1
|
Maluchenko A, Maksimov D, Antysheva Z, Krupinova J, Avsievich E, Glazova O, Bodunova N, Karnaukhov N, Feidorov I, Salimgereeva D, Voloshin M, Volchkov P. Molecular Basis of Pancreatic Neuroendocrine Tumors. Int J Mol Sci 2024; 25:11017. [PMID: 39456803 PMCID: PMC11507569 DOI: 10.3390/ijms252011017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/20/2024] [Accepted: 09/21/2024] [Indexed: 10/28/2024] Open
Abstract
Pancreatic neuroendocrine tumors (NETs) are rare well-differentiated neoplasms with limited therapeutic options and unknown cells of origin. The current classification of pancreatic neuroendocrine tumors is based on proliferative grading, and guides therapeutic strategies, however, tumors within grades exhibit profound heterogeneity in clinical manifestation and outcome. Manifold studies have highlighted intra-patient differences in tumors at the genetic and transcriptomic levels. Molecular classification might become an alternative or complementary basis for treatment decisions and reflect tumor biology, actionable cellular processes. Here, we provide a comprehensive review of genomic, transcriptomic, proteomic and epigenomic studies of pancreatic NETs to elucidate patterns shared between proposed subtypes that could form a foundation for new classification. We denote four NET subtypes with distinct molecular features, which were consistently reproduced using various omics technologies.
Collapse
Affiliation(s)
- Alesia Maluchenko
- Moscow Center for Advanced Studies, Kulakova Str. 20, Moscow 123592, Russia; (A.M.); (D.M.); (Z.A.); (E.A.); (O.G.); (P.V.)
| | - Denis Maksimov
- Moscow Center for Advanced Studies, Kulakova Str. 20, Moscow 123592, Russia; (A.M.); (D.M.); (Z.A.); (E.A.); (O.G.); (P.V.)
| | - Zoia Antysheva
- Moscow Center for Advanced Studies, Kulakova Str. 20, Moscow 123592, Russia; (A.M.); (D.M.); (Z.A.); (E.A.); (O.G.); (P.V.)
| | - Julia Krupinova
- Moscow Center for Advanced Studies, Kulakova Str. 20, Moscow 123592, Russia; (A.M.); (D.M.); (Z.A.); (E.A.); (O.G.); (P.V.)
- Moscow Clinical Scientific Center N.A. A.S. Loginov, Moscow 111123, Russia; (N.B.); (N.K.); (I.F.); (D.S.); (M.V.)
| | - Ekaterina Avsievich
- Moscow Center for Advanced Studies, Kulakova Str. 20, Moscow 123592, Russia; (A.M.); (D.M.); (Z.A.); (E.A.); (O.G.); (P.V.)
- Moscow Clinical Scientific Center N.A. A.S. Loginov, Moscow 111123, Russia; (N.B.); (N.K.); (I.F.); (D.S.); (M.V.)
| | - Olga Glazova
- Moscow Center for Advanced Studies, Kulakova Str. 20, Moscow 123592, Russia; (A.M.); (D.M.); (Z.A.); (E.A.); (O.G.); (P.V.)
- Moscow Clinical Scientific Center N.A. A.S. Loginov, Moscow 111123, Russia; (N.B.); (N.K.); (I.F.); (D.S.); (M.V.)
| | - Natalia Bodunova
- Moscow Clinical Scientific Center N.A. A.S. Loginov, Moscow 111123, Russia; (N.B.); (N.K.); (I.F.); (D.S.); (M.V.)
| | - Nikolay Karnaukhov
- Moscow Clinical Scientific Center N.A. A.S. Loginov, Moscow 111123, Russia; (N.B.); (N.K.); (I.F.); (D.S.); (M.V.)
| | - Ilia Feidorov
- Moscow Clinical Scientific Center N.A. A.S. Loginov, Moscow 111123, Russia; (N.B.); (N.K.); (I.F.); (D.S.); (M.V.)
| | - Diana Salimgereeva
- Moscow Clinical Scientific Center N.A. A.S. Loginov, Moscow 111123, Russia; (N.B.); (N.K.); (I.F.); (D.S.); (M.V.)
| | - Mark Voloshin
- Moscow Clinical Scientific Center N.A. A.S. Loginov, Moscow 111123, Russia; (N.B.); (N.K.); (I.F.); (D.S.); (M.V.)
| | - Pavel Volchkov
- Moscow Center for Advanced Studies, Kulakova Str. 20, Moscow 123592, Russia; (A.M.); (D.M.); (Z.A.); (E.A.); (O.G.); (P.V.)
- Moscow Clinical Scientific Center N.A. A.S. Loginov, Moscow 111123, Russia; (N.B.); (N.K.); (I.F.); (D.S.); (M.V.)
| |
Collapse
|
2
|
Taherifard E, Bakhtiar M, Mahnoor M, Ahmed R, Cavalcante L, Zhang J, Saeed A. Efficacy and safety of temozolomide-based regimens in advanced pancreatic neuroendocrine tumors: a systematic review and meta-analysis. BMC Cancer 2024; 24:192. [PMID: 38347461 PMCID: PMC10860315 DOI: 10.1186/s12885-024-11926-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 01/27/2024] [Indexed: 02/15/2024] Open
Abstract
BACKGROUND Recent advances in the management of pancreatic neuroendocrine tumors (pNETs) highlight the potential benefits of temozolomide, an alkylating agent, for these patients. In this meta-analysis, we aimed to assess the outcome of temozolomide, alone or in combination with other anticancer medications in patients with advanced pNET. METHODS Online databases of PubMed, Web of Science, Embase, the Cochrane Library, and ClinicalTrials.gov were searched systematically for clinical trials that reported the efficacy and safety of temozolomide in patients with advanced pNET. Random-effect model was utilized to estimate pooled rates of outcomes based on Response Evaluation Criteria in Solid Tumors criteria, biochemical response, and adverse events (AEs). RESULTS A total of 14 studies, providing details of 441 individuals with advanced pNET, were included. The quantitative analyses showed a pooled objective response rate (ORR) of 41.2% (95% confidence interval, CI, of 32.4%-50.6%), disease control rate (DCR) of 85.3% (95% CI of 74.9%-91.9%), and a more than 50% decrease from baseline chromogranin A levels of 44.9% (95% CI of 31.6%-49.0%). Regarding safety, the results showed that the pooled rates of nonserious AEs and serious AEs were 93.8% (95% CI of 88.3%-96.8%) and 23.7% (95% CI of 12.0%-41.5%), respectively. The main severe AEs encompassed hematological toxicities. CONCLUSIONS In conclusion, our meta-analysis suggests that treatment with temozolomide, either as a monotherapy or in combination with other anticancer treatments might be an effective and relatively safe option for patients with advanced locally unresectable and metastatic pNET. However, additional clinical trials are required to further strengthen these findings. This study has been registered in PROSPERO (CRD42023409280).
