151
|
Kasai Y, Mahuron K, Hirose K, Corvera CU, Kim GE, Hope TA, Shih BE, Warren RS, Bergsland EK, Nakakura EK. Prognostic impact of a large mesenteric mass >2 cm in ileal neuroendocrine tumors. J Surg Oncol 2019; 120:1311-1317. [DOI: 10.1002/jso.25727] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 09/14/2019] [Indexed: 12/17/2022]
Affiliation(s)
- Yosuke Kasai
- Department of SurgeryUniversity of California San Francisco California
- UCSF Helen Diller Family Comprehensive Cancer Center San Francisco California
| | - Kelly Mahuron
- Department of SurgeryUniversity of California San Francisco California
| | - Kenzo Hirose
- Department of SurgeryUniversity of California San Francisco California
- UCSF Helen Diller Family Comprehensive Cancer Center San Francisco California
| | - Carlos U. Corvera
- Department of SurgeryUniversity of California San Francisco California
- UCSF Helen Diller Family Comprehensive Cancer Center San Francisco California
| | - Grace E. Kim
- UCSF Helen Diller Family Comprehensive Cancer Center San Francisco California
- Department of PathologyUniversity of California San Francisco California
| | - Thomas A. Hope
- UCSF Helen Diller Family Comprehensive Cancer Center San Francisco California
- Department of Radiology and Biomedical ImagingUniversity of California San Francisco California
| | - Brandon E. Shih
- UCSF Helen Diller Family Comprehensive Cancer Center San Francisco California
| | - Robert S. Warren
- Department of SurgeryUniversity of California San Francisco California
- UCSF Helen Diller Family Comprehensive Cancer Center San Francisco California
| | - Emily K. Bergsland
- UCSF Helen Diller Family Comprehensive Cancer Center San Francisco California
- Department of MedicineUniversity of California San Francisco California
| | - Eric K. Nakakura
- Department of SurgeryUniversity of California San Francisco California
- UCSF Helen Diller Family Comprehensive Cancer Center San Francisco California
| |
Collapse
|
152
|
Singh S, Moody L, Chan DL, Metz DC, Strosberg J, Asmis T, Bailey DL, Bergsland E, Brendtro K, Carroll R, Cleary S, Kim M, Kong G, Law C, Lawrence B, McEwan A, McGregor C, Michael M, Pasieka J, Pavlakis N, Pommier R, Soulen M, Wyld D, Segelov E. Follow-up Recommendations for Completely Resected Gastroenteropancreatic Neuroendocrine Tumors. JAMA Oncol 2019; 4:1597-1604. [PMID: 30054622 DOI: 10.1001/jamaoncol.2018.2428] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
There is no consensus on optimal follow-up for completely resected gastroenteropancreatic neuroendocrine tumors. Published guidelines for follow-up are complex and emphasize closer surveillance in the first 3 years after resection. Neuroendocrine tumors have a different pattern and timescale of recurrence, and thus require more practical and tailored follow-up. The Commonwealth Neuroendocrine Tumour Collaboration convened an international multidisciplinary expert panel, in collaboration with the North American Neuroendocrine Tumor Society, to create patient-centered follow-up recommendations for completely resected gastroenteropancreatic neuroendocrine tumors. This panel used the RAND/UCLA (University of California, Los Angeles) Appropriateness Method to generate recommendations. A large international survey was conducted outlining current the surveillance practice of neuroendocrine tumor practitioners and shortcomings of the current guidelines. A systematic review of available data to date was supplemented by recurrence data from 2 large patient series. The resultant guidelines suggest follow-up for at least 10 years for fully resected small-bowel and pancreatic neuroendocrine tumors and also identify clinical situations in which no follow-up is required. These recommendations stratify follow-up strategies based on evidence-based prognostic factors that allow for a more individualized patient-centered approach to this complex and heterogeneous malignant neoplasm.
Collapse
Affiliation(s)
- Simron Singh
- Department of Medical Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Lesley Moody
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, Ontario, Canada
| | - David L Chan
- Department of Medical Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - David C Metz
- Perelman School of Medicine, Department of Gastroenterology, University of Pennsylvania, Philadelphia
| | - Jonathan Strosberg
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Centre, Tampa, Florida
| | - Timothy Asmis
- Department of Internal Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Dale L Bailey
- Department of Nuclear Medicine, Royal North Shore Hospital, Sydney, Australia
| | - Emily Bergsland
- Department of Medical Oncology, University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco
| | - Kari Brendtro
- North American Neuroendocrine Tumor Society, Albany, New York
| | - Richard Carroll
- Department of Endocrinology, Wellington Regional Hospital, Wellington, New Zealand
| | - Sean Cleary
- Department of Surgery, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Michelle Kim
- Department of Gastroenterology, Mount Sinai Hospital, New York, New York
| | - Grace Kong
- Department of Nuclear Medicine, Peter MacCullum Cancer Centre, Melbourne, Australia
| | - Calvin Law
- Department of Surgery, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Ben Lawrence
- Department of Medical Oncology, Auckland Hospital, Auckland, New Zealand
| | - Alexander McEwan
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
| | - Caitlin McGregor
- Department of Medical Imaging, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Michael Michael
- Department of Medical Oncology, Peter MacCullum Cancer Centre, Melbourne, Australia
| | - Janice Pasieka
- Department of Surgery, Tom Baker Cancer Centre, Calgary, Alberta, Canada
| | - Nick Pavlakis
- Department of Medical Oncology, Royal North Shore Hospital, Sydney, Australia
| | - Rodney Pommier
- Department of Surgery, Oregon Health & Science University, Portland
| | - Michael Soulen
- Perelman School of Medicine, Department of Medical Imaging, University of Pennsylvania, Philadelphia
| | - David Wyld
- Department of Medical Oncology, Royal Brisbane and Women's Hospital, Queensland, Australia
| | - Eva Segelov
- Department of Medical Oncology, Monash University, Clayton, Australia
| | | |
Collapse
|
153
|
Larouche V, Akirov A, Alshehri S, Ezzat S. Management of Small Bowel Neuroendocrine Tumors. Cancers (Basel) 2019; 11:cancers11091395. [PMID: 31540509 PMCID: PMC6770692 DOI: 10.3390/cancers11091395] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 09/08/2019] [Accepted: 09/15/2019] [Indexed: 12/20/2022] Open
Abstract
Several important landmark trials have reshaped the landscape of non-surgical management of small bowel neuroendocrine tumors over the last few years, with the confirmation of the antitumor effect of somatostatin analogue therapy in PROMID and CLARINET trials as well as the advent of therapies with significant potential such as mammalian target of rapamycin inhibitor (mTor) everolimus (RADIANT trials) and peptide receptor radionuclide therapy (PRRT) with 177-Lutetium (NETTER-1 trial). This narrative summarizes the recommended management strategies of small bowel neuroendocrine tumors. We review the main evidence behind each recommendation as well as compare and contrast four major guidelines, namely the 2016 Canadian Consensus guidelines, the 2017 North American Neuroendocrine Tumor Society guidelines, the 2018 National Comprehensive Cancer Network guidelines, and the 2016 European Neuroendocrine Tumor Society guidelines. Different clinical situations will be addressed, from loco-regional therapy to metastatic unresectable disease. Carcinoid syndrome, which is mostly managed by somatostatin analogue therapy and the serotonin antagonist telotristat etiprate for refractory diarrhea, as well as neuroendocrine carcinoma will be reviewed. However, several questions remain unanswered, such as the optimal management of neuroendocrine carcinomas or the effect of combining and sequencing of the aforementioned modalities where more randomized controlled trials are needed.
Collapse
Affiliation(s)
- Vincent Larouche
- Endocrine Oncology Site Group, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON M5G2C1, Canada; (A.A.); (S.A.); (S.E.)
- Division of Endocrinology and Metabolism, Department of Medicine, Jewish General Hospital, McGill University, Montreal, QC H3T1E2, Canada
- Correspondence: ; Tel.: +1-(514)-340-8222 (ext. 28521)
| | - Amit Akirov
- Endocrine Oncology Site Group, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON M5G2C1, Canada; (A.A.); (S.A.); (S.E.)
- Department of Medicine, Institute of Endocrinology, Beilinson Hospital, Petach Tikva 4941492, Israel
- Department of Medicine, Sackler School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Sameerah Alshehri
- Endocrine Oncology Site Group, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON M5G2C1, Canada; (A.A.); (S.A.); (S.E.)
| | - Shereen Ezzat
- Endocrine Oncology Site Group, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON M5G2C1, Canada; (A.A.); (S.A.); (S.E.)
| |
Collapse
|
154
|
Woodhouse B, Pattison S, Segelov E, Singh S, Parker K, Kong G, Macdonald W, Wyld D, Meyer-Rochow G, Pavlakis N, Conroy S, Gordon V, Koea J, Kramer N, Michael M, Wakelin K, Asif T, Lo D, Price T, Lawrence B. Consensus-Derived Quality Performance Indicators for Neuroendocrine Tumour Care. J Clin Med 2019; 8:jcm8091455. [PMID: 31547431 PMCID: PMC6780732 DOI: 10.3390/jcm8091455] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 09/05/2019] [Accepted: 09/07/2019] [Indexed: 12/16/2022] Open
Abstract
Quality performance indicators (QPIs) are used to monitor the delivery of cancer care. Neuroendocrine tumours (NETs) are a family of individually uncommon cancers that derive from neuroendocrine cells or their precursors, and can occur in most organs. There are currently no QPIs available for NETs and their heterogeneity makes QPI development difficult. CommNETs is a collaboration between NET clinicians, researchers and advocates in Canada, Australia and New Zealand. We created QPIs for NETs using a three-step consensus process. First, a multidisciplinary team used the nominal group technique to create candidates (n = 133) which were then curated into appropriateness statements (62 statements, 44 sub-statements). A two-stage modified RAND/UCLA Delphi consensus process was conducted: an online survey rated the statement appropriateness then the top-ranked statements (n = 20) were assessed in a face-to-face meeting. Finally, 10 QPIs met consensus criteria; documentation of primary site, proliferative index, differentiation, tumour board review, use of a structured pathology report, presence of distant metastasis, 5- and 10-year disease-free and overall survival. These NET QPIs will be trialed as a method to monitor and improve care for people with NETs and to facilitate international comparison.
Collapse
Affiliation(s)
- Braden Woodhouse
- Discipline of Oncology, Faculty of Medicine and Health Sciences, The University of Auckland, Auckland 1023, New Zealand.
| | - Sharon Pattison
- Department of Medicine, University of Otago, Dunedin 9016, New Zealand.
| | - Eva Segelov
- Department of Medical Oncology, Monash University and Monash Health, Melbourne 3800, Australia.
| | - Simron Singh
- Department of Medical Oncology, Sunnybrook Odette Cancer Center, University of Toronto, Toronto, ON M4N 3M5, Canada.
| | - Kate Parker
- Discipline of Oncology, Faculty of Medicine and Health Sciences, The University of Auckland, Auckland 1023, New Zealand.
| | - Grace Kong
- Department of Nuclear Medicine, Peter MacCallum Cancer Centre, Melbourne 3000, Australia.
| | - William Macdonald
- Department of Nuclear Medicine, Fiona Stanley Hospital, Perth 6150, Australia.
| | - David Wyld
- Department of Medical Oncology, Royal Brisbane and Women's Hospital, Brisbane 4029, Australia.
- School of Medicine, University of Queensland, Brisbane 4072, Australia.
| | - Goswin Meyer-Rochow
- Department of General Surgery, Waikato Hospital, Hamilton 3204, New Zealand.
| | - Nick Pavlakis
- Department of Medical Oncology, Royal North Shore Hospital, Sydney 2065, Australia.
| | | | - Vallerie Gordon
- Department of Medical Oncology, Cancer Care Manitoba, Winnipeg, MB R3E 0V9, Canada.
| | - Jonathan Koea
- Department of General Surgery, North Shore Hospital, Auckland 0620, New Zealand.
| | - Nicole Kramer
- Department of Pathology, North Shore Hospital, Auckland 0620, New Zealand.
| | - Michael Michael
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne 3000, Australia.
| | | | - Tehmina Asif
- Department of Medical Oncology, Saskatchewan Cancer Agency, Saskatoon, SK S4W 0G3, Canada.
| | - Dorothy Lo
- Department of Medical Oncology, St Joseph's Health Care, Toronto, ON M6R 1B5, Canada.
| | - Timothy Price
- Department of Medical Oncology, The Queen Elizabeth Hospital, Adelaide 5011, Australia.
| | - Ben Lawrence
- Discipline of Oncology, Faculty of Medicine and Health Sciences, The University of Auckland, Auckland 1023, New Zealand.
