract
INTRODUCTION A significant rise in incidence of rectal gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs) has been observed in the last decade. Most detected gastroenteropancreatic neuroendocrine tumors (GEP-NETs) are well differentiated and less than 2 cm in diameter. Endoscopic submucosal dissection (ESD) is a new method for endoscopic treatment of such tumors, difficult to resect by conventional endoscopic techniques and thus subject to surgical treatment. AIM To present the results of the endoscopic treatment of GEP NET tumors in the rectum using ESD in single academic center. MATERIAL AND METHODS From June 2013 to April 2014, 4 cases of GEP-NET in the rectum were treated by ESD in our center. Effectiveness of dissection, complications and tumor recurrence after 3 months of treatment were then retrospectively investigated. RESULTS The group contained 2 patients with primary rectal GEP-NET (1 male, 1 female; age range: 48-60 years) and 2 with scars after incomplete polypectomy of rectal GEP-NET (1 male, 1 female; 61-65 years). Primary rectal GEP-NET diameters were 0.6 cm and 1.5 cm. Scar resection specimen diameters were 0.7 cm and 1 cm. Mean resection time was 28 min. The en bloc resection rate was 100% (2 of 2) and the histologically complete resection was confirmed in both cases. No foci of neuroendocrine neoplasia were reported in dissected scars. No complications were observed. After 3 months, 3 patients underwent follow-up colonoscopy - no local recurrence was reported. CONCLUSIONS Endoscopic submucosal dissection of rectal GEP-NET should be recommended as a treatment of choice when dealing with lesions over 1 cm in diameter without invasion of the muscle layer. Due to technical difficulties, performing this procedure should be reserved for centers with appropriately trained endoscopic staff.
Collapse
Affiliation(s)
- Michał Spychalski
- Department of General and Colorectal Surgery, Medical University of Lodz, Lodz, Poland
| | - Włodzimierz Koptas
- Department of General and Colorectal Surgery, Medical University of Lodz, Lodz, Poland
| | - Piotr Zelga
- Department of General and Colorectal Surgery, Medical University of Lodz, Lodz, Poland
| | - Adam Dziki
- Department of General and Colorectal Surgery, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
217
|
Pavel ME, Becerra C, Grosch K, Cheung W, Hasskarl J, Yao JC. Effect of everolimus on the pharmacokinetics of octreotide long-acting repeatable in patients with advanced neuroendocrine tumors: An analysis of the randomized phase III RADIANT-2 trial. Clin Pharmacol Ther 2016; 101:462-468. [DOI: 10.1002/cpt.559] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 09/27/2016] [Accepted: 10/25/2016] [Indexed: 01/06/2023]
Affiliation(s)
- ME Pavel
- Department of Hepatology and Gastroenterology; Charité Universitätsmedizin Berlin, Campus Virchow Klinikum; Berlin Germany
| | - C Becerra
- Texas Oncology Charles A. Sammons Cancer Center at Baylor; Dallas Texas USA
| | - K Grosch
- Novartis Pharmaceuticals Corporation; Basel Switzerland
| | - W Cheung
- Novartis Pharmaceuticals Corporation; Florham Park New Jersey USA
| | - J Hasskarl
- Novartis Pharmaceuticals Corporation; Basel Switzerland
| | - JC Yao
- University of Texas MD Anderson Cancer Center; Houston Texas USA
| |
Collapse
|
218
|
Krug S, Boch M, Nimphius W, Gress TM, Michl P, Rinke A. Relevance of dihydropyrimidine-dehydrogenase and thymidylate-synthase in patients with pancreatic neuroendocrine neoplasms treated with 5-FU-based chemotherapy. Pancreatology 2016; 17:139-145. [PMID: 28027897 DOI: 10.1016/j.pan.2016.12.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 11/15/2016] [Accepted: 12/16/2016] [Indexed: 12/11/2022]
Abstract
BACKGROUND Chemotherapy with 5-FU and Streptozotocin (STZ) is recommended as first-line treatment in patients with metastatic pancreatic neuroendocrine neoplasms (PNEN). However, data about biomarkers involved in the 5-FU metabolism to predict response are still limited. OBJECTIVES Evaluation of clinicopathological features and potential predictive and prognostic markers of patients with PNEN treated with 5-FU based regimens. PATIENTS AND METHODS We retrospectively analyzed 41 patients with PNEN who were treated at the University Hospital Marburg between 2000 and 2013. Dihydropyrimidine-Dehydrogenase (DPD) and Thymidylate-Synthase (TS) expression was correlated with treatment response in 19 patients who had available tumour tissue and response data. The median overall survival (OS) and progression free survival (PFS) were calculated using Kaplan-Meier and Cox regression methods, respectively. RESULTS The median PFS in patients receiving 5-FU/STZ was 17 months with a median OS of 50 months. Objective response rate (ORR) and disease control rate (DCR) were 32% and 73%, respectively. Biochemical response (p = 0.005) and high DPD expression (p = 0.018) were predictive markers of response to 5-FU-based chemotherapy. Univariate analysis identified Ki-67 > 10%, no biochemical response, positive 5-HIAA levels and TS deficiency as independent risk factors for shorter PFS. Moreover, performance status (PS) ≥1 was an independent risk factors for impaired OS. CONCLUSIONS DPD expression and biochemical response represent promising predictive biomarkers for response to 5-FU based chemotherapy. Moreover, Ki-67, PS and TS are independent prognostic markers of OS and PFS in patients with PNEN.
Collapse
Affiliation(s)
- S Krug
- Department of Internal Medicine I, Martin Luther University, Halle (Saale), Germany; Department of Gastroenterology and Endocrinology, Philipps-University, Marburg, Germany
| | - M Boch
- Institute of Pathology, Philipps-University, Marburg, Germany
| | - W Nimphius
- Institute of Pathology, Philipps-University, Marburg, Germany
| | - T M Gress
- Department of Gastroenterology and Endocrinology, Philipps-University, Marburg, Germany
| | - P Michl
- Department of Internal Medicine I, Martin Luther University, Halle (Saale), Germany.
| | - A Rinke
- Department of Gastroenterology and Endocrinology, Philipps-University, Marburg, Germany
| |
Collapse
|
219
|
Faure M, Niccoli P, Autret A, Cavaglione G, Mineur L, Raoul JL. Systemic chemotherapy with FOLFOX in metastatic grade 1/2 neuroendocrine cancer. Mol Clin Oncol 2016; 6:44-48. [PMID: 28123727 PMCID: PMC5245060 DOI: 10.3892/mco.2016.1097] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 10/07/2016] [Indexed: 12/13/2022] Open
Abstract
Neuroendocrine tumors (NETs) comprise a heterogeneous group of malignancies with various clinical presentations and evolution. NETs are often diagnosed at a late stage, when they are already metastatic. Treatment is currently based on traditional chemotherapies, such as streptozocin, with serious side effects. The favorable toxicity profile of the combination of 5-fluorouracil with oxaliplatin, together with its significant antitumor activity in several gastrointestinal malignancies, led to the evaluation of its efficacy and tolerability in patients with advanced grade 1/2 (G1/G2) NETs. The endpoints of the study were tumor response (according to the Response Evaluation Criteria in Solid Tumors 1.1), overall survival (OS), progression-free survival (PFS) and symptom improvement. From January, 2013 to January, 2015, during our Regional Multidisciplinary Tumor Board dedicated to NETs (RENATEN network), FOLFOX was recommended for the treatment of metastatic NETs as first-line therapy or after failure of other therapies. The inclusion criteria were metastatic, well-differentiated G1/G2 NETs, progressing within the last 3 months. Cases with previous antitumor therapy were allowed. The patients received modified FOLFOX-6 and were assessed every 3 months by computed tomography or magnetic resonance imaging examinations. A total of 31 patients were included. The median follow-up was 20 months [95% confidence interval (CI): 15-27]. Nine patients (29%) exhibited a partial response, and 13 (41%) achieved stable disease; the disease control rate was 70%. A total of 9 patients exhibited disease progression. The control rate was 78% for pancreatic and 65% for extrapancreatic NETs. The median OS was not reached; the 1- and 2-year OS rates were 89 and 70%, respectively (Fig. 1). No significant difference in OS was observed between the <5 and 5-20% Ki-67 subgroups (P=0.41) (Fig. 2A) or according to primary tumor location (P=0.71) (Fig. 2B). The median PFS was 14.1 months (95% CI: 9.3-24.1), with no significant difference in PFS between the Ki-67 subgroups (P=0.26) (Fig. 3A) or by primary tumor location (P=0.995) (Fig. 3B). The median time to treatment failure was 14.72 months (95% CI: 10.0-not estimable). No unusual toxicity or toxicity-related deaths were reported. Finally, 7 of 9 patients who achieved a partial response benefited from a break in treatment of ≥3 months. The median duration of this break was 9.2 months (range, 3-42 months). Of the 13 patients with stable disease, 12 may have also benefited from a chemotherapy break. The median break duration was 10 months (range, 0.5-26 months).