Collapse
Affiliation(s)
- Erfan Taherifard
- Department of Medicine, Division of Hematology & Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Muhammad Bakhtiar
- Department of Medicine, Division of Hematology & Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Mahnoor Mahnoor
- Department of Medicine, Division of Hematology & Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Rabeea Ahmed
- Department of Medicine, Division of Hematology & Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | | | - Janie Zhang
- Department of Medicine, Division of Hematology & Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Anwaar Saeed
- Department of Medicine, Division of Hematology & Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA.
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
| |
Collapse
|
3
|
Hu C, Ye M, Bai J, Liu P, Lu F, Chen J, Xu Y, Yan L, Yu P, Xiao Z, Gu D, Xu L, Tian Y, Tang Q. FOXA2-initiated transcriptional activation of INHBA induced by methylmalonic acid promotes pancreatic neuroendocrine neoplasm progression. Cell Mol Life Sci 2024; 81:50. [PMID: 38252148 PMCID: PMC10803496 DOI: 10.1007/s00018-023-05084-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 12/01/2023] [Accepted: 12/06/2023] [Indexed: 01/23/2024]
Abstract
Pancreatic neuroendocrine neoplasms (PanNENs) are a group of highly heterogeneous neoplasms originating from the endocrine islet cells of the pancreas with characteristic neuroendocrine differentiation, more than 60% of which represent metastases when diagnosis, causing major tumor-related death. Metabolic alterations have been recognized as one of the hallmarks of tumor metastasis, providing attractive therapeutic targets. However, little is known about the molecular mechanism of metabolic changes regulating PanNEN progression. In this study, we first identified methylmalonic acid (MMA) as an oncometabolite for PanNEN progression, based on serum metabolomics of metastatic PanNEN compared with non-metastatic PanNEN patients. One of the key findings was the potentially novel mechanism of epithelial-mesenchymal transition (EMT) triggered by MMA. Inhibin βA (INHBA) was characterized as a key regulator of MMA-induced PanNEN progression according to transcriptomic analysis, which has been validated in vitro and in vivo. Mechanistically, INHBA was activated by FOXA2, a neuroendocrine (NE) specific transcription factor, which was initiated during MMA-induced progression. In addition, MMA-induced INHBA upregulation activated downstream MITF to regulate EMT-related genes in PanNEN cells. Collectively, these data suggest that activation of INHBA via FOXA2 promotes MITF-mediated EMT during MMA inducing PanNEN progression, which puts forward a novel therapeutic target for PanNENs.
Collapse
Affiliation(s)
- Chunhua Hu
- Department of Geriatric Gastroenterology, Neuroendocrine Tumor Center, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Institute of Neuroendocrine Tumor, Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, China
| | - Mujie Ye
- Department of Geriatric Gastroenterology, Neuroendocrine Tumor Center, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Institute of Neuroendocrine Tumor, Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, China
| | - Jianan Bai
- Department of Geriatric Gastroenterology, Neuroendocrine Tumor Center, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Institute of Neuroendocrine Tumor, Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, China
| | - Pengfei Liu
- Department of Gastroenterology, Jiangyin People's Hospital, Jiangyin, Jiangsu Province, China
| | - Feiyu Lu
- Department of Geriatric Gastroenterology, Neuroendocrine Tumor Center, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Institute of Neuroendocrine Tumor, Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, China
| | - Jinhao Chen
- Department of Geriatric Gastroenterology, Neuroendocrine Tumor Center, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Institute of Neuroendocrine Tumor, Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, China
| | - Yanling Xu
- Department of Geriatric Gastroenterology, Neuroendocrine Tumor Center, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Institute of Neuroendocrine Tumor, Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, China
| | - Lijun Yan
- Department of Geriatric Gastroenterology, Neuroendocrine Tumor Center, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Institute of Neuroendocrine Tumor, Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, China
| | - Ping Yu
- Department of Geriatric Gastroenterology, Neuroendocrine Tumor Center, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Institute of Neuroendocrine Tumor, Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, China
| | - Zequan Xiao
- Department of Gastroenterology, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Ili State, China
| | - Danyang Gu
- Department of Geriatric Gastroenterology, Neuroendocrine Tumor Center, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Institute of Neuroendocrine Tumor, Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, China
| | - Lin Xu
- Department of Geriatric Gastroenterology, Neuroendocrine Tumor Center, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Institute of Neuroendocrine Tumor, Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, China
| | - Ye Tian
- Department of Geriatric Gastroenterology, Neuroendocrine Tumor Center, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Institute of Neuroendocrine Tumor, Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, China
| | - Qiyun Tang
- Department of Geriatric Gastroenterology, Neuroendocrine Tumor Center, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Institute of Neuroendocrine Tumor, Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, China.