- Department of Medical Oncology, Auckland City Hospital, Auckland 1023, New Zealand.
| |
Collapse
|
155
|
Fang JM, Shi J. A Clinicopathologic and Molecular Update of Pancreatic Neuroendocrine Neoplasms With a Focus on the New World Health Organization Classification. Arch Pathol Lab Med 2019; 143:1317-1326. [PMID: 31509453 DOI: 10.5858/arpa.2019-0338-ra] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
CONTEXT.— According to the 2017 World Health Organization classification, pancreatic neuroendocrine neoplasms (PanNENs) include a new category of pancreatic neuroendocrine tumor, grade 3, which is often difficult to differentiate from pancreatic neuroendocrine carcinoma. However, pancreatic neuroendocrine tumor grade 3 and pancreatic neuroendocrine carcinoma are distinct entities with very different clinical presentation, prognosis, and therapeutic strategies. Recent discoveries on the molecular characteristics of pancreatic neuroendocrine tumors also play an essential role in the pathologic differential diagnosis of PanNENs. In addition, the histopathologic varieties of PanNENs bring in many differential diagnoses with other pancreatic neoplasms, especially acinar cell carcinoma, solid pseudopapillary neoplasm, and ductal adenocarcinoma. OBJECTIVE.— To provide a brief update of the World Health Organization classification; the clinical, histopathologic, immunohistochemical, and molecular characteristics; and the differential diagnoses and biological behavior of PanNENs. DATA SOURCES.— Analysis of the pertinent literature (PubMed) and authors' clinical practice experience based on institutional and consultation materials. CONCLUSIONS.— The evolving clinical, histopathologic, immunohistochemical, and molecular features of PanNENs are reviewed. Important differential diagnoses with other neoplasms of the pancreas are discussed.
Collapse
Affiliation(s)
- Jiayun M Fang
- From the Department of Pathology, University of Michigan, Ann Arbor
| | - Jiaqi Shi
- From the Department of Pathology, University of Michigan, Ann Arbor
| |
Collapse
|
156
|
Naraev BG, Halland M, Halperin DM, Purvis AJ, O'Dorisio TM, Halfdanarson TR. Management of Diarrhea in Patients With Carcinoid Syndrome. Pancreas 2019; 48:961-972. [PMID: 31425482 PMCID: PMC6867674 DOI: 10.1097/mpa.0000000000001384] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 07/10/2019] [Indexed: 02/06/2023]
Abstract
Neuroendocrine tumors (NETs) arise from enterochromaffin cells found in neuroendocrine tissues, with most occurring in the gastrointestinal tract. The global incidence of NETs has increased in the past 15 years, likely due to better diagnostic methods. Small-bowel NETs are frequently associated with carcinoid syndrome (CS). Carcinoid syndrome diarrhea occurs in 80% of CS patients and poses a substantial symptomatic and economic burden. Patients with CS diarrhea frequently suffer from diarrhea and flushing and report corresponding impairment in quality of life, requiring substantial changes in daily activities and lifestyle. Treatment paradigms range from surgical debulking to liver-directed therapies to treatment with somatostatin analogs, nonspecific anti-diarrheal agents, and a tryptophan hydroxylase inhibitor. Other causes of diarrhea, including steatorrhea, short bowel syndrome, and bile acid malabsorption, should be considered in NET patients with refractory diarrhea. More therapeutic options are needed for symptomatic management of patients with NETs, and better understanding of the pathophysiology can empower clinicians with improved patient care.
Collapse
Affiliation(s)
| | - Magnus Halland
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | - Daniel M. Halperin
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Amy J. Purvis
- University of Arizona Cancer Center (UACC), Phoenix, AZ
| | - Thomas M. O'Dorisio
- Neuroendocrine Cancer Program, University of Iowa Health Care, Iowa City, IA
| | | |
Collapse
|
157
|
Gaertner F, Plum T, Kreppel B, Eppard E, Meisenheimer M, Strunk H, Bundschuh R, Sinnes J, Rösch F, Essler M. Clinical evaluation of [68Ga]Ga-DATA-TOC in comparison to [68Ga]Ga-DOTA-TOC in patients with neuroendocrine tumours. Nucl Med Biol 2019; 76-77:1-9. [DOI: 10.1016/j.nucmedbio.2019.08.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 08/07/2019] [Accepted: 08/26/2019] [Indexed: 01/20/2023]
|
158
|
Gabrielsen J, Girone G, Bennett B, Jung A. Long-Acting Somatostatin Analogue Safety Monitoring Protocol for Outpatients With Neuroendocrine Tumors. J Adv Pract Oncol 2019; 10:646-659. [PMID: 33391850 PMCID: PMC7517774 DOI: 10.6004/jadpro.2019.10.7.2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Somatostatin analogues (SSAs) are widely used in the long-term treatment of neuroendocrine tumors (NETs) and have a relatively favorable safety profile. However, SSAs are associated with specific side effects that are important to monitor. Currently, there is no standardized safety monitoring protocol for health-care professionals to use as a reference when initiating patients on long-acting SSAs. With the expansion of SSA use from symptomatic control to include antiproliferative tumor treatment in patients with NETs, it is increasingly important that patients taking these medications are properly monitored. The purpose of this analysis was to develop a comprehensive, practical SSA safety monitoring protocol for patients with NETs in the outpatient setting. This strategy was based on side effect frequencies that were reported and the monitoring parameters used in influential clinical and safety trials. Based on our assessment, we consider monitoring gallbladder imaging, laboratory tests (including blood chemistry, thyroid-stimulating hormone, hemoglobin A1c, and stool studies), vital signs, and physical examinations as the most important parameters when evaluating the safety of long-term SSA therapy. Due to the frequency at which patients experienced diarrhea as a side effect in clinical trials, questions about urgency, frequency, timing, consistency, odor, and color of bowel movements should be asked as part of the follow-up visits every 6 months to help differentiate between drug-induced vs. disease-associated causes. This broad monitoring strategy for patients receiving long-term SSAs was developed specifically for patients with NETs; however, the use of this protocol could be expanded to other indications in the future.
Collapse
Affiliation(s)
| | - Gianna Girone
- Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Bonita Bennett
- Hospital of the University of Pennsylvania, Perelman Center for Advanced Medicine, Philadelphia, Pennsylvania
| | - Anna Jung
- Hospital of the University of Pennsylvania, Perelman Center for Advanced Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
159
|
Burden of Carcinoid Heart Disease in Patients With Carcinoid Syndrome Initiating Somatostatin Analogues. Clin Ther 2019; 41:1716-1723.e2. [DOI: 10.1016/j.clinthera.2019.06.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 06/17/2019] [Accepted: 06/21/2019] [Indexed: 12/25/2022]
|
160
|
Safety and Activity of Metronomic Temozolomide in Second-Line Treatment of Advanced Neuroendocrine Neoplasms. J Clin Med 2019; 8:jcm8081224. [PMID: 31443197 PMCID: PMC6723560 DOI: 10.3390/jcm8081224] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 08/07/2019] [Accepted: 08/12/2019] [Indexed: 02/07/2023] Open
Abstract
Background. Platinum-based chemotherapy is the mainstay of front-line treatment of patients affected by pluri-metastatic intermediate/high grade NeuroEndocrine Neoplasms (NENs). However, there are no standard second-line treatments at disease progression. Previous clinical experiences have evidenced that temozolomide (TMZ), an oral analog of dacarbazine, is active against NENs at standard doses of 150 to 200 mg/mq per day on days 1 to 5 of a 28-day cycle, even if a significant treatment-related toxicity is reported. Methods. Metastatic NENs patients were treated at the ENETS (European NeuroEndocrine Tumor Society) center of excellence of Naples (Italy), from 2014 to 2017 with a second-line alternative metronomic schedule of TMZ, 75 mg/m2per os “one week on/one week off”. Toxicity was graded with NCI-CTC criteria v4.0; objective responses with RECIST v1.1 and performance status (PS) according to ECOG. Results. Twenty-six consecutive patients were treated. Median age was 65.5 years. The predominant primary organs were pancreas and lung. Grading was G2 in 11 patients, G3 in 15. More than half of patients had a PS 2 (15 vs. 11 with PS 1). The median time-on-temozolomide therapy was 12.2 months (95% CI: 11.4–19.6). No G3/G4 toxicities were registered. Complete response was obtained in 1 patient, partial response in 4, stable disease in 19 (disease control rate: 92.3%), and progressive disease in 2. The median overall survival from TMZ start was 28.3 months. PS improved in 73% of patients. Conclusions. Metronomic TMZ is a suitable treatment for G2 and G3 NENs particularly in PS 2 patients. Prospective and larger trials are needed to confirm these results.
Collapse
|
161
|
Biological and Biochemical Basis of the Differential Efficacy of First and Second Generation Somatostatin Receptor Ligands in Neuroendocrine Neoplasms. Int J Mol Sci 2019; 20:ijms20163940. [PMID: 31412614 PMCID: PMC6720449 DOI: 10.3390/ijms20163940] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/1970] [Revised: 08/05/2019] [Accepted: 08/08/2019] [Indexed: 02/07/2023] Open
Abstract
Endogenous somatostatin shows anti-secretory effects in both physiological and pathological settings, as well as inhibitory activity on cell growth. Since somatostatin is not suitable for clinical practice, researchers developed synthetic somatostatin receptor ligands (SRLs) to overcome this limitation. Currently, SRLs represent pivotal tools in the treatment algorithm of neuroendocrine tumors (NETs). Octreotide and lanreotide are the first-generation SRLs developed and show a preferential binding affinity to somatostatin receptor (SST) subtype 2, while pasireotide, which is a second-generation SRL, has high affinity for multiple SSTs (SST5 > SST2 > SST3 > SST1). A number of studies demonstrated that first-generation and second-generation SRLs show distinct functional properties, besides the mere receptor affinity. Therefore, the aim of the present review is to critically review the current evidence on the biological effects of SRLs in pituitary adenomas and neuroendocrine tumors, by mainly focusing on the differences between first-generation and second-generation ligands.
Collapse
|
162
|
Calanchini M, Tadman M, Krogh J, Fabbri A, Grossman A, Shine B. Measurement of urinary 5-HIAA: correlation between spot versus 24-h urine collection. Endocr Connect 2019; 8:1082-1088. [PMID: 31265996 PMCID: PMC6652243 DOI: 10.1530/ec-19-0269] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 07/01/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND The 24-h urinary output of 5-hydroxyindoleacetic acid (5-HIAA) is used to monitor disease progression and treatment responses of neuroendocrine neoplasms (NENs). Several conditions are required for 5-HIAA assay, involving urine collection/preservation and food/drug restrictions. AIM To evaluate the correlation between 5-HIAA concentration in a spot urine sample and the output in a 24-h urine collection, and whether spot urine specimens can replace 24-h collection. METHODS Patients with NENs or symptoms suggestive of NENs were asked to provide a separate spot urine at the end of the 24-h urine collection for 5-HIAA assessment. The upper reference limit for 24-h urinary 5-HIAA was 40 µmol/24 h. 5-HIAA measurements in spot urine samples were corrected for variation in urine flow rate by expressing results as a ratio to creatinine concentration. RESULTS We included 136 paired urinary samples for 5-HIAA assessment from 111 patients (100 NENs). The correlation between 5-HIAA values measured in 24-h and spot urines was r = +0.863 (P < 0.001) and r = +0.840 (P < 0.001) including only NEN patients. Using the 24-h urinary 5-HIAA as reference method, the AUC on ROC analysis for spot urinary 5-HIAA was 0.948 (95% CI, 0.914-0.983; P < 0.001), attaining a sensitivity of 83% and specificity of 95% using 5.3 mol/mmol as cut-off for the spot urine. The AUC among NEN patients alone was 0.945 (95% CI, 0.904-0.987; P < 0.001). CONCLUSIONS The ratio of 5-HIAA to creatinine in a spot urine could replace the measurement of 5-HIAA output in a 24-h urine collection, especially for follow-up of patients with known elevated 5-HIAA levels.