Collapse
Affiliation(s)
- Marjorie Faure
- Department of Medical Oncology, Paoli-Calmettes Institute, 13009 Marseille, France
| | - Patricia Niccoli
- Department of Medical Oncology, Paoli-Calmettes Institute, 13009 Marseille, France; Department of Endocrinology and Medical Oncology, La Timone Hospital, Faculty of Medicine, University of Mediterranée, 13005 Marseille, France
| | - Aurelie Autret
- Biostatistics Department, Paoli-Calmettes Institute, 13009 Marseille, France
| | - Gerard Cavaglione
- Medical Oncology Department, Antoine Lacassagne Cancer Center, 06100 Nice, France
| | - Laurent Mineur
- Medical Oncology Department, Sainte-Catherine Institute, 84000 Avignon, France
| | - Jean-Luc Raoul
- Department of Medical Oncology, Paoli-Calmettes Institute, 13009 Marseille, France
| |
Collapse
|
220
|
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: 75] [Impact Index Per Article: 8.3] [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
|
221
|
Basu S, Ostwal V. The case for combined chemotherapy-peptide receptor radionuclide therapy (chemo-PRRT) strategy in metastatic neuroendocrine tumor: predicting and looking at the possible case scenarios. Eur J Nucl Med Mol Imaging 2016; 43:2453-2455. [PMID: 27542673 DOI: 10.1007/s00259-016-3497-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 08/15/2016] [Indexed: 11/27/2022]
Affiliation(s)
- Sandip Basu
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Hospital Annexe, Jerbai Wadia Road Parel, Mumbai, 400 012, India.
| | - Vikas Ostwal
- Department of Medical Oncology, Tata Memorial Hospital, Mumbai, India
| |
Collapse
|
222
|
Sevilla I, Segura Á, Capdevila J, López C, García-Carbonero R, Grande E, On behalf of GETNE (Spanish Group of NeuroEndocrine Tumors). Management of controversial gastroenteropancreatic neuroendocrine tumour clinical situations with somatostatin analogues: results of a Delphi questionnaire panel from the NETPraxis program. BMC Cancer 2016; 16:858. [PMID: 27821081 PMCID: PMC5100262 DOI: 10.1186/s12885-016-2901-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 10/30/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND There are clinical situations (CS) in which the use of somatostatin analogs (SSAs) in patients with neuroendocrine tumors (NET) is controversial due to lack of evidence. A Delphi study was conducted to develop common treatment guidelines for these CS, based on clinical practice and expert opinion of Spanish oncologists. METHODS A scientific committee identified 5 CS with a common core (c-c) [non-functioning NET, not susceptible of surgery/locoregional therapy, Ki67 < 10 % (except for CS5: >10 %), ECOG ≤ 2], and controversy regarding use of SSAs, and prepared a Delphi questionnaire of 48 treatment statements. Statements were rated on a 1 (completely disagree) to 9 (completely agree) scale. Responses were grouped by tertiles: 1-3: Disagreement, 4-6: Neutral, 7-9: Agreement. Consensus was reached when the responses of ≥2/3 participants were located in the same tertile as the median value of all reported responses for that statement. RESULTS Sixty five (81.2 %) of 80 invited oncologists with experience in the management of NETs answered a first round of the questionnaire and 57 (87.7 %) of those 65 answered a second round (mean age 43.5 years; 53.8 % women; median time of experience 9 years). Consensus was obtained in 42 (36 agreement and 6 disagreement) of the 48 statements (87.5 %). Regarding CS1 (Enteropancreatic NET, c-c, non-progressive in the last 3-6 months), overall, SSA treatment is recommended (a wait and see approach is anecdotal and reserved for fragile patients or with low tumor load or ki-67 < 2 %); CS2 (Pancreatic NET, c-c), overall, SSA monotherapy is recommended, except when high tumor load or tumor progression exists, where combination therapy would be considered; CS3 [Gastroenteropancreatic (GEP)-NET, c-c, in treatment with anti-proliferative dose of SSA and progressing], overall, SSA maintenance is recommended at the time of progression, with or without adding molecular targeted drugs; CS4 (GEP-NET, c-c, and negative octreoscan®), SSA in monotherapy is only considered in low-risk patients (low tumor load and Ki-67 < 5 %); CS5 [GEP-NET, c-c (ki67 > 10 %), and positive octreoscan®], monotherapy with SSA is mainly considered in patients with comorbidities. CONCLUSION Several recommendations regarding use of SSAs in controversial NET CS were reached in consensus and might be considered as treatment guideline.
Collapse
Affiliation(s)
- Isabel Sevilla
- Oncology Unit. Hospital Clínico y Regional de Málaga, Colonia Santa Inés s/n, Málaga, 29010 Spain
| | - Ángel Segura
- Oncology Unit. Hospital Universitario La Fe, Avda. de Fernando Abril Martorell 106, 46026 Valencia, Spain
| | - Jaume Capdevila
- Oncology Unit. Hospital Vall d’Hebron, Pg de la Vall d’Hebron 119-129, 08035 Barcelona, Spain
| | - Carlos López
- Oncology Unit. Hospital Marqués de Valdecilla, Avda. Valdecilla 25, 39008 Santander, Spain
| | - Rocío García-Carbonero
- Oncology Unit. Hospital Universitario 12 de Octubre, Avda. de Córdoba s/n, 28041 Madrid, Spain
| | - Enrique Grande
- Oncology Unit. Hospital Universitario Ramón y Cajal, Ctra. de Colmenar Viejo km. 9.100, 28034 Madrid, Spain
| | - On behalf of GETNE (Spanish Group of NeuroEndocrine Tumors)
- Oncology Unit. Hospital Clínico y Regional de Málaga, Colonia Santa Inés s/n, Málaga, 29010 Spain
- Oncology Unit. Hospital Universitario La Fe, Avda. de Fernando Abril Martorell 106, 46026 Valencia, Spain
- Oncology Unit. Hospital Vall d’Hebron, Pg de la Vall d’Hebron 119-129, 08035 Barcelona, Spain
- Oncology Unit. Hospital Marqués de Valdecilla, Avda. Valdecilla 25, 39008 Santander, Spain
- Oncology Unit. Hospital Universitario 12 de Octubre, Avda. de Córdoba s/n, 28041 Madrid, Spain
- Oncology Unit. Hospital Universitario Ramón y Cajal, Ctra. de Colmenar Viejo km. 9.100, 28034 Madrid, Spain
| |
Collapse
|
223
|
Jin K, Xu J, Chen J, Chen M, Chen R, Chen Y, Chen Z, Cheng B, Chi Y, Feng ST, Fu D, Hou B, Huang D, Huang H, Huang Q, Li J, Li Y, Liang H, Lin R, Liu A, Liu J, Liu X, Lu M, Luo J, Mai G, Ni Q, Qiu M, Shao C, Shen B, Sheng W, Sun J, Tan C, Tan H, Tang Q, Tang Y, Tian X, Tong D, Wang X, Wang J, Wang J, Wang W, Wang W, Wang Y, Wu Z, Xue L, Yan Q, Yang N, Yang Y, Yang Z, Yin X, Yuan C, Zeng S, Zhang R, Yu X. Surgical management for non-functional pancreatic neuroendocrine neoplasms with synchronous liver metastasis: A consensus from the Chinese Study Group for Neuroendocrine Tumors (CSNET). Int J Oncol 2016; 49:1991-2000. [PMID: 27826620 DOI: 10.3892/ijo.2016.3711] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Accepted: 09/19/2016] [Indexed: 02/06/2023] Open
Abstract
Pancreatic neuroendocrine neoplasms (p-NENs) are slowly growing tumors with frequent liver metastasis. There is a variety of approaches to treat non-functional p-NENs with synchronous liver metastasis (LM) which complicates the determination of optimal treatment. Based on updated literature review, we discussed the treatment strategy determinants for p-NEN with LM. According to the resectability of primary tumor, the WHO 2010 grade classification and the radiological type of liver metastasis, the CSNET group reached agreements on a number of issues, including the following. Prior to treatment, biopsy is required to confirm pathology. Liver biopsy is important for more accurate grading of tumor and percutaneous core needle biopsy is more available than EUS-FNA. In patients with unresectable primary, surgical resection for liver-metastatic lesions should be avoided. Curative surgery is recommended for G1/G2 p-NET with type I LM and R1 resection also seems to improve overall survival rate. Cytoreductive surgery is recommended for G1/G2 p-NET with type II LM in select patients, and should meet stated requirements. Surgical resection for G1/G2 p-NET with type III LM and p-NEC with LM should be avoided, and insufficient evidence exists to guide the surgical treatment of G3 p-NET with LM. Liver transplantation may be an option in highly select patients. In addition, the optimal time for surgical approach is still required for more evidence.