| |
Collapse
|
4
|
Chang L, Bi X, Li S, Tong Q, Gu Y, He Z, Li Y, Chen Q, Cui J, Yu H, He Q, Liu M. The comparison of three different molecular imaging methods in localization and grading of insulinoma. Front Endocrinol (Lausanne) 2023; 14:1163176. [PMID: 37455905 PMCID: PMC10348808 DOI: 10.3389/fendo.2023.1163176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 06/15/2023] [Indexed: 07/18/2023] Open
Abstract
Aims This cross-sectional study compared the value of molecular imaging (Exendin-4 positron emission tomography/computed tomography [PET/CT], 68Ga-DOTATATE PET/CT, 18F- fluorodeoxyglucose [FDG] PET/CT) in insulinoma localization by stratified tumor size and grading, and explored the correlation of the related the maximum standardized uptake value (SUVmax) with insulinoma grading, Ki-67, maximum tumor diameter, and glucose metabolism. Methods In 28 insulinoma patients, the sensitivity of three types of PET/CT for localizing insulinoma was calculated according to tumor size and grade. We compared the SUVmax for different insulinoma grades and analyzed the correlation of SUVmax with Ki-67, maximum tumor diameter, and glucose metabolism indicators. Results The study included 12 grade (G) 1 and 16 G2 cases, with maximum tumor diameters ranging from 9 to 40 mm. Without differentiation by size and grade, the sensitivity of Exendin-4 PET/CT to localize insulinoma was 100%, which significantly exceeded that of 68Ga-DOTATATE PET/CT and 18F-FDG PET/CT (75% and 57%, respectively). In tumors with a maximum diameter ≤ 20 mm and ≤ 15 mm, the sensitivity of Exendin-4 (both 100%) significantly exceeded that of 68Ga-DOTATATE PET/CT (74% and 64%, respectively) and 18F-FDG PET/CT (54% and 50%, respectively). In G1 tumors, the sensitivity of Exendin-4 PET/CT was significantly higher than that of 18F-FDG PET/CT, but not that of 68Ga-DOTATATE PET/CT, while in G2 tumors, the sensitivity of Exendin-4 PET/CT was significantly higher than that of both other types. However, all three PET/CT types missed a metastatic lymph node in one patient. The 18F-FDG PET/CT SUVmax was significantly lower than that of the other PET/CT types and that of 68Ga-DOTATATE PET/CT was significantly lower in G2 than in G1. 68Ga-DOTATATE PET/CT SUVmax correlated negatively with Ki-67. A receiver operating characteristic (ROC) curve suggested that 68Ga-DOTATATE PET/CT SUVmax > 19.9 could predict G1 tumors. Conclusion Exendin-4 PET/CT was superior to 68Ga-DOTATATE PET/CT and 18F-FDG PET/CT for insulinoma localization, particularly small and G2 tumors, but its diagnostic value in small metastatic lymph nodes requires further exploration. 68Ga-DOTATATE PET/CT SUVmax could be used as an adjunct to pathology, and a value > 19.9 could predict G1 tumors. No PET/CT SUVmax could predict tumor maximum diameter and glucose metabolism.
Collapse
Affiliation(s)
- Lina Chang
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Xinyu Bi
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Shuo Li
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Qi Tong
- Department of General Internal Medicine, Baodi District People’s Hospital, Tianjin, China
| | - Yian Gu
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Zonghao He
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Yansheng Li
- Department of PET/CT Examination Room, Tianjin Medical University General Hospital, Tianjin, China
| | - Qiusong Chen
- Department of PET/CT Examination Room, Tianjin Medical University General Hospital, Tianjin, China
| | - Jingqiu Cui
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Haonan Yu
- Department of PET/CT Examination Room, Tianjin Medical University General Hospital, Tianjin, China
| | - Qing He
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Ming Liu
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
5
|
Laser Capture Microdissection: A Gear for Pancreatic Cancer Research. Int J Mol Sci 2022; 23:ijms232314566. [PMID: 36498893 PMCID: PMC9741023 DOI: 10.3390/ijms232314566] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/16/2022] [Accepted: 11/19/2022] [Indexed: 11/24/2022] Open
Abstract
The advancement in molecular techniques has been attributed to the quality and significance of cancer research. Pancreatic cancer (PC) is one of the rare cancers with aggressive behavior and a high mortality rate. The asymptomatic nature of the disease until its advanced stage has resulted in late diagnosis as well as poor prognosis. The heterogeneous character of PC has complicated cancer development and progression studies. The analysis of bulk tissues of the disease was insufficient to understand the disease, hence, the introduction of the single-cell separating technique aided researchers to decipher more about the specific cell population of tumors. This review gives an overview of the Laser Capture Microdissection (LCM) technique, one of the single-cell separation methods used in PC research.
Collapse
|
6
|
Brown G, Monaghan AM, Fristedt R, Ramsey E, Al-Mrayat M, Rajak R, Armstrong T, Takhar A. Metastatic mixed VIPoma/PPoma-induced diarrhoea causing renal failure. Endocrinol Diabetes Metab Case Rep 2022; 2022:22-0231. [PMID: 36001009 PMCID: PMC9422256 DOI: 10.1530/edm-22-0231] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 07/26/2022] [Indexed: 11/29/2022] Open
Abstract
Summary Vasoactive intestinal peptide-secreting tumours (VIPomas) are an extremely rare form of functional pancreatic neuroendocrine tumour with an estimated annual incidence of 1 in 10 million. Associated tumour hypersecretion of other peptides, including pancreatic polypeptide (PPomas), may also be seen. These malignancies classically present with a defined triad of refractory diarrhoea, hypokalaemia and metabolic acidosis known as Verner-Morrison syndrome. Diagnosis is frequently delayed, and the majority of patients will have metastatic disease at presentation. Symptoms are usually well controlled with somatostatin analogue administration. Here we report a case of metastatic mixed VIPoma/PPoma-induced diarrhoea causing renal failure so severe that ultrafiltration was required to recover adequate renal function. Learning points Profuse, watery diarrhoea is a common presenting complaint with a multitude of aetiologies. This, combined with the rarity of these tumours, makes diagnosis difficult and frequently delayed. A functional neuroendocrine tumour should be suspected when diarrhoea is unusually extreme, prolonged and common causes have been promptly excluded. These patients are likely to be profoundly unwell on presentation. They are extremely hypovolaemic with dangerous electrolyte and metabolic abnormalities. Aggressive initial rehydration and electrolyte replacement are imperative. A somatostatin analogue should be commenced as soon as the diagnosis is suspected. This is an extreme example of Verner-Morrison syndrome. We are unaware of another case where renal failure secondary to diarrhoea and dehydration was so severe that renal replacement therapy was required to restore adequate renal function, further emphasising how critically unwell these patients can be. Both the primary tumour and metastases showed a remarkably good and rapid response to somatostatin analogue administration. Cystic change and involution were noted on repeat imaging within days. Prior to his illness, this patient was extremely high functioning with no medical history. His diagnosis was an enormous psychological shock, and the consideration and care for his psychological well-being were a crucial part of his overall management. It highlights the importance of a holistic approach to cancer care and the role of the clinical nurse specialist within the cancer multidisciplinary team.