Collapse
Affiliation(s)
- Matilde Calanchini
- Oxford Centre for Diabetes, Endocrinology & Metabolism, Churchill Hospital, University of Oxford, Oxford, UK
- Endocrinology & Metabolism Unit, CTO A. Alesini Hospital ASL Roma 2, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Correspondence should be addressed to M Calanchini:
| | - Michael Tadman
- Oxford Centre for Diabetes, Endocrinology & Metabolism, Churchill Hospital, University of Oxford, Oxford, UK
| | - Jesper Krogh
- Oxford Centre for Diabetes, Endocrinology & Metabolism, Churchill Hospital, University of Oxford, Oxford, UK
| | - Andrea Fabbri
- Endocrinology & Metabolism Unit, CTO A. Alesini Hospital ASL Roma 2, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Ashley Grossman
- Oxford Centre for Diabetes, Endocrinology & Metabolism, Churchill Hospital, University of Oxford, Oxford, UK
| | - Brian Shine
- Oxford Centre for Diabetes, Endocrinology & Metabolism, Churchill Hospital, University of Oxford, Oxford, UK
| |
Collapse
|
163
|
Cives M, Pelle’ E, Quaresmini D, Mandriani B, Tucci M, Silvestris F. The Role of Cytotoxic Chemotherapy in Well-Differentiated Gastroenteropancreatic and Lung Neuroendocrine Tumors. Curr Treat Options Oncol 2019; 20:72. [DOI: 10.1007/s11864-019-0669-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
164
|
Abstract
OPINION STATEMENT Carcinoid syndrome (CS) is a complex disorder caused by functional neuroendocrine tumors (NETs). This debilitating disease is characterized by hyper-secretion of biologically active substances eliciting major hormonal symptoms burden and fibrotic changes that are often challenging for management. There have been a number of insights that have substantially advanced treatments since the introduction of somatostatin analogs (SSAs). Second-line treatments are needed in a substantial proportion of patients with advanced disease that have uncontrolled hormone secretion on the highest labeled doses of SSAs. International guidelines suggest several available options including dose escalation of SSAs, interferon alpha, everolimus, radionuclide therapy, liver-directed therapies, and the novel tryptophan hydroxylase 1 inhibitor, telotristat ethyl. The clear preference of one second-line therapy over the other is not stated since their relative and long-term efficacy are largely unknown, and standardized approach of hormonal response assessment is lacking in the literature. In the clinical setting, the treatment of CS is guided in conjunction with patients' performance status, tumor origin, grade, stage, and growth rate, with regard to both anti-hormonal, as well as anti-proliferative effect. There is an unmet need for further well-designed randomized placebo-controlled and head-to-head studies that systematically assess CS symptom control and biochemical response following a specific intervention.
Collapse
|
165
|
Song L, Zhai X, Yu S, Ma Y, Wang F, Yu X, Tao S, Lian Y, Yang M, Tao W, Fan Q. Clinical analysis of 547 patients with neuroendocrine tumors in a Chinese population: A single-center study. Cancer Med 2019; 8:3729-3737. [PMID: 31127690 PMCID: PMC6639184 DOI: 10.1002/cam4.2259] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 04/16/2019] [Accepted: 05/06/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Neuroendocrine tumors (NETs) are rare, which has resulted in a lack of published data on their epidemiology and clinical features. We therefore aimed to investigate the epidemiology, clinical features, treatments, and prognosis of patients with NETs. METHODS The clinicopathologic characteristics of 547 patients who were pathologically diagnosed with NETs were retrospectively analyzed, including age, sex, primary and metastatic sites, symptoms, pathology, treatment, and prognosis. RESULTS The 547 patients had a wide age range (9-87 years), with a male to female ratio of 1:1.1. The primary tumor sites included 413 in the digestive system, 74 in the lung, 15 in the mediastinum, 8 in unknown sites, and 37 in other sites. Of the 413 patients with digestive system NETs, the pancreas, rectum, and stomach were the most common primary sites. Blood metastases were found in 84 patients at initial diagnosis, and the liver, bone, and lung were the most frequent sites of metastasis. Lymph node metastases were found in 82 patients at initial diagnosis. Surgery and chemotherapy were the most widely applied treatments. Statistical analysis showed that age <50 years, female sex, lower-grade tumor, no distant metastasis, intestinal NET and surgery indicated a favorable prognosis. CONCLUSIONS A difference between China and other countries is that small intestinal NETs are quite common in other countries but are rare in China. In China, the most common primary sites are the pancreas, rectum, and stomach. Furthermore, no unified treatments exist, though prognoses could be improved by using methods such as surgery, targeted therapies, and somatostatin analogs. CLINICAL TRIAL REGISTRATION This study was not a clinical trial.
Collapse
Affiliation(s)
- Lijie Song
- Department of OncologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Xuejia Zhai
- Department of OncologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Shunli Yu
- Department of UrologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Yihui Ma
- Department of PathologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Feng Wang
- Department of PathologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Xuxu Yu
- Department of OncologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Shuang Tao
- Department of OncologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Yujin Lian
- Department of OncologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Minjie Yang
- Department of OncologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Weili Tao
- Department of OncologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Qingxia Fan
- Department of OncologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| |
Collapse
|
166
|
Cook R, Hendifar AE. Evidence-Based Policy in Practice: Management of Carcinoid Syndrome Diarrhea. P & T : A PEER-REVIEWED JOURNAL FOR FORMULARY MANAGEMENT 2019; 44:424-427. [PMID: 31258314 PMCID: PMC6590927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Carcinoid syndrome causes substantial morbidity and reduces quality of life and survival. In a recent clinical trial, 97% of patients reported bowel movement-related issues, abdominal pain, flushing, and low energy. Combining somatostatin analogs with elotristat ethyl provides a new option for managing refractory CS symptoms. Health care providers should consider strategies that take advantage of approved dosing for patients with CS-related diarrhea.
Collapse
|
167
|
Peptide Receptor Radionuclide Therapy Combined With Chemotherapy in Patients With Neuroendocrine Tumors. Clin Nucl Med 2019; 44:e329-e335. [DOI: 10.1097/rlu.0000000000002532] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
168
|
Wolin E, Benson III A. Systemic Treatment Options for Carcinoid Syndrome: A Systematic Review. Oncology 2019; 96:273-289. [DOI: 10.1159/000499049] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 02/07/2019] [Indexed: 11/19/2022]
|
169
|
Kaderli RM, Spanjol M, Kollár A, Bütikofer L, Gloy V, Dumont RA, Seiler CA, Christ ER, Radojewski P, Briel M, Walter MA. Therapeutic Options for Neuroendocrine Tumors: A Systematic Review and Network Meta-analysis. JAMA Oncol 2019; 5:480-489. [PMID: 30763436 PMCID: PMC6459123 DOI: 10.1001/jamaoncol.2018.6720] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 11/26/2018] [Indexed: 12/17/2022]
Abstract
IMPORTANCE Multiple therapies are currently available for patients with neuroendocrine tumors (NETs), yet many therapies have not been compared head-to-head within randomized clinical trials (RCTs). OBJECTIVE To assess the relative safety and efficacy of therapies for NETs. DATA SOURCES PubMed, Embase, the Cochrane Central Register of Controlled Trials, trial registries, meeting abstracts, and reference lists from January 1, 1947, to March 2, 2018, were searched. Key search terms included neuroendocrine tumors, gastrointestinal neoplasms, therapy, and randomized controlled trial. STUDY SELECTION Randomized clinical trials comparing 2 or more therapies in patients with NETs (primarily gastrointestinal and pancreatic) were evaluated. Thirty RCTs met the selection criteria. DATA EXTRACTION AND SYNTHESIS Pairs of independent reviewers screened studies, extracted data, and assessed the risk of bias. A network meta-analysis with a frequentist approach was used to compare the efficacy of therapies; the Preferred Reporting Items for Systematic Reviews and Meta-analyses guideline was used. MAIN OUTCOMES AND MEASURES Disease control, progression-free survival, overall survival, adverse events, and quality of life. RESULTS The systematic review identified 30 relevant RCTs comprising 3895 patients (48.4% women) assigned to 22 different therapies for NETs. These therapies showed a broad range of risk for serious and nonserious adverse events. The network meta-analyses included 16 RCTs with predominantly a low risk of bias; nevertheless, precision-of-treatment estimates and estimated heterogeneity were limited. The network meta-analysis found 7 therapies for pancreatic NETs: everolimus (hazard ratio [HR], 0.35 [95% CI, 0.28-0.45]), everolimus plus somatostatin analogue (HR, 0.35 [95% CI, 0.25-0.51]), everolimus plus bevacizumab plus somatostatin analogue (HR, 0.44 [95% CI, 0.26-0.75]), interferon (HR, 0.37 [95% CI, 0.16-0.83]), interferon plus somatostatin analogue (HR, 0.31 [95% CI, 0.13-0.71]), somatostatin analogue (HR, 0.46 [95% CI, 0.33-0.66]), and sunitinib (HR, 0.42 [95% CI, 0.26-0.67]), and 5 therapies for gastrointestinal NETs: bevacizumab plus somatostatin analogue (HR, 0.22 [95% CI, 0.05-0.99]), everolimus plus somatostatin analogue (HR, 0.31 [95% CI, 0.11-0.90]), interferon plus somatostatin analogue (HR, 0.27 [95% CI, 0.07-0.96]), Lu 177-dotatate plus somatostatin analogue (HR, 0.08 [95% CI, 0.03-0.26], and somatostatin analogues (HR, 0.40 [95% CI, 0.21-0.78]) with higher efficacy than placebo and suggests an overall superiority of combination therapies. CONCLUSIONS AND RELEVANCE The findings from this study suggest that a range of efficient therapies with different safety profiles is available for patients with NETs.
Collapse
Affiliation(s)
- Reto M. Kaderli
- Department of Visceral Surgery and Medicine, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Marko Spanjol
- Department of Nuclear Medicine, University Hospital, University of Geneva, Geneva, Switzerland
| | - Attila Kollár
- Department of Medical Oncology, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Lukas Bütikofer
- Clinical Trials Unit Bern, Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
| | - Viktoria Gloy
- Department of Nuclear Medicine, University Hospital, University of Geneva, Geneva, Switzerland
| | - Rebecca A. Dumont
- Department of Nuclear Medicine, University Hospital, University of Geneva, Geneva, Switzerland
| | - Christian A. Seiler
- Department of Visceral Surgery and Medicine, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Emanuel R. Christ
- Department of Endocrinology, Diabetes, and Metabolism, Basel University Hospital, University of Basel, Basel, Switzerland
| | - Piotr Radojewski
- Department of Nuclear Medicine, University Hospital, University of Geneva, Geneva, Switzerland
| | - Matthias Briel
- Institute for Clinical Epidemiology and Biostatistics, Department of Clinical Research, Basel University Hospital, University of Basel, Basel, Switzerland
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada
| | - Martin A. Walter
- Department of Nuclear Medicine, University Hospital, University of Geneva, Geneva, Switzerland
| |
Collapse
|
170
|
Abstract
Telotristat ethyl (Xermelo®), a first-in-class peripheral tryptophan hydroxylase (TPH) inhibitor, is approved to treat carcinoid syndrome diarrhoea in combination with somatostatin analogue (SSA) therapy in adults inadequately controlled by SSA therapy alone. Some neuroendocrine tumours secrete serotonin (5-HT) into the blood, resulting in frequent bowel movements (BMs) and other symptoms. Telotristat ethyl inhibits TPH, thereby reducing the production of 5-HT and improving carcinoid syndrome diarrhoea. In the 12-week placebo-controlled phase of randomized trials in patients with carcinoid syndrome diarrhoea (most of whom were receiving SSA therapy), the addition of oral telotristat ethyl 250 three times daily provided significant reductions in the frequency of BMs and levels of urinary 5-hydroxyindolacetic acid (u5-HIAA; a metabolite of 5-HT) relative to placebo. Telotristat ethyl 250 mg three times daily was well tolerated, with the proportion of patients reporting at least one treatment-emergent adverse event being similar to that with placebo. With regard to adverse events of special interest, relative to placebo, telotristat ethyl had a comparable incidence of depression-related symptoms, a somewhat higher incidence of gastrointestinal (GI) disorders and a higher incidence of elevated hepatic enzyme levels.