Collapse
Affiliation(s)
- Kaizhou Jin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center; Pancreatic Cancer Institute, Fudan University, Shanghai, P.R. China
| | - Jin Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center; Pancreatic Cancer Institute, Fudan University, Shanghai, P.R. China
| | - Jie Chen
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Minhu Chen
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Rufu Chen
- Department of Biliary-Pancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Ye Chen
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, P.R. China
| | - Zhiyu Chen
- Department of Medical Oncology, Shanghai Cancer Center, Fudan University, Shanghai, P.R. China
| | - Bin Cheng
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China
| | - Yihebali Chi
- Department of Medical Oncology, Cancer Institute and Hospital, Chinese Academy of Medical Sciences, Beijing, P.R. China
| | - Shi-Ting Feng
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Deliang Fu
- Department of Pancreatic Surgery, Huashan Hospital, Fudan University, Shanghai, P.R. China
| | - Baohua Hou
- Department of General Surgery, Guangdong General Hospital, Guangzhou, Guangdong, P.R. China
| | - Dan Huang
- Department of Pathology, Shanghai Cancer Center, Fudan University, Shanghai, P.R. China
| | - Heguang Huang
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, P.R. China
| | - Qiang Huang
- Department of General Surgery, Anhui Provincial Hospital, Hefei, Anhui, P.R. China
| | - Jie Li
- Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Beijing Cancer Hospital, Beijing, P.R. China
| | - Ying Li
- Department of Radiology, Peking University Cancer Hospital, Beijing, P.R. China
| | - Houjie Liang
- Department of Oncology, Southwest Hospital, Third Military Medical University, Chongqing, P.R. China
| | - Rong Lin
- Department of Gastroenterology, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China
| | - An'an Liu
- Department of General Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, P.R. China
| | - Jixi Liu
- Department of Gastroenterology, China-Japan Friendship Hospital, Beijing, P.R. China
| | - Xubao Liu
- Department of Hepatobiliopancreatic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Ming Lu
- Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Beijing Cancer Hospital, Beijing, P.R. China
| | - Jie Luo
- Department of Pathology, China-Japan Friendship Hospital, Beijing, P.R. China
| | - Gang Mai
- Department of Hepatobiliopancreatic Surgery, The People's Hospital of Deyang, Chengdu, Sichuan, P.R. China
| | - Quanxing Ni
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center; Pancreatic Cancer Institute, Fudan University, Shanghai, P.R. China
| | - Meng Qiu
- Department of Abdominal Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Chenghao Shao
- Department of General Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, P.R. China
| | - Baiyong Shen
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Weiqi Sheng
- Department of Pathology, Shanghai Cancer Center, Fudan University, Shanghai, P.R. China
| | - Jian Sun
- Department of Biliary-Pancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Chunlu Tan
- Department of Hepatobiliopancreatic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Huangying Tan
- Department of Integrative Oncology, China-Japan Friendship Hospital, Beijing, P.R. China
| | - Qiyun Tang
- Department of Gastroenterology, Jiangsu People's Hospital, Nanjing, Jiangsu, P.R. China
| | - Yingmei Tang
- Department of Gastroenterology, The Second Affliated Hospital of Kunming Medical University, Yunnan Research Center for Liver Diseases, Kunming, Yunnan, P.R. China
| | - Xiaodong Tian
- Department of General Surgery, Peking University First Hospital, Beijing, P.R. China
| | - Danian Tong
- Department of Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, P.R. China
| | - Xiaohong Wang
- Department of Radiology, Shanghai Cancer Center, Fudan University, Shanghai, P.R. China
| | - Jian Wang
- Department of Biliary-Pancreatic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Jie Wang
- Department of Hepatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, P.R. China
| | - Wei Wang
- Department of Surgery, Huadong Hospital, Fudan University, Shanghai, P.R. China
| | - Wei Wang
- Department of Gastric and Pancreatic Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, P.R. China
| | - Yu Wang
- Department of Interventional Oncology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou Guangdong, P.R. China
| | - Zheng Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, P.R. China
| | - Ling Xue
- Department of Pathology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, P.R. China
| | - Qiang Yan
- Department of Hepatobiliopancreatic Surgery, Huzhou Central Hospital, Huzhou, Zhejiang, P.R. China
| | - Ning Yang
- Hepatobiliary Surgery Department V, Eastern Hepatobiliary Surgery Hospital, Shanghai, P.R. China
| | - Yinmo Yang
- Department of General Surgery, Peking University First Hospital, Beijing, P.R. China
| | - Zhiying Yang
- Department of Hepatobiliary Surgery, China-Japan Friendship Hospital, Beijing, P.R. China
| | - Xiaoyi Yin
- Department of Biliary-Pancreatic Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, P.R. China
| | - Chunhui Yuan
- Department of General Surgery, Peking University Third Hospital, Beijing, P.R. China
| | - Shan Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Renchao Zhang
- Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, P.R. China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center; Pancreatic Cancer Institute, Fudan University; Shanghai, P.R. China
| |
Collapse
|
224
|
Flaum N, Valle JW, Mansoor W, McNamara MG. Everolimus in the treatment of neuroendocrine tumors of the respiratory and gastroenteropancreatic systems. Future Oncol 2016; 12:2561-2578. [DOI: 10.2217/fon.16.23] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Neuroendocrine tumors (NETs) are a rare diverse group of malignancies occurring most commonly in the gastroenteropancreatic system and the lungs. The incidence of NETs is increasing worldwide; median survival for patients with metastatic NETs is 5–65 months. A growing body of evidence shows survival benefit in patients with advanced NETs (gastroenteropancreatic and lung) treated with mTOR inhibitor everolimus, with improvement in survival being demonstrated in the clinical trial and real-world setting. Everolimus has been shown to have a manageable safety profile, with the most common adverse events being stomatitis, rash, diarrhea, fatigue and infections. Due to the rarity of the condition, there are challenges in conducting clinical trials in these patients. Further research is required to clarify the role of adjuvant therapy, treatment sequencing and the use of multimodality treatments.
Collapse
Affiliation(s)
- Nicola Flaum
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Juan W Valle
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
- Institute of Cancer Sciences, University of Manchester, Manchester, UK
| | - Wasat Mansoor
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Mairéad G McNamara
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
- Institute of Cancer Sciences, University of Manchester, Manchester, UK
| |
Collapse
|
225
|
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
|
226
|
Jakoby J, Beuschlein F, Mentz S, Hantel C, Süss R. Liposomal doxorubicin for active targeting: surface modification of the nanocarrier evaluated in vitro and in vivo: challenges and prospects. Oncotarget 2016; 6:43698-711. [PMID: 26497207 PMCID: PMC4791260 DOI: 10.18632/oncotarget.6191] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 10/06/2015] [Indexed: 11/29/2022] Open
Abstract
Due to the inability of classical chemotherapeutic agents to exclusively target tumor cells, these treatments are associated with severe toxicity profiles. Thus, long-circulating liposomes have been developed in the past to enhance accumulation in tumor tissue by passive targeting. Accordingly, commercially available liposomal formulations of sterically stabilized liposomal doxorubicin (Caelyx®, Doxil®, Lipodox®) are associated with improved off-target profiles. However, these preparations are still not capable to selectively bind to target cells. Thus, in an attempt to further optimize existing treatment schemes immunoliposomes have been established to enable active targeting of tumor tissues. Recently, we have provided evidence for therapeutic efficacy of anti-IGF1R-targeted, surface modified doxorubicin loaded liposomes. Our approach involved a technique, which allows specific post-modifications of the liposomal surface by primed antibody-anchor conjugates thereby facilitating personalized approaches of commercially available liposomal drugs. In the current study, post-modification of sterically stabilized liposomal Dox was thoroughly investigated including the influence of different modification techniques (PIT, SPIT, SPIT60), lipid composition (SPC/Chol, HSPC/Chol), and buffers (HBS, SH). As earlier in vivo experiments did not take into account the presence of non-integrated ab-anchor conjugates this was included in the present study. Our experiments provide evidence that post-modification of commercially available liposomal preparations for active targeting is possible. Moreover, lyophilisation represents an applicable method to obtain a storable precursor of surface modifying antibody-anchor conjugates. Thus, these findings open up new approaches in patient individualized targeting of chemotherapeutic therapies.