Collapse
Affiliation(s)
- George Brown
- Department of Hepatobiliary & Pancreatic Surgery, University Hospital Southampton, Southampton, UK
| | - Anthony Mark Monaghan
- Department of Hepatobiliary & Pancreatic Surgery, University Hospital Southampton, Southampton, UK
| | - Richard Fristedt
- Department of Hepatobiliary & Pancreatic Surgery, University Hospital Southampton, Southampton, UK
| | - Emma Ramsey
- Department of Hepatobiliary & Pancreatic Surgery, University Hospital Southampton, Southampton, UK
| | - Ma’en Al-Mrayat
- Department of Endocrinology, University Hospital Southampton, Southampton, UK
| | - Rushda Rajak
- Department of Cellular Pathology, University Hospital Southampton, Southampton, UK
| | - Thomas Armstrong
- Department of Hepatobiliary & Pancreatic Surgery, University Hospital Southampton, Southampton, UK
| | - Arjun Takhar
- Department of Hepatobiliary & Pancreatic Surgery, University Hospital Southampton, Southampton, UK
| |
Collapse
|
7
|
Mapelli P, Bezzi C, Palumbo D, Canevari C, Ghezzo S, Samanes Gajate AM, Catalfamo B, Messina A, Presotto L, Guarnaccia A, Bettinardi V, Muffatti F, Andreasi V, Schiavo Lena M, Gianolli L, Partelli S, Falconi M, Scifo P, De Cobelli F, Picchio M. 68Ga-DOTATOC PET/MR imaging and radiomic parameters in predicting histopathological prognostic factors in patients with pancreatic neuroendocrine well-differentiated tumours. Eur J Nucl Med Mol Imaging 2022; 49:2352-2363. [DOI: 10.1007/s00259-022-05677-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 12/31/2021] [Indexed: 12/17/2022]
|
8
|
Hanan S, Luvsannyam E, Jain MS, Laller S, Cheema T, Mellon C, Teed D, Somagutta M, Ahmad LN, Tiesenga FM. A Rare Case of Three Distinct Gastrointestinal Neoplasms Occurring Simultaneously in an Elderly Patient. J Med Cases 2021; 12:419-423. [PMID: 34691340 PMCID: PMC8510667 DOI: 10.14740/jmc3765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 09/01/2021] [Indexed: 12/09/2022] Open
Abstract
Duodenal adenocarcinoma (DA), gastrointestinal stromal tumor (GIST), and pancreatic neuroendocrine tumor (PNET) are a set of rare gastrointestinal (GI) cancers characterized by nonspecific symptoms such as anemia, weight loss, and abdominal pain. We report an interesting case of DA, GIST, and PNET in a 79-year-old African American male who presented to the emergency department with syncope. The patient was tachycardic and found to have severe anemia. Further computed tomography (CT) of the abdomen and pelvis with contrast revealed a mass within the duodenal bulb along with a pancreatic cystic lesion. The patient underwent gastroduodenectomy and distal pancreatectomy with incidental findings of GIST in the posterior gastric wall. The patient after surgical removal of all tumors no longer experienced abdominal pain and had hematocrit and hemoglobin (H&H) level of 9.1/31.7 postoperation day (POD) 14. The case unfolds a rare presentation of three neoplasms that were moderate to well-differentiated with no metastases. It highlights the importance of assessment of rare GI neoplasms concurrently with colorectal cancer when geriatric patients present with nonspecific GI symptoms.
Collapse
Affiliation(s)
- Saad Hanan
- Saint James School of Medicine, Chicago, IL, USA
| | | | - Molly S Jain
- Saint James School of Medicine, Chicago, IL, USA
| | | | | | - Cory Mellon
- West Suburban Medical Centre, Oak Park, IL, USA
| | - Don Teed
- West Suburban Medical Centre, Oak Park, IL, USA
| | - Manoj Somagutta
- California Institute of Behavioral and Neurosciences and Psychology, Fairfield, CA, USA
| | | | | |
Collapse
|
9
|
Maharjan CK, Ear PH, Tran CG, Howe JR, Chandrasekharan C, Quelle DE. Pancreatic Neuroendocrine Tumors: Molecular Mechanisms and Therapeutic Targets. Cancers (Basel) 2021; 13:5117. [PMID: 34680266 PMCID: PMC8533967 DOI: 10.3390/cancers13205117] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/08/2021] [Accepted: 10/09/2021] [Indexed: 12/16/2022] Open
Abstract
Pancreatic neuroendocrine tumors (pNETs) are unique, slow-growing malignancies whose molecular pathogenesis is incompletely understood. With rising incidence of pNETs over the last four decades, larger and more comprehensive 'omic' analyses of patient tumors have led to a clearer picture of the pNET genomic landscape and transcriptional profiles for both primary and metastatic lesions. In pNET patients with advanced disease, those insights have guided the use of targeted therapies that inhibit activated mTOR and receptor tyrosine kinase (RTK) pathways or stimulate somatostatin receptor signaling. Such treatments have significantly benefited patients, but intrinsic or acquired drug resistance in the tumors remains a major problem that leaves few to no effective treatment options for advanced cases. This demands a better understanding of essential molecular and biological events underlying pNET growth, metastasis, and drug resistance. This review examines the known molecular alterations associated with pNET pathogenesis, identifying which changes may be drivers of the disease and, as such, relevant therapeutic targets. We also highlight areas that warrant further investigation at the biological level and discuss available model systems for pNET research. The paucity of pNET models has hampered research efforts over the years, although recently developed cell line, animal, patient-derived xenograft, and patient-derived organoid models have significantly expanded the available platforms for pNET investigations. Advancements in pNET research and understanding are expected to guide improved patient treatments.