Collapse
|
171
|
Fagan R, Bokhari SSN, Inayat F. Vitamin D and vitamin B 12 deficiencies in patients with small intestinal carcinoid tumour: is opioid use disorder a confounding factor in the diagnosis? BMJ Case Rep 2019; 12:12/3/e227430. [PMID: 30878964 DOI: 10.1136/bcr-2018-227430] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Carcinoid tumours have the ability to secrete various peptides and bioamines that lead to carcinoid syndrome manifested as cutaneous flushing, diarrhoea, bronchial constriction and cardiac involvement. The deficiencies of vitamins D and B12 have previously been reported in patients with carcinoid tumours presumably due to chronic diarrhoea associated with the carcinoid syndrome. Herein, we chronicle the case of a patient with opioid use disorder who presented with small bowel obstruction that was found to be caused by a midgut carcinoid tumour. Laboratory studies revealed deficiencies of vitamins D and B12 even though he denied diarrhoea and had no other aetiology of deficiencies of these vitamins. Additionally, this paper presents a review of the published medical literature pertaining to clinical features, diagnostic investigations and treatment of intestinal carcinoid tumours and explores possible explanations for the observed deficiencies in these patients.
Collapse
Affiliation(s)
- Richard Fagan
- West Suburban Medical Center, Oak Park, Illinois, USA
| | | | | |
Collapse
|
172
|
Clement DSVM, Tesselaar MET, van Leerdam ME, Srirajaskanthan R, Ramage JK. Nutritional and vitamin status in patients with neuroendocrine neoplasms. World J Gastroenterol 2019; 25:1171-1184. [PMID: 30886501 PMCID: PMC6421241 DOI: 10.3748/wjg.v25.i10.1171] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 02/13/2019] [Accepted: 02/22/2019] [Indexed: 02/06/2023] Open
Abstract
Symptoms of gastroenteropancreatic located neuroendocrine neoplasms (GEP-NENs) are often related to food intake and manifest as abdominal pain or diarrhoea which can influence patients nutritional status. Malnutrition is common in cancer patients and influences quality of life, treatment options and survival but is also present in up to 40% of patients with GEP-NENs. As part of malnutrition there are often deficiencies in fat-soluble vitamins, mainly vitamin D. Little knowledge exists on trace elements. Several factors influence the development of malnutrition such as size and localisation of the primary tumour as well as metastases, side effects from treatment but also hormone production of the tumour itself. One of the main influencing factors leading to malnutrition is diarrhoea which leads to dehydration and electrolyte disturbances. Treatment of diarrhoea should be guided by its cause. Screening for malnutrition should be part of routine care in every GEP-NEN patient. Multidisciplinary treatment including dietician support is necessary for all malnourished patients with GEP-NENs.
Collapse
Affiliation(s)
- Dominique SVM Clement
- Neuroendocrine Tumour Unit, King’s College Hospital ENETS Centre of Excellence, London SE5 9RS, United Kingdom
| | - Margot ET Tesselaar
- Department of Medical Oncology, Netherlands Cancer Institute ENETS Centre of Excellence, Amsterdam 1066 CX, Netherlands
| | - Monique E van Leerdam
- Department of Gastrointestinal Oncology, Netherlands Cancer Institute, Amsterdam 1066 CX, Netherlands
| | - Rajaventhan Srirajaskanthan
- Neuroendocrine Tumour Unit, King’s College Hospital ENETS Centre of Excellence, London SE5 9RS, United Kingdom
- Department of Gastroenterology, King’s College Hospital, London SE5 9RS, United Kingdom
| | - John K Ramage
- Neuroendocrine Tumour Unit, King’s College Hospital ENETS Centre of Excellence, London SE5 9RS, United Kingdom
| |
Collapse
|
173
|
Umesalma S, Kaemmer CA, Kohlmeyer JL, Letney B, Schab AM, Reilly JA, Sheehy RM, Hagen J, Tiwari N, Zhan F, Leidinger MR, O'Dorisio TM, Dillon J, Merrill RA, Meyerholz DK, Perl AL, Brown BJ, Braun TA, Scott AT, Ginader T, Taghiyev AF, Zamba GK, Howe JR, Strack S, Bellizzi AM, Narla G, Darbro BW, Quelle FW, Quelle DE. RABL6A inhibits tumor-suppressive PP2A/AKT signaling to drive pancreatic neuroendocrine tumor growth. J Clin Invest 2019; 129:1641-1653. [PMID: 30721156 DOI: 10.1172/jci123049] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 01/24/2019] [Indexed: 12/15/2022] Open
Abstract
Hyperactivated AKT/mTOR signaling is a hallmark of pancreatic neuroendocrine tumors (PNETs). Drugs targeting this pathway are used clinically, but tumor resistance invariably develops. A better understanding of factors regulating AKT/mTOR signaling and PNET pathogenesis is needed to improve current therapies. We discovered that RABL6A, a new oncogenic driver of PNET proliferation, is required for AKT activity. Silencing RABL6A caused PNET cell-cycle arrest that coincided with selective loss of AKT-S473 (not T308) phosphorylation and AKT/mTOR inactivation. Restoration of AKT phosphorylation rescued the G1 phase block triggered by RABL6A silencing. Mechanistically, loss of AKT-S473 phosphorylation in RABL6A-depleted cells was the result of increased protein phosphatase 2A (PP2A) activity. Inhibition of PP2A restored phosphorylation of AKT-S473 in RABL6A-depleted cells, whereas PP2A reactivation using a specific small-molecule activator of PP2A (SMAP) abolished that phosphorylation. Moreover, SMAP treatment effectively killed PNET cells in a RABL6A-dependent manner and suppressed PNET growth in vivo. The present work identifies RABL6A as a new inhibitor of the PP2A tumor suppressor and an essential activator of AKT in PNET cells. Our findings offer what we believe is a novel strategy of PP2A reactivation for treatment of PNETs as well as other human cancers driven by RABL6A overexpression and PP2A inactivation.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ryan M Sheehy
- Department of Pharmacology.,Free Radical & Radiation Biology Training Program
| | | | | | | | - Mariah R Leidinger
- Department of Pathology, in the College of Medicine, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa, USA
| | | | | | | | - David K Meyerholz
- Department of Pathology, in the College of Medicine, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa, USA
| | - Abbey L Perl
- Department of Pharmacology, Case Western Reserve University, Cleveland, Ohio, USA
| | | | | | | | | | - Agshin F Taghiyev
- Pediatrics, Colleges of Medicine, Engineering, or Public Health, University of Iowa, Iowa City, Iowa, USA
| | | | | | | | - Andrew M Bellizzi
- Department of Pathology, in the College of Medicine, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa, USA
| | - Goutham Narla
- Department of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Benjamin W Darbro
- Pediatrics, Colleges of Medicine, Engineering, or Public Health, University of Iowa, Iowa City, Iowa, USA
| | | | - Dawn E Quelle
- Department of Pharmacology.,Molecular Medicine Graduate Program.,Free Radical & Radiation Biology Training Program.,Department of Pathology, in the College of Medicine, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
174
|
Plasma Pancreastatin Predicts the Outcome of Surgical Cytoreduction in Neuroendocrine Tumors of the Small Bowel. Pancreas 2019; 48:356-362. [PMID: 30768573 DOI: 10.1097/mpa.0000000000001263] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVES Elevated pancreastatin (PST) levels have been shown to be associated with poor prognosis in small bowel neuroendocrine tumors (NETs). We hypothesized that plasma PST levels that remain elevated following surgical cytoreduction portend a poor prognosis in well-differentiated small bowel NETs. METHODS Patients diagnosed with small bowel NETs who underwent surgical cytoreduction at our institution were identified. Demographics, histopathologic characteristics, and biochemical data were collected. Only patients who had serial preoperative PST (PreopPST) and postoperative PST (PostopPST) levels were included in this study. Patients were sorted into groups by PST level to assess their response to surgical cytoreduction (group 1, PreopPST/PostopPST normal; group 2, PreopPST elevated/PostopPST normal; group 3, PreopPST/PostopPST elevated). Survival rates were calculated from the date of surgery. RESULTS PreopPST and PostopPST levels were collected from 300 patients. Patients in groups 1 (n = 74) and 2 (n = 81) had a significant survival advantage compared with patients in group 3 (n = 145) (P < 0.0001). Kaplan-Meier 5- and 10-year survival rates were as follows: group 1: 93% and 82%; group 2: 91% and 65%; and group 3: 58% and 34%, respectively. CONCLUSIONS Serial monitoring of plasma PST is useful in predicting long-term survival following surgical cytoreduction and can be helpful to identify patients who have a poor prognosis.
Collapse
|
175
|
Hofland J, Herrera-Martínez AD, Zandee WT, de Herder WW. Management of carcinoid syndrome: a systematic review and meta-analysis. Endocr Relat Cancer 2019; 26:R145-R156. [PMID: 30608900 DOI: 10.1530/erc-18-0495] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 01/03/2019] [Indexed: 12/18/2022]
Abstract
Carcinoid syndrome (CS) is a debilitating disease caused by functional neuroendocrine tumors. Several treatment options are available to alleviate the hormonal symptoms, but their relative efficacy is unknown. Online databases were searched for publications on the treatment of CS symptoms. Independent reviewers assessed relevant publications for study quality and outcome. Meta-analysis of the outcomes of the intervention on CS-related symptoms was stratified by the type of treatment. We found 3682 therapeutic interventions on CS-specific outcomes were collected from 93 studies. Overall, the study qualities were poor with only six randomized controlled clinical trials. The somatostatin analogs octreotide and lanreotide induced symptomatic improvement in 65-72% and biochemical response in 45-46% of patients. An increase in dose or frequency or interclass switch led to a reduction of flushes and/or diarrhea in 72-84% of cases. Retrospective, institutional series showed that liver-directed therapy can improve symptoms in 82% of CS patients with a liver-dominant disease. The serotonin synthesis inhibitor telotristat ethyl reduced bowel movements in 40% of patients with diarrhea refractory to somatostatin analogs. Interferon-alpha controlled CS symptoms in 45-63% of cases. Favorable response has been noted after radionuclide therapy in subgroup analyses of studies not specifically involving CS patients. Chemotherapy and everolimus did not induce a significant response in the CS. We conclude that several treatment lines can be offered to patients suffering from the carcinoid syndrome. Initiation of randomized controlled trials with a primary outcome on carcinoid syndrome symptoms is strongly recommended.
Collapse
Affiliation(s)
- Johannes Hofland
- ENETS Center of Excellence, Section of Endocrinology, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Aura D Herrera-Martínez
- ENETS Center of Excellence, Section of Endocrinology, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
- Endocrinology and Nutrition Service, Reina Sofia University Hospital, Córdoba, Spain
| | - Wouter T Zandee
- ENETS Center of Excellence, Section of Endocrinology, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Wouter W de Herder
- ENETS Center of Excellence, Section of Endocrinology, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| |
Collapse
|
176
|
Abstract
Carcinoid syndrome (CS) is a debilitating disease caused by functional neuroendocrine tumors. Several treatment options are available to alleviate the hormonal symptoms, but their relative efficacy is unknown. Online databases were searched for publications on the treatment of CS symptoms. Independent reviewers assessed relevant publications for study quality and outcome. Meta-analysis of the outcomes of the intervention on CS-related symptoms was stratified by the type of treatment. We found 3682 therapeutic interventions on CS-specific outcomes were collected from 93 studies. Overall, the study qualities were poor with only six randomized controlled clinical trials. The somatostatin analogs octreotide and lanreotide induced symptomatic improvement in 65–72% and biochemical response in 45–46% of patients. An increase in dose or frequency or interclass switch led to a reduction of flushes and/or diarrhea in 72–84% of cases. Retrospective, institutional series showed that liver-directed therapy can improve symptoms in 82% of CS patients with a liver-dominant disease. The serotonin synthesis inhibitor telotristat ethyl reduced bowel movements in 40% of patients with diarrhea refractory to somatostatin analogs. Interferon-alpha controlled CS symptoms in 45–63% of cases. Favorable response has been noted after radionuclide therapy in subgroup analyses of studies not specifically involving CS patients. Chemotherapy and everolimus did not induce a significant response in the CS. We conclude that several treatment lines can be offered to patients suffering from the carcinoid syndrome. Initiation of randomized controlled trials with a primary outcome on carcinoid syndrome symptoms is strongly recommended.