Collapse
Affiliation(s)
- Judith Jakoby
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical Technology and Biopharmacy, Albert Ludwig University Freiburg, Freiburg, Germany
| | - Felix Beuschlein
- Endocrine Research Unit, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Susanne Mentz
- Endocrine Research Unit, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Constanze Hantel
- Endocrine Research Unit, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Regine Süss
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical Technology and Biopharmacy, Albert Ludwig University Freiburg, Freiburg, Germany
| |
Collapse
|
227
|
Lee MS, O'Neil BH. Summary of emerging personalized medicine in neuroendocrine tumors: are we on track? J Gastrointest Oncol 2016; 7:804-818. [PMID: 27747094 DOI: 10.21037/jgo.2016.08.05] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Neuroendocrine tumors (NETs) comprise a heterogeneous group of malignancies, with differences in prognosis and effective therapies. Traditionally, NETs have been characterized by tumor grade, site of primary tumor, functional status, and presence of underlying familial syndrome. However, increased feasibility and utilization of next-generation sequencing and other methodologies have revealed new genomic and epigenetic aberrations. In the last decade, treatment options available for metastatic well-differentiated gastroenteropancreatic (GEP) NETs have expanded, with approval of antiangiogenic and mTOR-directed targeted therapies, and our armamentarium of active therapies is likely to further increase. Cytotoxic therapies also are an important option for pancreatic NETs, and MGMT promoter methylation and protein expression may be an important biomarker for efficacy of alkylating agents. Peptide receptor radioligand therapy is an emerging treatment that uses functional imaging to personalize dosimetry to the tumor and avoid nephrotoxicity. Nevertheless, there is a critical need for further biomarkers, particularly multianalyte biomarkers, to aid in prognostication and predict efficacy of therapies.
Collapse
Affiliation(s)
- Michael S Lee
- University of North Carolina at Chapel Hill, North Carolina, USA
| | | |
Collapse
|
228
|
Latteyer S, Tiedje V, Schilling B, Führer D. Perspectives for immunotherapy in endocrine cancer. Endocr Relat Cancer 2016; 23:R469-84. [PMID: 27485460 DOI: 10.1530/erc-16-0169] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 08/02/2016] [Indexed: 12/16/2022]
Abstract
The fight against cancer has seen major breakthroughs in recent years. More than a decade ago, tyrosine kinase inhibitors targeting constitutively activated signaling cascades within the tumor inaugurated a new era of oncological therapy. Recently, immunotherapy with immune checkpoint inhibitors has started to revolutionize the treatment of several malignancies, most notably malignant melanoma, leading to the renaissance and the long-awaited breakthrough of immunooncology. This review provides an overview of the basis of immunotherapy from its initial concepts of anti-tumor immunity and cell-based therapy to the development of immune checkpoint inhibitors and discusses published studies and the perspectives of immunooncology for the treatment of endocrine malignancies.
Collapse
Affiliation(s)
- S Latteyer
- Department of Endocrinology and MetabolismUniversity Hospital Essen, University of Duisburg-Essen, Essen, Germany Endocrine Tumour Center at West German Cancer Center (WTZ)Essen, Germany
| | - V Tiedje
- Department of Endocrinology and MetabolismUniversity Hospital Essen, University of Duisburg-Essen, Essen, Germany Endocrine Tumour Center at West German Cancer Center (WTZ)Essen, Germany
| | - B Schilling
- Department of DermatologyVenereology and Allergology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany German Cancer Consortium (DKTK)Heidelberg, Germany
| | - D Führer
- Department of Endocrinology and MetabolismUniversity Hospital Essen, University of Duisburg-Essen, Essen, Germany Endocrine Tumour Center at West German Cancer Center (WTZ)Essen, Germany
| |
Collapse
|
229
|
Imaging approaches to assess the therapeutic response of gastroenteropancreatic neuroendocrine tumors (GEP-NETs): current perspectives and future trends of an exciting field in development. Cancer Metastasis Rev 2016; 34:823-42. [PMID: 26433592 PMCID: PMC4661203 DOI: 10.1007/s10555-015-9598-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [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
Gastroenteropancreatic neuroendocrine tumors (GEP-NETs) are a family of neoplasms with a complex spectrum of clinical behavior. Although generally more indolent than carcinomas, once they progress beyond surgical resectability, they are essentially incurable. Systemic treatment options have substantially expanded in recent years for the management of advanced disease. Imaging plays a major role in new drug development, as it is the main tool used to objectively evaluate response to novel agents. However, current standard response criteria have proven suboptimal for the assessment of the antiproliferative effect of many targeted agents, particularly in the context of slow-growing tumors such as well-differentiated NETs. The aims of this article are to discuss the advantages and limitations of conventional radiological techniques and standard response assessment criteria and to review novel imaging modalities in development as well as alternative cancer- and therapy-specific criteria to assess drug efficacy in the field of GEP-NETs.
Collapse
|
230
|
Narayanan S, Kunz PL. Role of Somatostatin Analogues in the Treatment of Neuroendocrine Tumors. Hematol Oncol Clin North Am 2016; 30:163-77. [PMID: 26614375 DOI: 10.1016/j.hoc.2015.09.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Neuroendocrine tumors (NETs) are rare epithelial neoplasms with neuroendocrine differentiation originating most commonly in the lungs and gastroenteropancreatic. Treatment includes surgery and other local therapies; treatment of inoperable disease centers around symptom management and control of tumor growth. Somatostatin analogues (SSAs) have been a mainstay of managing hormone-related symptoms. Emerging evidence suggests that they are effective therapies for tumor control also. Peptide receptor radionuclide therapy with radiolabeled SSAs is a new, promising treatment for inoperable or metastatic NETs. This article reviews the role of SSAs in the treatment of NETs.
Collapse
Affiliation(s)
- Sujata Narayanan
- Medicine/Oncology, Stanford University School of Medicine, 875 Blake Wilbur Drive, Stanford, CA 94305-5826, USA
| | - Pamela L Kunz
- Medicine/Oncology, Stanford University School of Medicine, 875 Blake Wilbur Drive, Stanford, CA 94305-5826, USA.
| |
Collapse
|
231
|
|
232
|
Heikkonen J, Mäenpää H, Hippeläinen E, Reijonen V, Tenhunen M. Effect of calculation method on kidney dosimetry in 177Lu-octreotate treatment. Acta Oncol 2016; 55:1069-1076. [PMID: 27219529 DOI: 10.1080/0284186x.2016.1182642] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND 177Lu-octreotate is an effective treatment modality for patients with metastatic neuroendocrine tumors. The kidney is a critical dose-limiting organ in that modality. We investigated the absorbed doses in the kidney and compared whole kidney volume (WKV) and small (4 cm3) volume of the kidney (SV) methods. We also evaluated a new calculation method that was based on two single photon emission computed tomography/computed tomography (SPECT/CT) scans. METHODS Absorbed radiation doses in the kidneys were calculated for 24 patients with neuroendocrine tumors. All patients received four cycles of 177Lu-octreotate given at eight-week intervals with a mean activity of 7.1 GBq (range 3.28-8.79 GBq). Absorbed doses and half-lives were calculated by the WKV and SV methods. Dosimetry was determined for the cortex and medulla in the first treatment cycle. RESULTS The mean absorbed radiation dose was 0.44 ± 0.15 Gy/GBq for the WKV method and, 0.74 ± 0.28 Gy/GBq for the SV method. Three patients had a 20% increase of the absorbed dose over the four treatment cycles for the WKV method compared to eight patients for the SV method. The mean absorbed dose in the medulla was 0.62 ± 0.27 Gy/GBq, whereas the mean absorbed dose in the cortex was 0.41 ± 0.22 Gy/GBq. Both regions had similar half-lives. Patients who received lower activities for medical reasons still had similar absorbed doses to kidneys compared to those who received the full activities. Our study indicates that absorbed doses can be calculated reliably using two SPECT/CT scans, at 24 and 168 hours after each treatment. CONCLUSIONS Absorbed doses in the kidneys from systemic radionuclide therapy that are measured by the WKV method and SV method cannot be directly compared. There were regional differences within kidneys for the uptake of 177Lu-octreotate. Two SPECT/CTs are sufficient for kidney dosimetry based on our new calculation method.