Collapse
Affiliation(s)
- Chandra K. Maharjan
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA;
| | - Po Hien Ear
- Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; (P.H.E.); (C.G.T.); (J.R.H.)
| | - Catherine G. Tran
- Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; (P.H.E.); (C.G.T.); (J.R.H.)
| | - James R. Howe
- Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; (P.H.E.); (C.G.T.); (J.R.H.)
| | - Chandrikha Chandrasekharan
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA;
| | - Dawn E. Quelle
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA;
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
10
|
RABL6A Promotes Pancreatic Neuroendocrine Tumor Angiogenesis and Progression In Vivo. Biomedicines 2021; 9:biomedicines9060633. [PMID: 34199469 PMCID: PMC8228095 DOI: 10.3390/biomedicines9060633] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/26/2021] [Accepted: 05/28/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic neuroendocrine tumors (pNETs) are difficult-to-treat neoplasms whose incidence is rising. Greater understanding of pNET pathogenesis is needed to identify new biomarkers and targets for improved therapy. RABL6A, a novel oncogenic GTPase, is highly expressed in patient pNETs and required for pNET cell proliferation and survival in vitro. Here, we investigated the role of RABL6A in pNET progression in vivo using a well-established model of the disease. RIP-Tag2 (RT2) mice develop functional pNETs (insulinomas) due to SV40 large T-antigen expression in pancreatic islet β cells. RABL6A loss in RT2 mice significantly delayed pancreatic tumor formation, reduced tumor angiogenesis and mitoses, and extended survival. Those effects correlated with upregulation of anti-angiogenic p19ARF and downregulation of proangiogenic c-Myc in RABL6A-deficient islets and tumors. Our findings demonstrate that RABL6A is a bona fide oncogenic driver of pNET angiogenesis and development in vivo.
Collapse
|
11
|
Deng S, Fan Z, Xia H, Gong Y, Qian Y, Huang Q, Cheng H, Jin K, Xiao Z, Luo G, Yu X, Liu C. Fibrinogen/Albumin Ratio as a Promising Marker for Predicting Survival in Pancreatic Neuroendocrine Neoplasms. Cancer Manag Res 2021; 13:107-115. [PMID: 33447083 PMCID: PMC7802789 DOI: 10.2147/cmar.s275173] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 12/10/2020] [Indexed: 12/16/2022] Open
Abstract
Background The fibrinogen/albumin ratio (FAR) has been widely reported to be a possible biomarker for predicting prognosis in several types of tumors, but the prognostic value of the FAR in pancreatic neuroendocrine neoplasms (Pan-NENs) has not been systematically studied. Patients and Methods In total, 324 patients with Pan-NENs were recruited. The patients were divided into 2 subgroups according to the FAR cutoff value, and clinicopathological characteristics of the 2 subgroups were compared. Overall survival (OS) was the primary endpoint, and progression-free survival (PFS) was the secondary endpoint. The prognostic value of the FAR was analyzed in univariate and multivariate analyses. Results The optimal cutoff value for the FAR was calculated to be 0.08 for OS. The patients with a FAR ≥0.08 had higher proportions of nonfunctioning tumors, Pan-NECs, grade 3 tumors, and stage IV tumors than those with a FAR <0.08. In the univariate analysis, a FAR ≥ 0.08 was associated with poor OS (hazard ratio (HR) = 2.37, P < 0.001) and PFS (HR = 2.37, P < 0.001). In the multivariate analysis, a FAR ≥0.08 was an independent risk factor for poor OS (HR = 4.70, P < 0.001) and PFS (HR = 1.80, P = 0.006). Conclusion The pretreatment FAR, which includes fibrinogen and albumin, was a feasible and predictive biomarker for prognosis in patients with Pan-NENs. An elevated FAR, based on a cutoff value of 0.08, was an independent risk factor for poor OS and PFS.
Collapse
Affiliation(s)
- Shengming Deng
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.,Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China.,Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Zhiyao Fan
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.,Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China.,Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Huanyu Xia
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.,Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China.,Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Yitao Gong
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.,Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China.,Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Yunzhen Qian
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.,Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China.,Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Qiuyi Huang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.,Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China.,Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - He Cheng
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.,Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China.,Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Kaizhou Jin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.,Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China.,Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Zhiwen Xiao
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.,Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China.,Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Guopei Luo
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.,Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China.,Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.,Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China.,Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Chen Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.,Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China.,Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
12
|
Lu L, Shang Y, Mullins CS, Zhang X, Linnebacher M. Epidemiologic trends and prognostic risk factors of patients with pancreatic neuroendocrine neoplasms in the US: an updated population-based study. Future Oncol 2021; 17:549-563. [PMID: 33401958 DOI: 10.2217/fon-2020-0543] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Background: We aimed to evaluate the incidence, mortality and survival outcome for patients with pancreatic neuroendocrine neoplasms (pNEN). Methods: Patients with pNEN were collected from the Surveillance, Epidemiology, and End Results (SEER) database. Incidence, mortality and average annual percentage change (AAPC) were calculated using SEER stat 8.3.6 and Joinpoint software. Survival outcome was estimated using Kaplan-Meier and Cox proportional hazard model. Results: During 2000-2016, the incidence of pNEN significantly rose from 0.2647 to 1.0618 per 100,000 persons with an AAPC of 9.4; AAPC of mortality was 6.7. Prognostic improvement was revealed in 2010-2016, but not for late-stage pNEN, which had the highest risk of death. Conclusion: Efforts to improve prognosis of pNEN patients must focus on not only early detection, but also on improving therapy for late-stage disease.