Collapse
|
177
|
Lee CHA, Holubar SD. Pelvic pouch cancers associated with inflammatory bowel disease. SEMINARS IN COLON AND RECTAL SURGERY 2019. [DOI: 10.1053/j.scrs.2019.01.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
178
|
Werner RA, Bundschuh RA, Bundschuh L, Fanti S, Javadi MS, Higuchi T, Weich A, Pienta KJ, Buck AK, Pomper MG, Gorin MA, Herrmann K, Lapa C, Rowe SP. Novel Structured Reporting Systems for Theranostic Radiotracers. J Nucl Med 2019; 60:577-584. [PMID: 30796171 DOI: 10.2967/jnumed.118.223537] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 12/29/2019] [Indexed: 12/13/2022] Open
Abstract
Standardized reporting is more and more routinely implemented in clinical practice, and such structured reports have a major impact on a large variety of medical fields, such as laboratory medicine, pathology, and, recently, radiology. Notably, the field of nuclear medicine is constantly evolving as novel radiotracers for numerous clinical applications are developed. Thus, framework systems for standardized reporting in this field may increase clinical acceptance of new radiotracers, allow for inter- and intracenter comparisons for quality assurance, and be used in global multicenter studies to ensure comparable results and enable efficient data abstraction. In the last couple of years, several standardized framework systems for PET radiotracers with potential theranostic applications have been proposed. These include systems for prostate-specific membrane antigen-targeted PET agents to diagnose and treat prostate cancer, and systems for somatostatin receptor-targeted PET agents to diagnose and treat neuroendocrine neoplasia. In the present review, the framework systems for these 2 types of cancer will be briefly introduced, followed by an overview of their advantages and limitations. In addition, potential applications will be defined, approaches to validate such concepts will be proposed, and future perspectives will be discussed.
Collapse
Affiliation(s)
- Rudolf A Werner
- Division of Nuclear Medicine and Molecular Imaging, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Nuclear Medicine/Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany.,European Neuroendocrine Tumor Society Center of Excellence, NET Zentrum, University Hospital Würzburg, Würzburg, Germany
| | - Ralph A Bundschuh
- Department of Nuclear Medicine, University Medical Center Bonn, Bonn, Germany
| | - Lena Bundschuh
- Department of Nuclear Medicine, University Medical Center Bonn, Bonn, Germany
| | - Stefano Fanti
- Nuclear Medicine Unit, University of Bologna, S. Orsola Hospital Bologna, Bologna, Italy
| | - Mehrbod S Javadi
- Division of Nuclear Medicine and Molecular Imaging, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Takahiro Higuchi
- Department of Nuclear Medicine/Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany.,Department of Bio Medical Imaging, National Cardiovascular and Cerebral Research Center, Suita, Japan
| | - Alexander Weich
- European Neuroendocrine Tumor Society Center of Excellence, NET Zentrum, University Hospital Würzburg, Würzburg, Germany.,Gastroenterology, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Kenneth J Pienta
- James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Andreas K Buck
- Department of Nuclear Medicine/Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany.,European Neuroendocrine Tumor Society Center of Excellence, NET Zentrum, University Hospital Würzburg, Würzburg, Germany
| | - Martin G Pomper
- Division of Nuclear Medicine and Molecular Imaging, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland.,James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Michael A Gorin
- Division of Nuclear Medicine and Molecular Imaging, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland.,James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ken Herrmann
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany; and.,Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Constantin Lapa
- Department of Nuclear Medicine/Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| | - Steven P Rowe
- Division of Nuclear Medicine and Molecular Imaging, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland .,James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
179
|
Thomas KEH, Voros BA, Boudreaux JP, Thiagarajan R, Woltering EA, Ramirez RA. Current Treatment Options in Gastroenteropancreatic Neuroendocrine Carcinoma. Oncologist 2019; 24:1076-1088. [PMID: 30635447 DOI: 10.1634/theoncologist.2018-0604] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 11/14/2018] [Indexed: 12/12/2022] Open
Abstract
Poorly differentiated gastroenteropancreatic neuroendocrine carcinomas (GEPNECs) are a rare neoplasm with a bleak prognosis. Currently there are little prospective data available for optimal treatment. This review discusses the current available regimens and the future direction for the treatment of GEPNECs. Treatment plans for GEPNECs are often adapted from those devised for small cell lung cancer; however, differences in these malignancies exist, and GEPNECs require their own treatment paradigms. As such, current first-line treatment for GEPNECs is platinum-based chemotherapy with etoposide. Studies show that response rate and overall survival remain comparable between cisplatin and carboplatin versus etoposide and irinotecan; however, prognosis remains poor, and more efficacious therapy is needed to treat this malignancy. Additional first-line and second-line treatment options beyond platinum-based chemotherapy have also been investigated and may offer further treatment options, but again with suboptimal outcomes. Recent U.S. Food and Drug Administration approval of peptide receptor radionuclide therapy in low- and intermediate-grade neuroendocrine tumors may open the door for further research in its usefulness in GEPNECs. Additionally, the availability of checkpoint inhibitors lends promise to the treatment of GEPNECs. This review highlights the lack of large, prospective studies that focus on the treatment of GEPNECs. There is a need for randomized control trials to elucidate optimal treatment regimens specific to this malignancy. IMPLICATIONS FOR PRACTICE: There are limited data available for the treatment of poorly differentiated gastroenteropancreatic neuroendocrine carcinomas (GEPNECs) because of the rarity of this malignancy. Much of the treatment regimens used in practice today come from research in small cell lung cancer. Given the poor prognosis of GEPNECs, it is necessary to have treatment paradigms specific to this malignancy. The aim of this literature review is to summarize the available first- and second-line GEPNEC therapy, outline future treatments, and highlight the vast gap in the literature.
Collapse
Affiliation(s)
- Katharine E H Thomas
- Department of Internal Medicine, Ochsner Medical Center, New Orleans, Louisiana, USA
| | - Brianne A Voros
- Department of Surgery, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
- Neuroendocrine Tumor Clinic, Ochsner Medical Center-Kenner, Kenner, Louisiana, USA
| | - J Philip Boudreaux
- Department of Surgery, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
- Neuroendocrine Tumor Clinic, Ochsner Medical Center-Kenner, Kenner, Louisiana, USA
| | - Ramcharan Thiagarajan
- Department of Surgery, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
- Neuroendocrine Tumor Clinic, Ochsner Medical Center-Kenner, Kenner, Louisiana, USA
| | - Eugene A Woltering
- Department of Surgery, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
- Neuroendocrine Tumor Clinic, Ochsner Medical Center-Kenner, Kenner, Louisiana, USA
| | - Robert A Ramirez
- Division of Hematology/Oncology, Department of Internal Medicine, Ochsner Medical Center, New Orleans, Louisiana, USA
- Neuroendocrine Tumor Clinic, Ochsner Medical Center-Kenner, Kenner, Louisiana, USA
| |
Collapse
|
180
|
Viknesh M, Bin Hisham F, Sankaran P, Kong Choon S, Azim Bin Nik Abdullah N. Bleeding and malignant pancreatic neuroendocrine neoplasms: A Malaysian case series. INTERNATIONAL JOURNAL OF HEPATOBILIARY AND PANCREATIC DISEASES 2019. [DOI: 10.5348/100084z04mv2019cs] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
181
|
Sigel CS. Advances in the cytologic diagnosis of gastroenteropancreatic neuroendocrine neoplasms. Cancer Cytopathol 2018; 126:980-991. [PMID: 30485690 DOI: 10.1002/cncy.22073] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 09/18/2018] [Accepted: 09/18/2018] [Indexed: 02/01/2023]
Abstract
Two-thirds of neuroendocrine neoplasms arising in the human body originate from the gastrointestinal system or pancreas. Gastroenteropancreatic neuroendocrine neoplasms are heterogeneous, comprising both well differentiated neuroendocrine tumors (NETs) and poorly differentiated neuroendocrine carcinomas (NECs). The clinical presentation, molecular characteristics, and behavior are distinct for NETs and NECs. Fine-needle aspiration is an important modality for the primary diagnosis and staging of these neoplasms and can provide information of prognostic and therapeutic significance. Our evolving understanding of neuroendocrine neoplasm biology has led to several iterations of classification. In this review, new concepts and issues most relevant to cytology diagnosis of gastroenteropancreatic neuroendocrine neoplasms are discussed, such as newer detection methods that aid in diagnosis and staging, recent changes in World Health Organization classification, practical issues related to grading these neoplasms on cytology, guidelines for diagnostic reporting, and panels of immunohistochemical stains for the diagnosis of metastasis. The current understanding of genetic and epigenetic events related to tumor development and potential applications for cytology also are presented as they relate to prognostication and recent therapeutic advances.
Collapse
Affiliation(s)
- Carlie S Sigel
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
182
|
Somatostatin receptor SSTR2A and SSTR5 expression in neuroendocrine breast cancer. Ann Diagn Pathol 2018; 38:62-66. [PMID: 30476894 DOI: 10.1016/j.anndiagpath.2018.11.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 11/18/2018] [Indexed: 12/31/2022]
Abstract
Neuroendocrine breast cancer (NEBC) is a group of rare tumors, which could benefit from therapy targeting the somatostatin receptors (SSTRs). In particular, SSTR2A and SSTR5 are potential targets given their consistent expression in gastrointestinal and pancreatic primary and metastatic neuroendocrine cancers. Currently, there are no studies describing the expression of SSTRs in NEBC. The purpose of our study was to characterize the immunohistochemical expression of SSTR2A and SSTR5 in a cohort of NEBC. Thirty-one primary NEBC cases were analyzed, and SSTR2A and SSTR5 immunohistochemistry performed and scored using the modified immunoreactive score proposed by Remmele and Stanger. All patients were females with a mean age of 66.6 years (SD = 14). 77% of cases were histological grade 2. SSTR2A showed a weak positivity in 11 cases (35.5%), moderate positivity in 6 cases (19.4%) and strong positivity in 5 cases (16.1%). Nine cases were negative for SSTR2A (29%). SSTR5 showed a weak positivity in 16 cases (51.6%), moderate positivity in 6 cases (19.4%), while no cases showed strong positivity. Nine cases were negative for SSTR5 (29%). Five cases were negative for both SSTR2A and SSTR5. A weak to moderate SSTR2A and SSTR5 expression was observed in 50-70% of the cases. A subset of NEBCs with strong SSR2A expression may benefit from SSTRs targeted therapy. These results need further validation in a larger series including metastatic NEBC, to provide potential therapeutic targets for patients with advanced disease.
Collapse
|
183
|
Abstract
OPINION STATEMENT Small bower cancer is a rare disease, despite its incidence is increasing in the last decade. Both benign and malignant tumors can arise from the small intestine. The main histological cancer types are adenocarcinomas, neuroendocrine tumors, sarcomas, gastrointestinal stromal tumors (GISTs), and lymphomas. Due to the rarity of these malignances, all the currently available data are based on small studies or retrospective series, although recent breakthroughs are redirecting our approach to these patients. Immunotherapy for small bowel adenocarcinomas, several multikinase inhibitors in resistant GIST patients, as well as everolimus and 177Lu-DOTATATE in neuroendocrine tumors are only few of the novel therapeutic options that have changed, or may change in the future, the therapeutic landscape of these rare cancers. Larger and more powerful studies on the molecular profile of these tumors may lead to a better design of clinical trials, which eventually would provide our patients with more efficacious treatments to improve both overall survival and quality of life.