Collapse
Affiliation(s)
- Jorma Heikkonen
- Nuclear Medicine, Helsinki University Central Hospital, Cancer Center, Helsinki, Finland
| | - Hanna Mäenpää
- Nuclear Medicine, Helsinki University Central Hospital, Cancer Center, Helsinki, Finland
| | - Eero Hippeläinen
- HUS Medical Imaging Center, Helsinki University Central Hospital, Helsinki, Finland
| | - Vappu Reijonen
- Nuclear Medicine, Helsinki University Central Hospital, Cancer Center, Helsinki, Finland
| | - Mikko Tenhunen
- Department of Radiotherapy, Helsinki University Central Hospital, Cancer Center, Helsinki, Finland
| |
Collapse
|
233
|
Paul D, Ostwal V, Bose S, Basu S, Gupta S. Personalized treatment approach to gastroenteropancreatic neuroendocrine tumors: a medical oncologist's perspective. Eur J Gastroenterol Hepatol 2016; 28:985-990. [PMID: 27257869 DOI: 10.1097/meg.0000000000000674] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
The medical management of gastroenteropancreatic neuroendocrine tumors involves treatment of symptomatic disease related to hormone secretions or bulky unresectable metastatic disease. Combining gallium DOTA with fluorine-18 fluorodeoxyglucose-PET along with histopathological grading helps to determine tumor heterogeneity and seek reasons for poor response to therapy. In the light of adding chemotherapy in selected patients with intermediate-grade tumors, the newer scan helps in personalization of the therapy along with the biopsy. The tumor dedifferentiation over the particular time period leading to aggressive behavior, a well-known entity, is contrasted with the redifferentiation phenomenon in some patients as a result of chemotherapy or targeted drug therapy. This may support the basis for combining peptide receptor-targeted radiotherapy/octreotide therapy with chemotherapy or mTOR inhibitors such as everolimus.
Collapse
Affiliation(s)
- Davinder Paul
- aDepartment of Medical Oncology, Tata Memorial Hospital bRadiation Medicine Centre, Mumbai, India
| | | | | | | | | |
Collapse
|
234
|
Cives M, Ghayouri M, Morse B, Brelsford M, Black M, Rizzo A, Meeker A, Strosberg J. Analysis of potential response predictors to capecitabine/temozolomide in metastatic pancreatic neuroendocrine tumors. Endocr Relat Cancer 2016; 23:759-67. [PMID: 27552969 DOI: 10.1530/erc-16-0147] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 07/21/2016] [Indexed: 12/25/2022]
Abstract
The capecitabine and temozolomide (CAPTEM) regimen is active in the treatment of metastatic pancreatic neuroendocrine tumors (pNETs), with response rates ranging from 30 to 70%. Small retrospective studies suggest that O(6)-methylguanine DNA methyltransferase (MGMT) deficiency predicts response to temozolomide. High tumor proliferative activity is also commonly perceived as a significant predictor of response to cytotoxic chemotherapy. It is unclear whether chromosomal instability (CIN), which correlates with alternative lengthening of telomeres (ALT), is a predictive factor. In this study, we evaluated 143 patients with advanced pNET who underwent treatment with CAPTEM for radiographic and biochemical response. MGMT expression (n=52), grade (n=128) and ALT activation (n=46) were investigated as potential predictive biomarkers. Treatment with CAPTEM was associated with an overall response rate (ORR) of 54% by RECIST 1.1. Response to CAPTEM was not influenced by MGMT expression, proliferative activity or ALT pathway activation. Based on these results, no biomarker-driven selection criteria for use of the CAPTEM regimen can be recommended at this time.
Collapse
Affiliation(s)
- M Cives
- Department of Gastrointestinal OncologyH. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - M Ghayouri
- Department of Gastrointestinal OncologyH. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - B Morse
- Department of Gastrointestinal OncologyH. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - M Brelsford
- Department of Gastrointestinal OncologyH. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - M Black
- Department of Gastrointestinal OncologyH. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - A Rizzo
- Department of PathologyJohns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - A Meeker
- Department of PathologyJohns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - J Strosberg
- Department of Gastrointestinal OncologyH. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| |
Collapse
|
235
|
Dumars C, Foubert F, Touchefeu Y, Regenet N, Senellart H, Matysiak-Budnik T, Heymann MF. Can PPH3 be helpful to assess the discordant grade in primary and metastatic enteropancreatic neuroendocrine tumors? Endocrine 2016; 53:395-401. [PMID: 27048356 DOI: 10.1007/s12020-016-0944-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 03/25/2016] [Indexed: 02/06/2023]
Abstract
Therapeutic strategy in neuroendocrine tumors (NETs) is based on histological characteristics of the primary tumor (PT), even in case of metastatic disease. Our aim was to compare the tumor grade between PT and their liver metastases (LM) in patients with enteropancreatic NETs. Forty-one patients treated for sporadic NETs (10 pancreatic, 31 intestinal) were included. All presented synchronous (35) or metachronous (6) LM. Tumor grade was evaluated for PT and LM according to the WHO classification, using Ki-67 labeling and mitotic count (MC) evaluated with or without phospho-histone H3 (PPH3). Tumor grade differed between primary and metastatic tumor in 16/41 patients (39 %), with an increase of grade in 13 of them (32 %). The median Ki-67, MC, and PPH3 in metastases were statistically higher than in PT (p = 0.0002, 0.02, and 0.01). In 17 of 65 cases tested with PPH3 (26 %), this antibody was more efficient in assessing the grade compared to the usual MC, and in 2/65 cases compared to the Ki-67. A better correlation was observed between Ki-67 and PPH3 (p = 0.0001) than between Ki-67 and MC without immunohistochemistry. There is a significant difference in tumor grade between primary and metastatic NETs, underlining the necessity of a systematic biopsy on LM for patient management. Moreover, PPH3 appears to be a powerful antibody to assess the MC and the tumor grade much more accurately when associated with Ki-67.
Collapse
Affiliation(s)
- Clotilde Dumars
- Department of Anatomopathology A, Nantes University Hospital, Nantes, France
- INSERM, UMR U957, Nantes University, Nantes, France
| | - Fanny Foubert
- IMAD, Department of Gastroenterology & Digestive Oncology, Nantes University Hospital, Nantes, France
| | - Yann Touchefeu
- IMAD, Department of Gastroenterology & Digestive Oncology, Nantes University Hospital, Nantes, France
- INSERM, UMR 892, Nantes University, Nantes, France
| | - Nicolas Regenet
- Department of Digestive Surgery, Nantes University Hospital, Nantes, France
| | | | - Tamara Matysiak-Budnik
- IMAD, Department of Gastroenterology & Digestive Oncology, Nantes University Hospital, Nantes, France
- INSERM, UMR 913, Nantes University, Nantes, France
| | - Marie-Françoise Heymann
- Department of Anatomopathology A, Nantes University Hospital, Nantes, France.
- INSERM, UMR U957, Nantes University, Nantes, France.
- Department of Oncology and Metabolism, The University of Sheffield, Medical School (Room FU 32), Beech Hill Road, Sheffield, S10 2RX, UK.
| |
Collapse
|
236
|
Elevated Serum Pancreastatin Is an Indicator of Hepatic Metastasis in Patients With Small Bowel Neuroendocrine Tumors. Pancreas 2016; 45:1032-5. [PMID: 26684860 DOI: 10.1097/mpa.0000000000000572] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVES Serum pancreastatin is a sensitive and specific diagnostic biomarker in neuroendocrine tumors (NETs). Elevated pancreastatin levels are associated with worse progression-free survival and overall survival in small bowel and pancreatic NETs. In this study, we investigated the clinical significance of elevated serum pancreastatin in identifying metastatic disease to the liver. METHODS Retrospective chart review of patients with NET managed at a single institution was performed. The site of primary tumor, laboratory data, and presence of metastatic disease were reviewed. The sensitivity, specificity, and positive and negative predictive values for pancreastatin as indicator of liver metastasis were ascertained. RESULTS Data were abstracted from 77 patient records. Small bowel was the primary tumor site in 44 patients (57%), and 49 patients had metastasis to the liver (64%). Sensitivity and specificity of serum pancreastatin was 85.7% and 66.7%, respectively, which compared with 61.5% and 43.8% for chromogranin A, in identifying liver metastasis in patients with primary tumors of the small bowel. CONCLUSIONS Elevated serum pancreastatin is a sensitive and specific assay for detecting the incidence of liver metastasis in patients with small-bowel NET. Routine measurement of pancreastatin in patients with NET, especially in patients with small bowel primaries, is supported.