Collapse
Affiliation(s)
- Lili Lu
- Department of General Surgery, Molecular Oncology & Immunotherapy, Rostock University Medical Center, Schillingallee 69, 18057, Rostock, Germany
| | - Yuru Shang
- Department of Plastic Surgery, Shenzhen University General Hospital, Xueyuan Road 1098, 518055, Shenzhen, PR China
| | - Christina Susanne Mullins
- Department of General Surgery, Molecular Oncology & Immunotherapy, Rostock University Medical Center, Schillingallee 69, 18057, Rostock, Germany
| | - Xianbin Zhang
- Department of General Surgery, Shenzhen University General Hospital & Carson International Cancer Research Centre, Xueyuan Road 1098, 518055, Shenzhen, PR China
| | - Michael Linnebacher
- Department of General Surgery, Molecular Oncology & Immunotherapy, Rostock University Medical Center, Schillingallee 69, 18057, Rostock, Germany
| |
Collapse
|
13
|
Li Y, Fan G, Yu F, Tian C, Tan H. Meta-Analysis of Prognostic Factors for Recurrence of Resected Well-Differentiated Pancreatic Neuroendocrine Tumors. Neuroendocrinology 2021; 111:1231-1237. [PMID: 33370727 DOI: 10.1159/000514047] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 12/17/2020] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Well-differentiated pancreatic neuroendocrine tumors (WDPNETs) are a group of rare and heterogeneous tumors. However, the prognostic factors for recurrence after curative resection still remain controversial. We aim to illustrate the prognostic factors for recurrence of resected WDPNETs. METHODS All relevant articles published through June 2020 were identified via PubMed, Embase, Web of Science, and Cochrane Library. Articles that examined the prognostic factors of WDPNETs were enrolled. RESULTS Ten articles were finally included in this study. From 1993 to 2018, 2,863 patients underwent curative resection and 358 patients had recurrence, and the combined recurrence rate was 13%. Furthermore, the pooled data indicated that patients with G2, positive lymph node and surgical resection margin, vascular invasion, and perineural invasion had a decreased disease-free survival for WDPNETs. However, gender, function, and tumor size had no significant relationship with WDPNETs recurrence. CONCLUSION These findings demonstrated that G2, positive lymph node and surgical resection margin, vascular invasion, and perineural invasion could be prognostic factors for recurrence of resected WDPNETs, indicating that patients with these high-risk factors need closer postoperative follow-up and may benefit from adjuvant therapy.
Collapse
Affiliation(s)
- Yuanliang Li
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- Department of Integrative Oncology, China-Japan Friendship Hospital, Beijing, China
| | - Guohui Fan
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Fuhuan Yu
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- Department of Integrative Oncology, China-Japan Friendship Hospital, Beijing, China
| | - Chao Tian
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- Department of Integrative Oncology, China-Japan Friendship Hospital, Beijing, China
| | - Huangying Tan
- Department of Integrative Oncology, China-Japan Friendship Hospital, Beijing, China,
| |
Collapse
|
14
|
Xie VK, He J, Xie K. Protein arginine methylation promotes therapeutic resistance in human pancreatic cancer. Cytokine Growth Factor Rev 2020; 55:58-69. [PMID: 32739260 DOI: 10.1016/j.cytogfr.2020.07.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 07/05/2020] [Accepted: 07/06/2020] [Indexed: 12/26/2022]
Abstract
Pancreatic cancer is a lethal disease with limited treatment options for cure. A high degree of intrinsic and acquired therapeutic resistance may result from cellular alterations in genes and proteins involved in drug transportation and metabolism, or from the influences of cancer microenvironment. Mechanistic basis for therapeutic resistance remains unclear and should profoundly impact our ability to understand pancreatic cancer pathogenesis and its effective clinical management. Recent evidences have indicated the importance of epigenetic changes in pancreatic cancer, including posttranslational modifications of proteins. We will review new knowledge on protein arginine methylation and its consequential contribution to therapeutic resistance of pancreatic cancer, underlying molecular mechanism, and clinical application of potential strategies of its reversal.
Collapse
Affiliation(s)
- Victoria Katie Xie
- Department of Gastroenterology, Guangzhou First People's Hospital Affiliated to The South China University of Technology School of Medicine, Guangzhou, Guangdong, People's Republic of China
| | - Jie He
- Department of Gastroenterology, Guangzhou First People's Hospital Affiliated to The South China University of Technology School of Medicine, Guangzhou, Guangdong, People's Republic of China
| | - Keping Xie
- Department of Gastroenterology, Guangzhou First People's Hospital Affiliated to The South China University of Technology School of Medicine, Guangzhou, Guangdong, People's Republic of China.
| |
Collapse
|
15
|
The Role of Tachykinins in the Initiation and Progression of Gastrointestinal Cancers: A Review. INTERNATIONAL JOURNAL OF CANCER MANAGEMENT 2020. [DOI: 10.5812/ijcm.100717] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
16
|
Kit OI, Gvaldin DY, Trifanov VS, Kolesnikov EN, Timoshkina NN. Molecular-Genetic Features of Pancreatic Neuroendocrine Tumors. RUSS J GENET+ 2020. [DOI: 10.1134/s1022795420020064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
17
|
Viti M, Lombardi PM, Marinelli M, Onorati M, D'Urbano C. Ampullary Adenocarcinoma with Incidental Pancreatic Neuroendocrine Tumor: Report of an Extremely Rare Case and Review of Literature. Cureus 2019; 11:e6143. [PMID: 31886078 PMCID: PMC6907710 DOI: 10.7759/cureus.6143] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Periampullary neoplasms are a heterogeneous group of tumors arising within 2 cm of the ampulla of Vater. Neuroendocrine tumors can originate throughout the entire body, from neuroendocrine cells. These neoplasms exhibit deep differences, according to their origin and biological behavior. We describe a case of a 79-year-old man who underwent pancreaticoduodenectomy for adenocarcinoma of the ampulla of Vater after proper staging. At gross histology, an incidental pancreatic neuroendocrine tumor was also documented. Despite two synchronous neoplasms, the patient survived 34 months with no evidence of recurrence at follow-up. The synchronous presence of a second primitive tumor in patients affected by a neuroendocrine tumor is reported in the literature; incidence is variable and the most common site is the gastrointestinal tract. Diagnostic workup for ampullary neoplasms includes abdominal computed tomography (CT) scan, magnetic resonance imaging (MRI) and endoscopic ultrasound (EUS). These investigations infrequently may detect subcentimetric lesions. We believe this case is currently extremely rare. Preoperative diagnosis of synchronous PanNET would not have changed our approach since surgical therapy represents the gold standard in resectable ampullary neoplasms, and it has a primary role in the prognosis of the present patient.