Collapse
Affiliation(s)
- Alberto Puccini
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, 1441 Eastlake Avenue, Suite 3456, Los Angeles, CA, 90033, USA.,Department of Medical Oncology, Ospedale Policlinico San Martino, Genoa, Italy
| | - Francesca Battaglin
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, 1441 Eastlake Avenue, Suite 3456, Los Angeles, CA, 90033, USA.,Medical Oncology Unit 1, Clinical and Experimental Oncology Department, Veneto Institute of Oncology IOV-IRCCS, 35128, Padua, Italy
| | - Heinz-Josef Lenz
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, 1441 Eastlake Avenue, Suite 3456, Los Angeles, CA, 90033, USA.
| |
Collapse
|
184
|
Pavel ME, Phan AT, Wolin EM, Mirakhur B, Liyanage N, Pitman Lowenthal S, Fisher GA, Vinik AI. Effect of Lanreotide Depot/Autogel on Urinary 5-Hydroxyindoleacetic Acid and Plasma Chromogranin A Biomarkers in Nonfunctional Metastatic Enteropancreatic Neuroendocrine Tumors. Oncologist 2018; 24:463-474. [PMID: 30355775 DOI: 10.1634/theoncologist.2018-0217] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 08/24/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Urinary 5-hydroxyindoleacetic acid (5-HIAA) is an established biomarker in neuroendocrine tumors and carcinoid syndrome; however, its role in nonfunctional neuroendocrine tumors is not defined. We present post hoc data on urinary 5-HIAA and plasma chromogranin A (CgA) from the CLARINET study. METHODS Patients with well- or moderately differentiated, nonfunctioning, locally advanced or metastatic enteropancreatic neuroendocrine tumors were randomized to deep subcutaneous lanreotide depot/autogel 120 mg or placebo once every 28 days for 96 weeks. Tumor response, evaluated centrally (RECIST 1.0), and progression-free survival (PFS) were assessed by treatment and biochemical response, defined as (a) baseline >upper limit of normal (ULN, 41.6 μmol per day 5-HIAA; 98.1 μg/L CgA) and (b) ≥50% decrease from baseline and to ≤ULN value on study. RESULTS Forty-eight percent (82 of 171; lanreotide, n = 45; placebo, n = 37) and 66% (129 of 195; lanreotide, n = 65; placebo, n = 64) of randomized patients had 5-HIAA and CgA > ULN at baseline. Among patients with >ULN baseline values who did not progress after 96 weeks of treatment, significantly greater reductions in 5-HIAA and CgA were observed in lanreotide-treated versus placebo-treated patients throughout the study (all p < .05). PFS was significantly prolonged among 5-HIAA responders versus nonresponders (median not reached vs. 16.2 months, p < .0001; hazard ratio [HR] = 0.21, 95% confidence interval [CI], 0.09-0.48) and CgA responders versus nonresponders (median not reached vs. 16.2 months, p = .0070; HR = 0.30, 95% CI, 0.12-0.76), regardless of treatment arm. PFS was also significantly prolonged among lanreotide-treated 5-HIAA responders versus nonresponders (p = .0071) but was not significantly different among placebo-treated 5-HIAA responders versus nonresponders. There were no significant differences in PFS between lanreotide-treated CgA responders versus nonresponders or between placebo-treated CgA responders versus nonresponders. CONCLUSIONS The 5-HIAA findings are noteworthy because they occurred in patients with nonfunctioning enteropancreatic neuroendocrine tumors. Monitoring 5-HIAA and CgA may be useful when treating patients with nonfunctional neuroendocrine tumors. IMPLICATIONS FOR PRACTICE Current guidelines focus only on the monitoring of 5-hydroxyindoleacetic acid (5-HIAA) in the diagnosis and management of functional neuroendocrine tumors with carcinoid syndrome. The current post hoc analysis of patients with nonfunctional enteropancreatic neuroendocrine tumors in the CLARINET study demonstrated that measuring and following both 5-HIAA and chromogranin A as biomarkers of disease progression may be useful in the management of patients with nonfunctional neuroendocrine tumors.
Collapse
Affiliation(s)
- Marianne E Pavel
- Charité University Medicine Berlin, Berlin, Germany
- Friedrich Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Alexandria T Phan
- University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico, USA
| | - Edward M Wolin
- Tisch Cancer Institute, Mount Sinai School of Medicine, New York New York, USA
| | - Beloo Mirakhur
- Ipsen Biopharmaceuticals, Basking Ridge, New Jersey, USA
| | | | | | - George A Fisher
- Stanford University School of Medicine, Stanford, California, USA
| | - Aaron I Vinik
- Eastern Virginia Medical School, Norfolk, Virginia, USA
| |
Collapse
|
185
|
Sackstein PE, O'Neil DS, Neugut AI, Chabot J, Fojo T. Epidemiologic trends in neuroendocrine tumors: An examination of incidence rates and survival of specific patient subgroups over the past 20 years. Semin Oncol 2018; 45:249-258. [PMID: 30348533 DOI: 10.1053/j.seminoncol.2018.07.001] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 07/25/2018] [Indexed: 02/03/2023]
Abstract
INTRODUCTION Neuroendocrine tumors (NETs) represent a small proportion of cancers, but are increasing in incidence due to incidental diagnosis. We examined NET incidence and survival over time in a population-based registry. MATERIALS/METHODS We identified all NET cases diagnosed between 1995 and 2014 in the Surveillance, Epidemiology, and End Results database, November 2016 submission. We determined incidence rates and calculated overall and cancer-specific survival curves in different subgroups stratified by grade, stage, and age at diagnosis. RESULTS We identified 85,133 patients with a diagnosis of NET between 1995 and 2014. Patients with grade 1, localized NETs had the best median overall survival (233 months, 95% confidence intervals [CI] not estimable) and 5-year cancer-specific survival (97.6%; 95% CI, 97.4%, 97.8%). The median overall survival decreased with age across the entire spectrum of ages, with patients >70 years having a particularly poor prognosis (28.0 months; 95% CI, 26.5, 29.5). Patients >70 years old often had distant (34.3%) or grade 3 disease (40.8%), but even elderly patients with lower grade and/or stage disease had worse median overall survival compared with younger subjects. CONCLUSIONS Age appears to be associated with a worse prognosis independent of NET stage, and grade at the time of diagnosis. Patients with grade 1, localized NETs have an excellent long-term prognosis. Further research is warranted on reducing intensity of surveillance in these patients.
Collapse
Affiliation(s)
- Paul E Sackstein
- University of Connecticut School of Medicine, UConn Health, Farmington, CT, USA
| | - Daniel S O'Neil
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center/New York-Presbyterian Hospital, New York, NY, USA; Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA.
| | - Alfred I Neugut
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center/New York-Presbyterian Hospital, New York, NY, USA; Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - John Chabot
- Department of Surgery, Pancreas Center, Columbia University, New York, NY, USA
| | - Tito Fojo
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center/New York-Presbyterian Hospital, New York, NY, USA
| |
Collapse
|
186
|
Safety and Feasibility of Integrating Yttrium-90 Radioembolization With Capecitabine-Temozolomide for Grade 2 Liver-Dominant Metastatic Neuroendocrine Tumors. Pancreas 2018; 47:980-984. [PMID: 30028446 DOI: 10.1097/mpa.0000000000001115] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
OBJECTIVES An integrated protocol combining capecitibine-temozolomide with yttrium-90 radioembolization (CapTemY90) for liver-dominant grade 2 neuroendocrine tumors (NETs) was designed in the hope of achieving synergistic improvement in liver disease control with no more than additive toxicities. This report describes the feasibility and safety of this regimen. METHODS Twenty-one patients with unresectable grade 2 NET liver-dominant metastases without contraindications to radioembolization or to CapTem initiated therapy with capecitabine 600 mg/m twice daily for 14 days and temozolomide 150 to 200 mg/m in 2 divided doses on days 10 to 14, with 14 days between cycles. During the first cycle, simulation angiography was performed. The dominant lobe was radioembolized on day 7 of the second cycle. In patients with bilobar disease, the other lobe was treated on day 7 of the third or fourth cycle. RESULTS Nineteen of 21 patients completed the protocol. Adverse events were as expected. Objective response rate was 74% in the liver and 55% for extrahepatic tumor. Median progression-free survival was not reached. Progression-free survival at 3 years was 67%, with 74% progression-free in the liver. CONCLUSIONS CapTemY90 is feasible and safe for grade 2 NETs. Toxicities were additive. Oncologic outcomes suggest synergy.
Collapse
|
187
|
Abstract
Neuroendocrine tumors, including carcinoids, are rare and insidiously growing tumors. Related to their site of origin, tumors can be functional, causing various forms of the carcinoid syndrome, owing to the overproduction of serotonin, histamine, or other bioactive substances. They often invade adjacent structures or metastasize to the liver and elsewhere. Treatment includes multimodal approaches, including cytoreductive surgery, locoregional embolization, cytotoxic therapy, peptide receptor radionuclide therapy, and various targeted therapies with goals of symptom relief and control of tumor growth. This article summarizes current and emerging approaches to management and reviews several promising future therapies.
Collapse
Affiliation(s)
- Paul Benjamin Loughrey
- Department of Ophthalmology, Royal Victoria Hospital, Belfast Trust, Grosvenor Road, Belfast, BT12 6BA, UK; Department of Endocrinology and Diabetes, Royal Victoria Hospital, Belfast Trust, Grosvenor Road, Belfast, BT12 6BA, UK
| | - Dongyun Zhang
- Department of Medicine, David Geffen School of Medicine, University of California, 700 Tiverton Avenue, Los Angeles, CA 90095, USA
| | - Anthony P Heaney
- Department of Medicine, David Geffen School of Medicine, University of California, 700 Tiverton Avenue, Los Angeles, CA 90095, USA; Department of Neurosurgery, David Geffen School of Medicine, University of California, 700 Tiverton Avenue, Los Angeles, CA 90095, USA.
| |
Collapse
|
188
|
Liu E, Paulson S, Gulati A, Freudman J, Grosh W, Kafer S, Wickremesinghe PC, Salem RR, Bodei L. Assessment of NETest Clinical Utility in a U.S. Registry-Based Study. Oncologist 2018; 24:783-790. [PMID: 30158287 DOI: 10.1634/theoncologist.2017-0623] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 04/09/2018] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND The clinical relevance of molecular biomarkers in oncology management has been recognized in breast and lung cancers. We evaluated a blood-based multigene assay for management of neuroendocrine tumors (NETs) in a real-world study (U.S. registry NCT02270567). Diagnostic accuracy and relationship to clinical disease status in two cohorts (treated and watch-and-wait) were evaluated. MATERIALS AND METHODS Patients with NETs (n = 100) were followed for 6-12 months. Patients' primary tumors were gastroenteropancreatic (68%), lung 20%, and of unknown origin (12%). Characteristics included well-differentiated, low-grade tumors (97%), stage IV disease (96%); treatment with surgery (70%); and drug treatment (56%). NETest was measured at each visit and disease status determined by RECIST. Scores categorized as low (NETest 14%-40%) or high (≥80%) defined disease as stable or progressive. Multivariate analyses determined the strength of the association with progression-free survival (PFS). RESULTS NETest diagnostic accuracy was 96% and concordant (95%) with image-demonstrable disease. Scores were reproducible (97%) and concordant with clinical status (98%). The NETest was the only feature linked to PFS (odds ratio, 6.1; p < .0001). High NETest correlated with progressive disease (81%; median PFS, 6 months), and low NETest correlated with stable disease (87%; median PFS, not reached). In the watch-and-wait cohort, low NETest was concordant with stable disease in 100% of patients, and high NETest was associated with management changes in 83% of patients. In the treated cohort, all low NETest patients (100%) remained stable. A high NETest was linked to intervention and treatment stabilization (100%). Use of NETest was associated with reduced imaging (biannual to annual) in 36%-38% of patients. CONCLUSION Blood NETest is an accurate diagnostic and can be of use in monitoring disease status and facilitating management change in both watch-and-wait and treatment cohorts. IMPLICATIONS FOR PRACTICE A circulating multigene molecular biomarker to guide neuroendocrine tumor (NET) management has been developed because current biomarkers have limited clinical utility. NETest is diagnostic (96%) and in real time defines the disease status (>95%) as stable or progressive. It is >90% effective in guiding treatment decisions in conjunction with diagnostic imaging. Monitoring was effective in watch-and-wait or treatment groups. Low levels supported no management change and reduced the need for imaging. High levels indicated the need for management intervention. Real-time liquid biopsy assessment of NETs has clinical utility and can contribute additional value to patient management strategies and outcomes.