Collapse
|
237
|
Cavalcanti MS, Gönen M, Klimstra DS. The ENETS/WHO grading system for neuroendocrine neoplasms of the gastroenteropancreatic system: a review of the current state, limitations and proposals for modifications. INTERNATIONAL JOURNAL OF ENDOCRINE ONCOLOGY 2016; 3:203-219. [PMID: 30338051 PMCID: PMC6190579 DOI: 10.2217/ije-2016-0006] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The understanding of neuroendocrine neoplasms has evolved significantly since their initial descriptions in the 1800s to early 1900s. In the gastroenteropancreatic system, this group of malignant tumors is subdivided into well and poorly differentiated neuroendocrine neoplasms based on morphologic, proliferative and biologic differences. However, it has become increasingly apparent that well-differentiated neuroendocrine tumors are not a homogeneous group. Attempting to better predict outcome of these tumors has been the motivation behind numerous proposed classification systems, the evolution of which culminated with the currently used system, the ENETS/WHO classification. Herein, we review the genesis of this classification system and some of its shortcomings. In addition, we discuss some of the most recent proposals that suggest modifications to the current system.
Collapse
Affiliation(s)
- Marcela S Cavalcanti
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mithat Gönen
- Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - David S Klimstra
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
238
|
Johnbeck CB, Knigge U, Langer SW, Loft A, Berthelsen AK, Federspiel B, Binderup T, Kjaer A. Prognostic Value of 18F-FLT PET in Patients with Neuroendocrine Neoplasms: A Prospective Head-to-Head Comparison with 18F-FDG PET and Ki-67 in 100 Patients. J Nucl Med 2016; 57:1851-1857. [DOI: 10.2967/jnumed.116.174714] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2016] [Accepted: 06/27/2016] [Indexed: 12/20/2022] Open
|
239
|
Calin ML, Sadiq A, Arevalo G, Fuentes R, Flanders VL, Gupta N, Nasri B, Singh K. The First Case Report of Robotic Multivisceral Resection for Synchronous Liver Metastasis from Pancreatic Neuroendocrine Tumor: A Case Report and Literature Review. J Laparoendosc Adv Surg Tech A 2016; 26:816-824. [PMID: 27454160 DOI: 10.1089/lap.2016.0342] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION Surgery for liver metastases in pancreatic neuroendocrine tumor (PNET) improves overall survival rate. We present the first case report for robotic multivisceral resection of distal pancreas, spleen, and left liver for metastatic PNET. MATERIALS AND METHODS We present a case of 52-year-old female diagnosed with PNET in the pancreatic neck metastatic to the liver, responding to somatostatin and bland embolization, who underwent surgical debulking using da Vinci robotic platform. Intraoperative Doppler ultrasound was used to define the vascular distribution and tumor extension. The parenchymal liver transection was performed with vessel sealer. The distal pancreas and the spleen were approached medial to lateral and resected in an en-bloc fashion. The left liver inflow, outflow, and splenic artery and vein were transected with vascular stapler device. RESULTS Da Vinci robot-assisted multivisceral resection has been performed with good postoperative outcome. Operative time was 369 minutes and the estimated blood loss was 100 mL. The patient had a short hospital stay with quick recovery and good outcome at 5 months follow-up after the surgery. DISCUSSION Liver metastases in PNETs are considered an adverse factor. Aggressive surgical management is a mainstay. The laparoscopic approach to pancreatic or hepatic surgery is difficult in inexperienced hands with steep learning curve. The recent robotic system seems to overcome many limitations. This is the first case of robotic multivisceral resection for synchronous liver metastasis from PNET. Concurrent primary tumor resection with hepatectomy offers potential curative intention.
Collapse
Affiliation(s)
- Marius L Calin
- 1 Department of General Surgery, Saint Vincent Hospital Indianapolis , Indianapolis, Indiana
| | - Aziz Sadiq
- 1 Department of General Surgery, Saint Vincent Hospital Indianapolis , Indianapolis, Indiana
| | - Gabriel Arevalo
- 1 Department of General Surgery, Saint Vincent Hospital Indianapolis , Indianapolis, Indiana
| | - Rocio Fuentes
- 1 Department of General Surgery, Saint Vincent Hospital Indianapolis , Indianapolis, Indiana
| | - Vincent L Flanders
- 2 Department of Interventional Radiology, Saint Vincent Hospital Indianapolis , Indianapolis, Indiana
| | - Niraj Gupta
- 3 Department of Oncology, Saint Vincent Hospital Indianapolis , Indianapolis, Indiana
| | - Baongoc Nasri
- 4 Department of Surgery, Tokyo Metropolitan Matsuzawa Hospital , Tokyo, Japan
| | - Kirpal Singh
- 1 Department of General Surgery, Saint Vincent Hospital Indianapolis , Indianapolis, Indiana
| |
Collapse
|
240
|
Grillo F, Albertelli M, Annunziata F, Boschetti M, Caff A, Pigozzi S, Ferone D, Mastracci L. Twenty years of gastroenteropancreatic neuroendocrine tumors: is reclassification worthwhile and feasible? Endocrine 2016; 53:58-62. [PMID: 26362059 DOI: 10.1007/s12020-015-0734-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 09/01/2015] [Indexed: 12/19/2022]
Abstract
Gastroenteropancreatic (GEP) neuroendocrine tumors (NETs) are rare neoplasms with heterogeneous clinical behavior and potential long-term survival. In 2006/2007, the European Neuroendocrine Tumors Society introduced an important parameter, grade (based on mitoses and Ki-67 proliferation rate), which became part of the latest 2010-WHO classification. Since this is an important tool in the choice of therapeutic algorithm of patients with NETs, our aim was to audit whether retrospective reclassification is possible and feasible and correlate pathological findings with survival. From the histopathology archive, 338 GEP-NETs (1994-2014) were identified, of which 250 were diagnosed pre-2010 and 80 of these have needed, up till now, classification (morphology and grade-mitotic count/Ki-67). Morphology was well differentiated (WD) in 74 cases while only 6 cases were poorly differentiated (PD). Grade was reclassified: G1-45 cases (56 %); G2-28 cases (35 %); G3-7 cases (9 %). Overall survival (OS) in WD NETs was strikingly better compared to PD neoplasms. Differences in OS between grade were statistically significant (p < 0.0001) and, in particular, grade identified a subgroup of patients with WD lesions but with less favorable clinical behavior (OS at 5 years: G1-89 %; G2-48 %; G3-0 %; G1 vs G2 p = 0.03). Feasibility analysis quantified time for reclassification to be between 45 and 64 min/case. Our series confirms the importance of grade in prognostic stratification and underlines that reclassification is feasible, and may prove worthwhile in patient management, especially in view of the potential long survival of patients with NETs and risk of use of inappropriate therapies.
Collapse
Affiliation(s)
- Federica Grillo
- Histopathology, Department of Surgical Science (DISC), University of Genoa, Genoa, Italy
- IRCCS AOU San Martino IST, Largo R. Benzi, 10, 16132, Genoa, Italy
| | - Manuela Albertelli
- Endocrinology, Department of Internal Medicine (DIMI), University of Genoa, Viale Benedetto XV, 6, 16132, Genoa, Italy.