Collapse
Affiliation(s)
- Matteo Viti
- General Surgery, G. Salvini Hospital Garbagnate Milanese, Asst Rhodense, Garbagnate Milanese, ITA
| | - Pietro Maria Lombardi
- General Surgery, G. Salvini Hospital Garbagnate Milanese, Asst Rhodense, Garbagnate Milanese, ITA
| | - Mattia Marinelli
- General Surgery, G. Salvini Hospital Garbagnate Milanese, Asst Rhodense, Garbagnate Milanese, ITA
| | - Monica Onorati
- Pathology, G. Salvini Hospital Garbagnate Milanese, Asst Rhodense, Garbagnate Milanese, ITA
| | - Corrado D'Urbano
- General Surgery, G. Salvini Hospital Garbagnate Milanese, Asst Rhodense, Garbagnate Milanese, ITA
| |
Collapse
|
18
|
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: 44] [Impact Index Per Article: 7.3] [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
|
19
|
Saller J, Seydafkan S, Shahid M, Gadara M, Cives M, Eschrich SA, Boulware D, Strosberg JR, Aejaz N, Coppola D. EPB41L5 is Associated With the Metastatic Potential of Low-grade Pancreatic Neuroendocrine Tumors. Cancer Genomics Proteomics 2019; 16:309-318. [PMID: 31467225 PMCID: PMC6727072 DOI: 10.21873/cgp.20136] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 05/15/2019] [Accepted: 05/29/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND/AIM Low-grade pancreatic neuroendocrine tumors (LG-PNETs) behave unpredictably. The aim of the study was to identify biomarkers that predict PNET metastasis to improve treatment selection. PATIENTS AND METHODS Five patients with primary non-metastatic LG-PNETs, six with primary LG-PNETs with synchronous or metachronous metastases (M-PNETs), and six metastatic to liver LG-PNETs (ML-PNETs) from the group of six M-PNET patients were selected. RNA data were normalized using iterative rank-order normalization. Student's t-test identified differentially-expressed genes in LG-PNETs versus M-PNETs. A 2-fold difference in expression was considered to be significant. Results were validated with an independent dataset of LG-PNETs and metastatic LG-PNETs. RESULTS Overall, 195 genes had a >2-fold change (in either direction). A total of 29 genes were differentially overexpressed in M-PNETs. Erythrocyte membrane protein band 4.1-like 5 (EPB41L5) had a 2.07-fold change increase in M-PNETs and the smallest p-value. EPB41L5 was not statistically different between M-PNETs and ML-PNETs. EPB41L5 differential expression between primary and metastatic LG-PNETs was confirmed by immunohistochemistry. CONCLUSION These results support further investigation into whether EPB41L5 is a biomarker of PNETs with high risk for metastases.
Collapse
Affiliation(s)
- James Saller
- Department of Anatomic Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, U.S.A
| | - Shabnam Seydafkan
- Department of Anatomic Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, U.S.A
| | - Mohammad Shahid
- Department of Anatomic Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, U.S.A
| | - Manoj Gadara
- Department of Anatomic Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, U.S.A
| | - Mauro Cives
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, U.S.A
| | - Steven A Eschrich
- Department of Bioinformatics and Biostatistics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, U.S.A
| | - David Boulware
- Department of Bioinformatics and Biostatistics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, U.S.A
| | - Jonathan R Strosberg
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, U.S.A
| | - Nasir Aejaz
- Diagnostic and Experimental Pathology, Eli Lilly and Company, Indianapolis, IN, U.S.A
| | - Domenico Coppola
- Department of Anatomic Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, U.S.A.
- Department of Chemical Biology and Molecular Medicine, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, U.S.A
- Department of Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, U.S.A
- Department of Oncological Sciences, University of South Florida, Tampa, FL, U.S.A
| |
Collapse
|
20
|
Alsadik S, Yusuf S, AL-Nahhas A. Peptide Receptor Radionuclide Therapy for Pancreatic Neuroendocrine Tumours. Curr Radiopharm 2019; 12:126-134. [PMID: 30714538 DOI: 10.2174/1874471012666190201164132] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 05/15/2018] [Accepted: 10/19/2018] [Indexed: 12/29/2022]
Abstract
Background:
The incidence of pancreatic Neuroendocrine Tumours (pNETs) has increased
considerably in the last few decades. The characteristic features of this tumour and the development of
new investigative and therapeutic methods had a great impact on its management.
Objective:
The aim of this review is to investigate the outcome of Peptide Receptor Radionuclide Therapy
(PRRT) in the treatment of pancreatic neuroendocrine tumours.
Methods:
A comprehensive literature search strategy was used based on two databases (SCOPUS, and
PubMed). We considered all studies published in English, evaluating the use of PRRT (177Luteciuim-
DOTA-conjugated peptides and 90Yetrium- DOTA- conjugated peptides) in the treatment of pancreatic
neuroendocrine tumours as a standalone entity or as a subgroup within the wider category of Gastroenteropancreatic
Neuroendocrine Tumours (GEP NETs).
Results:
PRRT was found to be an effective treatment modality as a monotherapy or in combination
with other therapies in the treatment of non-operable and metastatic pNETs where other options are
limited. Complete response was reported to be between 2-6% while partial response was achieved in up
to 60% of cases. Survival analysis was also impressive. Progression Free Survival (PFS) reached a mean
of 34 months and Overall Survival (OS) of 53 months. PRRT also proved to improve patients’ Quality
of Life (QoL). Acute and sub-acute side effects like nephrotoxicity and haematotoxicity are usually mild
and reversible.
Conclusion:
PRRT is well tolerated and effective treatment option for non-operable and/or metastatic
pNETs. Side effects are usually mild and reversible. Larger randomized controlled trails need to be done
to compare PRRT with other treatment modalities and to provide more detailed guidelines regarding
patient selections, the choice of PRRT, follow up and response assessment to maximum potential benefit.