Collapse
Affiliation(s)
- Eric Liu
- Rocky Mountain Cancer Center, Denver, Colorado, USA
| | - Scott Paulson
- Texas Oncology - Baylor Charles A Sammons Cancer Center, Dallas, Texas, USA
| | | | - Jon Freudman
- Freudman Healthcare Consulting, San Rafael, California, USA
| | - William Grosh
- Emily Couric Cancer Center, University of Virginia, Charlottesville, Virginia, USA
| | | | | | - Ronald R Salem
- Yale University School of Medicine, Department of Surgery, New Haven, Connecticut, USA
| | - Lisa Bodei
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
189
|
Gabriel M, Nilica B, Kaiser B, Virgolini IJ. Twelve-Year Follow-up After Peptide Receptor Radionuclide Therapy. J Nucl Med 2018; 60:524-529. [PMID: 30115690 DOI: 10.2967/jnumed.118.215376] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 08/10/2018] [Indexed: 02/01/2023] Open
Abstract
Peptide receptor radionuclide therapy (PRRT) has been used for more than 20 y as a systemic treatment approach in inoperable or metastatic somatostatin receptor-positive tumors. The purpose of this study was to analyze the long-term outcome of PRRT with regard to the most commonly used radiopharmaceuticals, 90Y-DOTATOC and 177Lu-DOTATATE. Methods: This retrospective clinical study included a total of 44 consecutive patients (27 men) with advanced tumors and enhanced somatostatin receptor expression. Mean age at initial diagnosis was 60 y (SD, 11.3 y; range, 40-84 y). Median follow-up was 80 mo. For 177Lu-PRRT, the mean number of cycles administered was 5.3 ± 2.5 and the mean activity was 27.2 ± 14.9 GBq per patient. For 90Y-PRRT, the mean number of cycles administered was 5.5 ± 2.6 and the mean activity was 14.7 ± 7.3 GBq per patient. Overall, 378 cycles were administered (mean, 8.6 ± 3.4 cycles per patient), with an overall cumulative activity of 1,514.1 GBq. Results: Median overall survival was 79 mo. Twenty-one (77.8%) of the 27 men and 9 (52.9%) of the 17 women had died 12 y after commencement of PRRT. The shortest duration of illness was 8 mo and the longest 155 mo. Severe side effects (World Health Organization grades III and IV) were seen in 9 of the 14 patients still alive. Chronic kidney disease in combination with anemia was the most common finding in the 9 patients with severe side effects. A poor prognosis was found for those patients who showed progressive disease, in comparison with patients with cumulative disease control after initial PRRT (log rank, P < 0.001), whereas women and patients with no more than 2 tumor sites seemed to especially benefit from PRRT (not reaching significance levels). Conclusion: PRRT is encouraging in terms of long-term outcome. Thirty-two percent (14/44 patients) of the patients with metastatic or inoperable disease were still alive more than 12 y after the beginning of radionuclide therapy. Possible predictors for favorable outcome are having an initial response to PRRT, having a low number of affected sites, and being female.
Collapse
Affiliation(s)
- Michael Gabriel
- Department of Nuclear Medicine, Medical University of Innsbruck, Innsbruck, Austria; and .,Institute of Nuclear Medicine and Endocrinology, Kepler University Hospital, Linz, Austria
| | - Bernhard Nilica
- Department of Nuclear Medicine, Medical University of Innsbruck, Innsbruck, Austria; and
| | - Bernhard Kaiser
- Institute of Nuclear Medicine and Endocrinology, Kepler University Hospital, Linz, Austria
| | - Irene J Virgolini
- Department of Nuclear Medicine, Medical University of Innsbruck, Innsbruck, Austria; and
| |
Collapse
|
190
|
Abstract
OBJECTIVE Endocrine tumor markers (ETMs) are important and indispensable tools in the diagnosis and follow-up of endocrine tumors. Unexpectedly high level of ETM (UHLETM) is often encountered in clinical practice. The objectives of this article are to discuss the approach to UHLETMs and describe the most common UHLETMs. METHODS Literature review and personal experience with UHLETMs. RESULTS A UHLETM is defined as an ETM level much higher than what an endocrinologist would expect based on the patient's clinical information, other biochemical test results, and imaging findings. Most UHLETMs are false positive, in that they do not indicate the existence or growth of an endocrine tumor. The key issue, however, is how to convincingly prove that a UHLETM is false positive. The most important question to help resolve UHLETMs is whether the UHLETMs are due to false or true results. Some UHLETMs are due to interpretation errors, laboratory errors, or spurious test results, while the true levels of the ETMs are normal or unchanged from previous results. Other UHLETMs are due to nontumor conditions or medications, and the true levels of the ETMs are indeed very high. When it is not straightforward to assess whether certain UHLETMs are due to false or true results, they may be due to novel conditions. CONCLUSION UHLETMs provide endocrinologists great opportunities to learn the basic biology and measurement of ETMs. Unresolved UHLETMs are exciting clinical research opportunities which may lead to discovery of new diseases and new mechanisms of measurement interference. ABBREVIATIONS 5-HIAAA = 5-hydroxyindoleacetic acid; 5-HTP = 5-hydroxytryptophan; CGA = chromogranin A; ETM = endocrine tumor marker; MTC = medullary thyroid carcinoma; UHLETM = unexpectedly high level of endocrine tumor marker.
Collapse
|
191
|
Abstract
OBJECTIVES Elevated neurokinin A (NKA) levels are associated with poor prognosis in patients with small bowel neuroendocrine tumors. We hypothesized that patients with NKA levels that remain elevated despite treatment with surgical cytoreduction have a poor prognosis. METHODS Patients diagnosed with small bowel neuroendocrine tumors who underwent surgical cytoreduction at our institution were identified. Demographics, histopathologic characteristics, and biochemical data were collected. Patients were grouped by the trend of their NKA levels (group 1, continuously normal; group 2, transiently elevated but normalized after therapy; group 3, remained elevated despite therapy). Survival rates were calculated from the date of the patient's first NKA level. RESULTS Serial NKA values after surgical cytoreduction were monitored in 267 patients. Kaplan-Meier 2-year, 5-year, and 10-year survival rates were as follows: group 1 (n = 157), 97%, 89%, and 62%; group 2 (n = 78), 99%, 90%, and 78%; and group 3 (n = 32), 88%, 69%, and 0%. Survival rates were statistically significant between groups 1 and 3 and between groups 2 and 3 (P < 0.01). CONCLUSIONS Serial monitoring of plasma NKA levels is useful in identifying patients who have a poor prognosis. Elevated NKA levels can indicate the need for immediate therapeutic intervention.
Collapse
|
192
|
Abstract
Small bowel neuroendocrine tumors (NETs) are increasing in incidence and are now the most common primary malignancies of the small intestine. Despite this increase, the vague presentation and slow growth of these tumors lead to long delays in diagnosis, and many patients present with metastases. Patients with metastatic small bowel NETs have a favorable disease prognosis, particularly when contrasted with other GI malignancies, and benefit from aggressive, multimodal therapy. During the past decade, the options for the diagnosis and treatment of small bowel NETs have increased considerably. This review provides a practical framework for the physician who seek to understand the epidemiology, presentation, diagnosis, and management of small bowel NETs.
Collapse
Affiliation(s)
- Aaron T. Scott
- University of Iowa Carver College of Medicine, Iowa City, IA
| | - James R. Howe
- University of Iowa Carver College of Medicine, Iowa City, IA
| |
Collapse
|
193
|
Jin XF, Spampatti MP, Spitzweg C, Auernhammer CJ. Supportive therapy in gastroenteropancreatic neuroendocrine tumors: Often forgotten but important. Rev Endocr Metab Disord 2018; 19:145-158. [PMID: 29464446 DOI: 10.1007/s11154-018-9443-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Neuroendocrine tumors (NETs) are a group of rare and heterogeneous malignancies that can develop in various organs. A significant number of gastroenteropancreatic neuroendocrine tumours (GEP-NETs) is functionally active and presents with symptoms related to the secretion of biologically active substances, leading to the development of distinct clinical syndromes. There are various therapeutic approaches for GEP-NETs, including curative surgery, palliative surgery, local-ablative and loco-regional therapies as well as systemic therapeutic options including peptide receptor radionuclide therapy, cytotoxic therapy, and molecularly targeted therapies. Specific supportive therapy of patients with NETs includes management or prevention of hormone-related clinical syndromes and paraneoplastic states. Supportive therapy plays a key role in NET treatment. Supportive therapy includes debulking surgery and interventional radiologic techniques to reduce tumour bulk or load, as well as systemic medical treatment options to manage or prevent hypersecretion syndromes and treatment-related side effects. Supportive therapies are a type of of comprehensive treatment addressing the patient as a whole person throughout the process of NET treatment. Therefore, supportive therapy also encompasses psychosocial support, expert nursing, nutritional support and management of cancer related pain.
Collapse
Affiliation(s)
- Xi-Feng Jin
- Department of Internal Medicine IV, University-Hospital Campus Grosshadern, Ludwig-Maximilian University of Munich, Munich, Germany
| | - Matilde P Spampatti
- Department of Internal Medicine II, University-Hospital Campus Grosshadern, Ludwig-Maximilian University of Munich, Munich, Germany
- Interdisciplinary Center of Neuroendocrine Tumours of the GastroEnteroPancreatic System (GEPNET-KUM), Klinikum der Universitaet Muenchen, Ludwig-Maximilians-University of Munich, Campus Grosshadern, Marchioninistr, 15, 81377, Munich, Germany
| | - Christine Spitzweg
- Department of Internal Medicine IV, University-Hospital Campus Grosshadern, Ludwig-Maximilian University of Munich, Munich, Germany
- Interdisciplinary Center of Neuroendocrine Tumours of the GastroEnteroPancreatic System (GEPNET-KUM), Klinikum der Universitaet Muenchen, Ludwig-Maximilians-University of Munich, Campus Grosshadern, Marchioninistr, 15, 81377, Munich, Germany
| | - Christoph J Auernhammer
- Department of Internal Medicine IV, University-Hospital Campus Grosshadern, Ludwig-Maximilian University of Munich, Munich, Germany.
- Interdisciplinary Center of Neuroendocrine Tumours of the GastroEnteroPancreatic System (GEPNET-KUM), Klinikum der Universitaet Muenchen, Ludwig-Maximilians-University of Munich, Campus Grosshadern, Marchioninistr, 15, 81377, Munich, Germany.
| |
Collapse
|
194
|
Ariotti R, Partelli S, Muffatti F, Andreasi V, Della Sala F, Falconi M. How should incidental NEN of the pancreas and gastrointestinal tract be followed? Rev Endocr Metab Disord 2018. [PMID: 29527619 DOI: 10.1007/s11154-018-9445-4] [Citation(s) in RCA: 5] [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] [Indexed: 02/07/2023]
Abstract
Neuroendocrine gastro-entero-pancreatic neoplasms (GEP-NENs) constitute a heterogeneous group of tumors, whose incidence has increased over the years. The most frequent site for primary disease is the stomach followed by small and large intestine, and pancreas. In the last decade, a dramatic growing in the incidence of small, incidental GEP-NENs has been recorded. In parallel, an increasing attitude toward more conservative approaches instead of surgical management has being widely spreading. This is particularly true for small, asymptomatic, pancreatic NEN as for these tumor forms an active surveillance has proven to be safe and feasible. Primary site and biological features of the neoplasms lead to different strategies and indications for surveillance and follow-up. This review focuses on the current evidence on modality and timing of surveillance and conservative treatment of incidentally discovered lesions.
Collapse
Affiliation(s)
- Riccardo Ariotti
- Pancreatic Surgery Unit, Pancreas Translational & Clinical Research Center, San Raffaele Scientific Institute, "Vita-Salute" University, Via Olgettina 60, 20132, Milan, Italy
| | - Stefano Partelli
- Pancreatic Surgery Unit, Pancreas Translational & Clinical Research Center, San Raffaele Scientific Institute, "Vita-Salute" University, Via Olgettina 60, 20132, Milan, Italy
| | - Francesca Muffatti
- Pancreatic Surgery Unit, Pancreas Translational & Clinical Research Center, San Raffaele Scientific Institute, "Vita-Salute" University, Via Olgettina 60, 20132, Milan, Italy
| | - Valentina Andreasi
- Pancreatic Surgery Unit, Pancreas Translational & Clinical Research Center, San Raffaele Scientific Institute, "Vita-Salute" University, Via Olgettina 60, 20132, Milan, Italy
| | - Francesca Della Sala
- Pancreatic Surgery Unit, Pancreas Translational & Clinical Research Center, San Raffaele Scientific Institute, "Vita-Salute" University, Via Olgettina 60, 20132, Milan, Italy
| | - Massimo Falconi
- Pancreatic Surgery Unit, Pancreas Translational & Clinical Research Center, San Raffaele Scientific Institute, "Vita-Salute" University, Via Olgettina 60, 20132, Milan, Italy.
| |
Collapse
|
195
|
Lee L, Ito T, Jensen RT. Everolimus in the treatment of neuroendocrine tumors: efficacy, side-effects, resistance, and factors affecting its place in the treatment sequence. Expert Opin Pharmacother 2018; 19:909-928. [PMID: 29757017 PMCID: PMC6064188 DOI: 10.1080/14656566.2018.1476492] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Since the initial approval of everolimus in 2011, there have been a number of important changes in therapeutic/diagnostic modalities as well as classification/staging systems of neuroendocrine tumors (NETs), which can significantly impact the use of everolimus in patients with advanced NETs. Areas covered: The efficacy of everolimus monotherapy and combination therapy demonstrated in clinical studies involving patients with advanced NETs are reviewed. Several factors affecting everolimus use are described including: the development and routine use of NET classification/staging systems; widespread use of molecular imaging modalities; side effects; drug resistance; and the availability of other treatment options. Furthermore, the current position of everolimus in the treatment approach is discussed, taking into account the recommendations from the recent guidelines. Expert opinion: Although everolimus demonstrated its high efficacy and tolerability in the RADIANT trials and other clinical studies, there still remain a number of controversies related to everolimus treatment in the management of NETs. The synergistic anti-growth effect of other agents in combination with everolimus or its effect on overall survival have not been established. The appropriate order of the use of everolimus in the treatment of advanced NETs still remains unclear, which needs to be defined in further studies and will be addressed in the new guidelines.