| | - Francesca Annunziata
- Endocrinology, Department of Internal Medicine (DIMI), University of Genoa, Viale Benedetto XV, 6, 16132, Genoa, Italy
| | - Mara Boschetti
- IRCCS AOU San Martino IST, Largo R. Benzi, 10, 16132, Genoa, Italy
- Endocrinology, Department of Internal Medicine (DIMI), University of Genoa, Viale Benedetto XV, 6, 16132, Genoa, Italy
| | - Andrea Caff
- Endocrinology, Department of Internal Medicine (DIMI), University of Genoa, Viale Benedetto XV, 6, 16132, Genoa, Italy
- Endocrinology, Department of Clinical and Experimental Medicine, Garibaldi-Nesima Medical Center, University of Catania, Via Palermo 636, 95122, Catania, Italy
| | - Simona Pigozzi
- Histopathology, Department of Surgical Science (DISC), University of Genoa, Genoa, Italy
- IRCCS AOU San Martino IST, Largo R. Benzi, 10, 16132, Genoa, Italy
| | - Diego Ferone
- IRCCS AOU San Martino IST, Largo R. Benzi, 10, 16132, Genoa, Italy
- Endocrinology, Department of Internal Medicine (DIMI), University of Genoa, Viale Benedetto XV, 6, 16132, Genoa, Italy
| | - Luca Mastracci
- Histopathology, Department of Surgical Science (DISC), University of Genoa, Genoa, Italy
- IRCCS AOU San Martino IST, Largo R. Benzi, 10, 16132, Genoa, Italy
| |
Collapse
|
241
|
Evaluation of the concordance between the stage of the disease and Ki-67 proliferation index in gastroenteropancreatic neuroendocrine tumors. Eur J Gastroenterol Hepatol 2016; 28:836-41. [PMID: 26945127 DOI: 10.1097/meg.0000000000000619] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
AIM This study aimed to determine the degree of concordance between TNM staging used in the determination of the prognosis of gastroenteropancreatic neuroendocrine tumor (GEP-NET) patients and the Ki-67 proliferation index value used in the grading of these tumors and investigate the most reliable prognostic parameter among them. MATERIALS AND METHODS The medical files of the patients with GEP-NET who were diagnosed or followed up in Erciyes University Faculty of Medicine were retrospectively examined and demographic characteristics, survival times, grade of these tumors, histopathologically detected Ki-67 values, and histopathological characteristics were recorded and evaluated statistically. RESULTS The mean age (53.09±14.6 years; range, 16-85 years) of all (n=141) the patients was estimated. The patient population included 72 (51.1%) female and 69 (48.9%) male patients, with a male/female ratio of 0.95. The most frequently encountered primary sites were the stomach (33.3%), and then in decreasing oder of frequency the pancreas (27%), colon-rectum (15.6%), the small intestine (12.8%), and the appendix (11.3%). The GEP-NET of the patients was in grade 1 (G1) (n: 103; 73%), grade 2 (G2) (n: 24; 17%), and grade 3 (G3) (n: 14; 10%). The GEP-NET of the patients was stage I (n: 66; 46.8%), stage II (n: 14; 9.9%), stage III (n: 12; 8.5%), and stage IV (n: 49; 34.8%). In the statistical analysis, Ki-67 increased in parallel with the stage of the disease (P<0.001). As Ki-67 increased at a rate of 1%, survival rates of the patients decreased 1.027 times (P=0.01). Five-year survival rates of the patients were 88% in G1, 44% in G2, and 24% in G3. Patients in G2 and G3 had a 6.67 and 12.38 times lower chance of survival compared with G1 patients, respectively. Survival rates of stage IV patients were 5.6 times lower relative to stages I and II patients, respectively (P<0.001). The median 5-year survival rates of the patients were 90% in stage I, 100% in stage II, 47% in stage III, and 46% in stage IV. In univariate analysis, age of the patients, grade, stage of the tumor, and lymph node metastases were found to be parameters that affected overall survival, whereas no significant correlation was found between the sex of the patient and the primary organ from which the tumor originated and survival rates. However, in the multivariate analysis, survival rates decreased inversely with age, whereas no significant correlation was found between grade and stage of the tumor and survival rates. CONCLUSION In conclusion, a decrease in the average survival rate in parallel with an increase in the grade of the tumor was more prominent compared with a decrease in survival rates in accordance with an increase in the stage of the tumor. This indicates that in the prediction of prognosis in patients with GEP-NET, the Ki-67 value can be a more important evaluation factor relative to staging.
Collapse
|
242
|
Basu S, Ranade R, Ostwal V, Shrikhande SV. PET-Based Molecular Imaging in Designing Personalized Management Strategy in Gastroenteropancreatic Neuroendocrine Tumors. PET Clin 2016; 11:233-241. [PMID: 27321028 DOI: 10.1016/j.cpet.2016.02.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
In recent years, PET-based molecular functional imaging has been increasingly used in neuroendocrine tumors for tailoring of treatment strategies to the individual characteristics of each patient. For each particular patient, the relative tracer uptake by the dual-tracer PET imaging approach (with 68Ga-DOTANOC/TATE and 18F-FDG) frequently plays an important role along with the histopathologic tumor grades for selecting the optimal treatment approach for advanced/metastatic cases. Various tumor-specific parameters have resulted in development of such precision-medicine type model in this biologically heterogeneous group of tumors. The traditional advantages of PET/computed tomography in terms of disease staging are also applicable for personalization of management. From the medical oncologist's standpoint, multitracer PET-based information and staging is of significant importance (in addition to the histologic grades) in selecting the appropriate chemotherapy regimen and monitoring response on an individual basis in the course of treatment.
Collapse
Affiliation(s)
- Sandip Basu
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Hospital Annexe, Jerbai Wadia Road, Parel, Mumbai 400 012, India.
| | - Rohit Ranade
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Hospital Annexe, Jerbai Wadia Road, Parel, Mumbai 400 012, India
| | - Vikas Ostwal
- Department of Medical Oncology, Tata Memorial Hospital, Mumbai, India
| | - Shailesh V Shrikhande
- Gastrointestinal and Hepato-Pancreato-Biliary Service, Department of Surgical Oncology, Tata Memorial Hospital, Mumbai, India
| |
Collapse
|
243
|
Vinik AI, Wolin EM, Liyanage N, Gomez-Panzani E, Fisher GA. EVALUATION OF LANREOTIDE DEPOT/AUTOGEL EFFICACY AND SAFETY AS A CARCINOID SYNDROME TREATMENT (ELECT): A RANDOMIZED, DOUBLE-BLIND, PLACEBO-CONTROLLED TRIAL. Endocr Pract 2016; 22:1068-80. [PMID: 27214300 DOI: 10.4158/ep151172.or] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
OBJECTIVE To evaluate the efficacy and safety of lanreotide depot/autogel 120 mg for the control of carcinoid syndrome (CS) symptoms in patients with neuroendocrine tumors (NETs). METHODS This was a 16-week, randomized, double-blind, phase 3 trial (Clinicaltrials.gov: NCT00774930). Patients with/without prior somatostatin analog (SSA) use were randomized to lanreotide depot/autogel 120 mg or placebo every 4 weeks, with access to short-acting octreotide as rescue medication. The primary endpoint was the percentage of days in which short-acting octreotide was used, which was assessed from daily diaries using an analysis of covariance including the stratification variables baseline short-acting octreotide use and frequency of diarrhea/flushing. The proportions of patients experiencing treatment success was a supportive analysis. Adverse events were recorded at all visits. RESULTS A total of 115 patients were enrolled (lanreotide, n = 59; placebo, n = 56). The adjusted mean (95% confidence interval [CI]) percentage of days with rescue octreotide use (primary endpoint) was significantly lower in the lanreotide (33.7%; 95% CI, 25.0%-42.4%) versus the placebo group (48.5%; 95% CI, 39.6%-57.4%), representing an absolute difference of -14.8% (95% CI, -26.8% to -2.8%; P = .017). The odds ratio of full/partial treatment success (≤3 days short-acting octreotide use weeks 12 to 15) was significantly greater with lanreotide than placebo (2.4; 95% CI, 1.1-5.3; P = .036). No new safety concerns were identified, and lanreotide was well tolerated. CONCLUSION Lanreotide depot/autogel is effective for the control of CS symptoms in patients (SSA-naïve or experienced) with NETs. ABBREVIATIONS AE = adverse event BMI = body mass index CS = carcinoid syndrome ELECT = Evaluating Lanreotide Efficacy and safety as a Carcinoid-syndrome Treatment HRQoL = health-related quality of life LTOLE = long-term open-label extension NET = neuroendocrine tumor OL = open label SSA = somatostatin analog.
Collapse
|
244
|
Singh S, Asa SL, Dey C, Kennecke H, Laidley D, Law C, Asmis T, Chan D, Ezzat S, Goodwin R, Mete O, Pasieka J, Rivera J, Wong R, Segelov E, Rayson D. Diagnosis and management of gastrointestinal neuroendocrine tumors: An evidence-based Canadian consensus. Cancer Treat Rev 2016; 47:32-45. [PMID: 27236421 DOI: 10.1016/j.ctrv.2016.05.003] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 05/07/2016] [Indexed: 02/07/2023]
Abstract
The majority of neuroendocrine tumors originate in the digestive system and incidence is increasing within Canada and globally. Due to rapidly evolving evidence related to diagnosis and clinical management, updated guidance on the diagnosis and treatment of gastrointestinal neuroendocrine tumors (GI-NETs) are of clinical importance. Well-differentiated GI-NETs may exhibit indolent clinical behavior and are often metastatic at diagnosis. Some NET patients will develop secretory disease requiring symptom control to optimize quality of life and clinical outcomes. Optimal management of GI-NETs is in a multidisciplinary environment and is multimodal, requiring collaboration between medical, surgical, imaging and pathology specialties. Clinical application of advances in pathological classification and diagnostic technologies, along with evolving surgical, radiotherapeutic and medical therapies are critical to the advancement of patient care. We performed a systematic literature search to update our last set of published guidelines (2010) and identified new level 1 evidence for novel therapies, including telotristat etiprate (TELESTAR), lanreotide (CLARINET), everolimus (RADIANT-2; RADIANT-4) and peptide receptor radionuclide therapy (PRRT; NETTER-1). Integrating these data with the clinical knowledge of 16 multi-disciplinary experts, we devised consensus recommendations to guide state of the art clinical management of GI-NETs.
Collapse
Affiliation(s)
- Simron Singh
- Sunnybrook Health Sciences Centre, Department of Medicine, University of Toronto, 2075 Bayview Ave. Room T2-047, Toronto, Ontario M4N 3M5, Canada.
| | - Sylvia L Asa
- University Health Network, Department of Pathology, University of Toronto, Toronto, Ontario M5G 2C4, Canada.
| | - Chris Dey
- Sunnybrook Health Sciences Centre, Department of Medical Imaging, University of Toronto, 2075 Bayview Ave. Room MG-182, Toronto, Ontario M4N 3M5, Canada.
| | - Hagen Kennecke
- BC Cancer Agency, Division of Medical Oncology, University of British Columbia, 600 West 10th Avenue, Vancouver, BC V5Z 4E1, Canada.
| | - David Laidley
- St. Joseph's Health Care London, Division of Nuclear Medicine, University of Western Ontario, 268 Grosvenor Street, London, Ontario N6A 4V2, Canada.
| | - Calvin Law
- Sunnybrook Health Sciences Centre, Department of Surgery, University of Toronto, 2075 Bayview Ave. Room T2-001, Toronto, Ontario M4N 3M5, Canada.
| | - Timothy Asmis
- The Ottawa Hospital Cancer Centre, Division of Medical Oncology, University of Ottawa, 501 Smyth Road, Ottawa, Ontario K1H8L6, Canada.
| | - David Chan
- Sunnybrook Health Sciences Centre, Department of Medicine, University of Toronto, 2075 Bayview Ave. Room T2-047, Toronto, Ontario M4N 3M5, Canada.
| | - Shereen Ezzat
- Princess Margaret Cancer Centre, Departments of Medicine & Oncology, University of Toronto, 610 University Ave. Room 7-327, Toronto, Ontario M5G 2N2, Canada.
| | - Rachel Goodwin
- The Ottawa Hospital Research Institute, Department of Medical Oncology, University of Ottawa, 501 Smyth Road, Ottawa, Ontario K1H8L6, Canada.
| | - Ozgur Mete
- University Health Network, Department of Pathology, University of Toronto, Toronto, Ontario M5G 2C4, Canada.
| | - Janice Pasieka
- Tom Baker Cancer Center and Foothills Medical Centre, Departments of Surgery & Oncology, University of Calgary, 1403 29th Street NW, North Tower Floor 10, Calgary, Alberta T2N 2T9, Canada.
| | - Juan Rivera
- McGill University Health Centre - Glen Campus, Bloc C - C04.5190, 1001 Decarie Blvd, Montreal, QC H4A 3J1, Canada.
| | - Ralph Wong
- CancerCare Manitoba, St Boniface General Hospital, 407 Tache Avenue, Winnipeg, Manitoba R2H 2A6, Canada.
| | - Eva Segelov
- St Vincent's Clinical School, University of New South Wales, 438 Victoria St, Darlinghurst, NSW 2010, Australia.
| | - Daniel Rayson
- QEII Health Sciences Centre, Division of Medical Oncology, Dalhousie University, Suite 457A Bethune Building, 1276 South Park Street, Halifax, NS B3H 2Y9, Canada.
| |
Collapse
|
245
|
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
|
246
|
de Galiza Barbosa F, Delso G, Ter Voert EEGW, Huellner MW, Herrmann K, Veit-Haibach P. Multi-technique hybrid imaging in PET/CT and PET/MR: what does the future hold? Clin Radiol 2016; 71:660-72. [PMID: 27108800 DOI: 10.1016/j.crad.2016.03.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 03/11/2016] [Accepted: 03/22/2016] [Indexed: 12/19/2022]
Abstract
Integrated positron-emission tomography and computed tomography (PET/CT) is one of the most important imaging techniques to have emerged in oncological practice in the last decade. Hybrid imaging, in general, remains a rapidly growing field, not only in developing countries, but also in western industrialised healthcare systems. A great deal of technological development and research is focused on improving hybrid imaging technology further and introducing new techniques, e.g., integrated PET and magnetic resonance imaging (PET/MRI). Additionally, there are several new PET tracers on the horizon, which have the potential to broaden clinical applications in hybrid imaging for diagnosis as well as therapy. This article aims to highlight some of the major technical and clinical advances that are currently taking place in PET/CT and PET/MRI that will potentially maintain the position of hybrid techniques at the forefront of medical imaging technologies.
Collapse
Affiliation(s)
- F de Galiza Barbosa
- Department of Nuclear Medicine, University Hospital Zurich, Switzerland; University of Zurich, Switzerland
| | - G Delso
- Department of Nuclear Medicine, University Hospital Zurich, Switzerland; GE Healthcare, Waukesha, WI, USA
| | - E E G W Ter Voert
- Department of Nuclear Medicine, University Hospital Zurich, Switzerland; University of Zurich, Switzerland
| | - M W Huellner
- Department of Nuclear Medicine, University Hospital Zurich, Switzerland; University of Zurich, Switzerland; Department of Neuroradiology, University Hospital Zurich, Switzerland
| | - K Herrmann
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, United States; Department of Nuclear Medicine, Universitätsklinikum Würzburg, Oberdürrbacher, Str. 6, Würzburg, Germany
| | - P Veit-Haibach
- Department of Nuclear Medicine, University Hospital Zurich, Switzerland; University of Zurich, Switzerland; Department of Diagnostic and Interventional Radiology, University Hospital Zurich, Switzerland.
| |
Collapse
|
247
|
Krabbe JG, Monaghan PJ, Russell J, de Rijke YB. Analytical evaluation of a second generation assay for chromogranin A; a dual-site study. Clin Chem Lab Med 2016; 54:e139-e142. [PMID: 26393331 DOI: 10.1515/cclm-2015-0782] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 08/26/2015] [Indexed: 01/06/2025]
|
248
|
Correlation and discordance of tumour proliferation index and molecular imaging characteristics and their implications for treatment decisions and outcome pertaining to peptide receptor radionuclide therapy in patients with advanced neuroendocrine tumour: developing a personalized model. Nucl Med Commun 2016; 36:766-74. [PMID: 25920048 DOI: 10.1097/mnm.0000000000000321] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
This communication critically illustrates the molecular imaging features and correlates them with the histopathological characteristics (focussing primarily on Ki-67 or MiB1 index) of neuroendocrine tumours (NETs), their implications for important treatment decision-making, and their response to peptide receptor radionuclide therapy. Such multiparametric analysis of functional imaging (along with conventional size, site-specific and stage-specific morphological assessment and histopathology) is crucial for developing a personalized model for the treatment of advanced and metastatic NET and to fine-tune the multimodal therapies, including the combination regimens. Illustrations with case examples have been made with respect to (a) concordant and (b) discordant tumour proliferation index and functional imaging features, (c) the variable molecular imaging parameters at the intermediate MiB1 indices, including (d) outliers with respect to their MiB1 index, and (e) interlesional heterogeneity between primary and metastatic sites in the same individual as demonstrated by molecular imaging features and its possible implications for therapeutic strategy. In each case, the treatment outcome has been described that would aid in better understanding of the potential usefulness of functional imaging in managing patients with NETs on an individual basis.
Collapse
|
249
|
Folkert IW, Hernandez P, Roses RE. Multidisciplinary management of nonfunctional neuroendocrine tumor of the pancreas. World J Gastroenterol 2016; 22:3105-3116. [PMID: 27003988 PMCID: PMC4789986 DOI: 10.3748/wjg.v22.i11.3105] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 01/05/2016] [Accepted: 01/11/2016] [Indexed: 02/06/2023] Open
Abstract
Pancreatic neuroendocrine tumors (PNETs) are a rare and diverse group of tumors; nonfunctional (NF) PNETs account for the majority of cases. Most patients with NF-PNETs have metastatic disease at the time of presentation. A variety of treatment modalities exist, including medical, liver directed, and surgical treatments. Aggressive surgical management is associated with prolonged survival, however available data are limited by selection bias and the frequent combination of PNETs with carcinoid tumors. Although few patients with metastatic disease will be cured, application of currently available therapies in a multidisciplinary setting can lead to excellent outcomes with prolonged patient survival.
Collapse
|
250
|
Ludwig JM, Ambinder EM, Ghodadra A, Xing M, Prajapati HJ, Kim HS. Lung Shunt Fraction prior to Yttrium-90 Radioembolization Predicts Survival in Patients with Neuroendocrine Liver Metastases: Single-Center Prospective Analysis. Cardiovasc Intervent Radiol 2016; 39:1007-14. [DOI: 10.1007/s00270-016-1323-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 02/21/2016] [Indexed: 12/20/2022]
|