Collapse
Affiliation(s)
- Lingaku Lee
- a Digestive Diseases Branch , National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health , Bethesda , MD , USA
| | - Tetsuhide Ito
- b Neuroendocrine Tumor Centre , Fukuoka Sanno Hospital, International University of Health and Welfare , Fukuoka , Japan
| | - Robert T Jensen
- a Digestive Diseases Branch , National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health , Bethesda , MD , USA
| |
Collapse
|
196
|
Auernhammer CJ, Spitzweg C, Angele MK, Boeck S, Grossman A, Nölting S, Ilhan H, Knösel T, Mayerle J, Reincke M, Bartenstein P. Advanced neuroendocrine tumours of the small intestine and pancreas: clinical developments, controversies, and future strategies. Lancet Diabetes Endocrinol 2018; 6:404-415. [PMID: 29229497 DOI: 10.1016/s2213-8587(17)30401-1] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Revised: 10/14/2017] [Accepted: 10/18/2017] [Indexed: 12/18/2022]
Abstract
In this Review, we discuss clinical developments and controversies in the treatment of neuroendocrine tumours (NETs) that are relevant for clinicians and clinical researchers. We describe advances in genetics, blood-based biomarkers, functional imaging, and systemic therapy of advanced NETs and discuss results of recent phase 3 studies, systemic treatment of advanced disease with peptide receptor radionuclide therapy, biotherapy, chemotherapy, and molecularly targeted therapy, and the potential role of immunotherapy in the treatment of NETs. Suggested treatment algorithms for NETs of ileal or jejunal origin and of pancreatic origin are presented.
Collapse
Affiliation(s)
- Christoph J Auernhammer
- Interdisciplinary Center of Neuroendocrine Tumours of the GastroEnteroPancreatic System, Ludwig-Maximilians-University of Munich, Munich, Germany; Department of Internal Medicine 4, Ludwig-Maximilians-University of Munich, Munich, Germany.
| | - Christine Spitzweg
- Interdisciplinary Center of Neuroendocrine Tumours of the GastroEnteroPancreatic System, Ludwig-Maximilians-University of Munich, Munich, Germany; Department of Internal Medicine 4, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Martin K Angele
- Interdisciplinary Center of Neuroendocrine Tumours of the GastroEnteroPancreatic System, Ludwig-Maximilians-University of Munich, Munich, Germany; Department of General, Visceral, Transplantation, Vascular and Thoracic Surgery, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Stefan Boeck
- Interdisciplinary Center of Neuroendocrine Tumours of the GastroEnteroPancreatic System, Ludwig-Maximilians-University of Munich, Munich, Germany; Department of Internal Medicine 3, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Ashley Grossman
- Neuroendocrine Tumour Centre, Royal Free Hospital, London, UK
| | - Svenja Nölting
- Interdisciplinary Center of Neuroendocrine Tumours of the GastroEnteroPancreatic System, Ludwig-Maximilians-University of Munich, Munich, Germany; Department of Internal Medicine 4, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Harun Ilhan
- Interdisciplinary Center of Neuroendocrine Tumours of the GastroEnteroPancreatic System, Ludwig-Maximilians-University of Munich, Munich, Germany; Department of Nuclear Medicine, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Thomas Knösel
- Interdisciplinary Center of Neuroendocrine Tumours of the GastroEnteroPancreatic System, Ludwig-Maximilians-University of Munich, Munich, Germany; Institute of Pathology, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Julia Mayerle
- Interdisciplinary Center of Neuroendocrine Tumours of the GastroEnteroPancreatic System, Ludwig-Maximilians-University of Munich, Munich, Germany; Department of Internal Medicine 2, Klinikum der Universitaet Muenchen, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Martin Reincke
- Interdisciplinary Center of Neuroendocrine Tumours of the GastroEnteroPancreatic System, Ludwig-Maximilians-University of Munich, Munich, Germany; Department of Internal Medicine 4, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Peter Bartenstein
- Interdisciplinary Center of Neuroendocrine Tumours of the GastroEnteroPancreatic System, Ludwig-Maximilians-University of Munich, Munich, Germany; Department of Nuclear Medicine, Ludwig-Maximilians-University of Munich, Munich, Germany
| |
Collapse
|
197
|
Merath K, Bagante F, Beal EW, Lopez-Aguiar AG, Poultsides G, Makris E, Rocha F, Kanji Z, Weber S, Fisher A, Fields R, Krasnick BA, Idrees K, Smith PM, Cho C, Beems M, Schmidt CR, Dillhoff M, Maithel SK, Pawlik TM. Nomogram predicting the risk of recurrence after curative-intent resection of primary non-metastatic gastrointestinal neuroendocrine tumors: An analysis of the U.S. Neuroendocrine Tumor Study Group. J Surg Oncol 2018; 117:868-878. [PMID: 29448303 PMCID: PMC5992105 DOI: 10.1002/jso.24985] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 01/01/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND The risk of recurrence after resection of non-metastatic gastro-entero-pancreatic neuroendocrine tumors (GEP-NET) is poorly defined. We developed/validated a nomogram to predict risk of recurrence after curative-intent resection. METHODS A training set to develop the nomogram and test set for validation were identified. The predictive ability of the nomogram was assessed using c-indices. RESULTS Among 1477 patients, 673 (46%) were included in the training set and 804 (54%) in y the test set. On multivariable analysis, Ki-67, tumor size, nodal status, and invasion of adjacent organs were independent predictors of DFS. The risk of death increased by 8% for each percentage increase in the Ki-67 index (HR 1.08, 95% CI, 1.05-1.10; P < 0.001). GEP-NET invading adjacent organs had a HR of 1.65 (95% CI, 1.03-2.65; P = 0.038), similar to tumors ≥3 cm (HR 1.67, 95% CI, 1.11-2.51; P = 0.014). Patients with 1-3 positive nodes and patients with >3 positive nodes had a HR of 1.81 (95% CI, 1.12-2.87; P = 0.014) and 2.51 (95% CI, 1.50-4.24; P < 0.001), respectively. The nomogram demonstrated good ability to predict risk of recurrence (c-index: training set, 0.739; test set, 0.718). CONCLUSION The nomogram was able to predict the risk of recurrence and can be easily applied in the clinical setting.
Collapse
Affiliation(s)
- Katiuscha Merath
- Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, Ohio
| | - Fabio Bagante
- Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, Ohio
- Department of Surgery, University of Verona, Verona, Italy
| | - Eliza W Beal
- Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, Ohio
| | - Alexandra G Lopez-Aguiar
- Division of Surgical Oncology, Department of Surgery, Winship Cancer Institute, Emory University, Atlanta, Georgia
| | | | | | - Flavio Rocha
- Department of Surgery, Virginia Mason Medical Center, Seattle, Washington
| | - Zaheer Kanji
- Department of Surgery, Virginia Mason Medical Center, Seattle, Washington
| | - Sharon Weber
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Alexander Fisher
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Ryan Fields
- Department of Surgery, Washington University School of Medicine, St. Louis, Wisconsin
| | - Bradley A Krasnick
- Department of Surgery, Washington University School of Medicine, St. Louis, Wisconsin
| | - Kamran Idrees
- Division of Surgical Oncology, Department of Surgery, Vanderbilt University, Nashville, Tennessee
| | - Paula M Smith
- Division of Surgical Oncology, Department of Surgery, Vanderbilt University, Nashville, Tennessee
| | - Cliff Cho
- Division of Hepatopancreatobiliary and Advanced Gastrointestinal Surgery, Department of Surgery, University of Michigan, Ann Arbor, Wisconsin
| | - Megan Beems
- Division of Hepatopancreatobiliary and Advanced Gastrointestinal Surgery, Department of Surgery, University of Michigan, Ann Arbor, Wisconsin
| | - Carl R Schmidt
- Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, Ohio
| | - Mary Dillhoff
- Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, Ohio
| | - Shishir K Maithel
- Division of Surgical Oncology, Department of Surgery, Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Timothy M Pawlik
- Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, Ohio
| |
Collapse
|
198
|
Cives M, Pellè E, Silvestris F. The management of refractory carcinoid syndrome: challenges and opportunities ahead. J Med Econ 2018; 21:241-243. [PMID: 29039712 DOI: 10.1080/13696998.2017.1394312] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Mauro Cives
- a Department of Biomedical Sciences and Human Oncology , University of Bari 'Aldo Moro' , Bari , Italy
| | - Eleonora Pellè
- a Department of Biomedical Sciences and Human Oncology , University of Bari 'Aldo Moro' , Bari , Italy
| | - Franco Silvestris
- a Department of Biomedical Sciences and Human Oncology , University of Bari 'Aldo Moro' , Bari , Italy
| |
Collapse
|
199
|
Abstract
PURPOSE OF REVIEW To review recent advances and controversies in all aspects of carcinoid-syndrome. RECENT FINDINGS Over the last few years there have been a number of advances in all aspects of carcinoid syndrome as well as new therapies. These include new studies on its epidemiology which demonstrate it is increasing in frequency; increasing insights into the pathogenesis of its various clinical manifestations and into its natural history: definition of prognostic factors; new methods to verify its presence; the development of new drugs to treat its various manifestations, both initially and in somatostatin-refractory cases; and an increased understanding of the pathogenesis, natural history and management of carcinoid heart disease. These advances have generated several controversies and these are also reviewed. SUMMARY There have been numerous advances in all aspects of the carcinoid-syndrome, which is the most common functional syndrome neuroendocrine tumors produce. These advances are leading to new approaches to the management of these patients and in some cases to new controversies.
Collapse
Affiliation(s)
- Tetsuhide Ito
- Neuroendocrine Tumor Centre, Fukuoka Sanno Hospital, International University of Health and Welfare
| | - Lingaku Lee
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Digestive Diseases Branch, NIDDK, NIH, Bethesda, Maryland, USA
| | - Robert T Jensen
- Digestive Diseases Branch, NIDDK, NIH, Bethesda, Maryland, USA
| |
Collapse
|
200
|
Dillon JS, Chandrasekharan C. Telotristat ethyl: a novel agent for the therapy of carcinoid syndrome diarrhea. Future Oncol 2018; 14:1155-1164. [PMID: 29350062 DOI: 10.2217/fon-2017-0340] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Carcinoid syndrome (CS), characterized by diarrhea and flushing, is present in 20% of patients with neuroendocrine tumors at diagnosis and becomes more frequent with progression. The diarrhea of CS is caused mainly by tumoral secretion of serotonin. It may not be fully controlled by somatostatin analogs, the currently indicated drugs for symptomatic relief. Telotristat ethyl is a novel inhibitor of tryptophan hydroxylase, the rate-limiting enzyme in serotonin biosynthesis. Administration of the drug decreases diarrhea in patients with CS. Telotristat ethyl was approved in February 2017 (USA) and September 2017 (European Commission) for the treatment of CS diarrhea in adults inadequately controlled by somatostatin analog alone. This drug is expected to greatly improve the health and quality of life of patients with CS diarrhea.
Collapse
Affiliation(s)
- Joseph S Dillon
- Division of Endocrinology, University of Iowa Hospital & VA Medical Center, Iowa City, IA 52242, USA
| | - Chandrikha Chandrasekharan
- Division of Hematology Oncology, University of Iowa Hospital & VA Medical Center, Iowa City, IA 52242, USA
| |
Collapse
|