Collapse
Affiliation(s)
- Shahad Alsadik
- Department of Nuclear Medicine, Hammersmith Hospital, Imperial College NHS Trust, London, United Kingdom
| | - Siraj Yusuf
- Department of Nuclear Medicine, Hammersmith Hospital, Imperial College NHS Trust, London, United Kingdom
| | - Adil AL-Nahhas
- Department of Nuclear Medicine, Hammersmith Hospital, Imperial College NHS Trust, London, United Kingdom
| |
Collapse
|
21
|
Varas-Lorenzo M, Cugat E, Capdevila J, Sánchez-Vizcaíno Mengual E. Detection of pancreatic neuroendocrine tumors: 23 years of experience. REVISTA DE GASTROENTEROLOGÍA DE MÉXICO (ENGLISH EDITION) 2019. [DOI: 10.1016/j.rgmxen.2018.08.001] [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: 02/07/2023] Open
|
22
|
Neoplasms of the Neuroendocrine Pancreas: An Update in the Classification, Definition, and Molecular Genetic Advances. Adv Anat Pathol 2019; 26:13-30. [PMID: 29912000 DOI: 10.1097/pap.0000000000000201] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
This review focuses on discussing the main modifications of the recently published 2017 WHO Classification of Neoplasms of the Neuroendocrine Pancreas (panNEN). Recent updates separate pancreatic neuroendocrine tumors into 2 broad categories: well-differentiated pancreatic neuroendocrine tumors (panNET) and poorly differentiated pancreatic neuroendocrine carcinoma (panNEC), and incorporates a new subcategory of "well-differentiated high-grade NET (G3)" to the well-differentiated NET category. This new classification algorithm aims to improve the prediction of clinical outcomes and survival and help clinicians select better therapeutic strategies for patient care and management. In addition, these neuroendocrine neoplasms are capable of producing large quantity of hormones leading to clinical hormone hypersecretion syndromes. These functioning tumors include, insulinomas, glucagonomas, somatostatinomas, gastrinomas, VIPomas, serotonin-producing tumors, and ACTH-producing tumors. Although most panNENs arise as sporadic diseases, a subset of these heterogeneous tumors present as parts on inherited genetic syndromes, such as multiple endocrine neoplasia type 1, von Hippel-Lindau, neurofibromatosis type 1, tuberous sclerosis, and glucagon cell hyperplasia and neoplasia syndromes. Characteristic clinical and morphologic findings for certain functioning and syndromic panNENs should alert both pathologists and clinicians as appropriate patient management and possible genetic counseling may be necessary.
Collapse
|
23
|
Ciaravino V, De Robertis R, Tinazzi Martini P, Cardobi N, Cingarlini S, Amodio A, Landoni L, Capelli P, D'Onofrio M. Imaging presentation of pancreatic neuroendocrine neoplasms. Insights Imaging 2018; 9:943-953. [PMID: 30302635 PMCID: PMC6269331 DOI: 10.1007/s13244-018-0658-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 08/07/2018] [Accepted: 08/28/2018] [Indexed: 02/07/2023] Open
Abstract
Abstract Pancreatic neuroendocrine neoplasms (P-NENs) are the second most common solid pancreatic neoplasms. P-NENs have a wide range of imaging features presentations and they can be detected with typical and atypical imaging presentations. Typical and atypical appearances can be explained by pathologic correlations. P-NENs are generally hypervascular lesions, showing a typical enhancement behavior after contrast media injection during imaging methods, but they could also have different imaging features, creating some difficulty in differential diagnosis. For this reason, radiologists should be aware of different imaging presentations of these neoplasms. Radiological evaluation has a critical role in P-NENs identification, characterization, and staging of these neoplasms, especially in those cases in which surgery is the treatment of choice. The present paper shows, indicating the underlying pathologic correlations, typical and atypical presentations of NENs. Key Points • P-NENs have a wide range of imaging features presentations, typical and atypical. • Pathology could help in better understanding the typical P-NENs appearance at imaging. • P-NENs are generally hypervascular lesions. • Radiological evaluation has a critical role in P-NENs identification and management. • Radiologists should know every type of different imaging presentation of P-NENs to better diagnose these kinds of lesions.
Collapse
Affiliation(s)
- Valentina Ciaravino
- Department of Radiology, University Hospital G.B. Rossi, University of Verona, Verona, Italy. .,Department of Radiology, Hospital Morgagni Pierantoni, Via Carlo Forlanini 4, 47121, Forlì, FC, Italy.
| | - Riccardo De Robertis
- Department of Radiology, Hospital "Casa di Cura Dott. Pederzoli", Peschiera del Garda, Verona, Italy
| | - Paolo Tinazzi Martini
- Department of Radiology, Hospital "Casa di Cura Dott. Pederzoli", Peschiera del Garda, Verona, Italy
| | - Nicolò Cardobi
- Department of Radiology, Hospital "Casa di Cura Dott. Pederzoli", Peschiera del Garda, Verona, Italy
| | - Sara Cingarlini
- Department of Oncology, University Hospital G.B. Rossi, University of Verona, Verona, Italy
| | - Antonio Amodio
- Department of Gastroenterology, University Hospital G.B. Rossi, University of Verona, Verona, Italy
| | - Luca Landoni
- Department of Surgery, University Hospital G.B. Rossi, University of Verona, Verona, Italy
| | - Paola Capelli
- Department of Pathology, University Hospital G.B. Rossi, University of Verona, Verona, Italy
| | - Mirko D'Onofrio
- Department of Radiology, University Hospital G.B. Rossi, University of Verona, Verona, Italy
| |
Collapse
|
24
|
Zhang B, Li C, Sun Z. Long non-coding RNA LINC00346, LINC00578, LINC00673, LINC00671, LINC00261, and SNHG9 are novel prognostic markers for pancreatic cancer. Am J Transl Res 2018; 10:2648-2658. [PMID: 30210701 PMCID: PMC6129514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 12/14/2017] [Indexed: 06/08/2023]
Abstract
Pancreatic cancer (PC) is a devastating human disease with aggressive course and extremely poor prognosis. Long non-coding RNAs (lncRNAs) have been studied to serve as a critical role in pancreatic development and progression. However, little is known about its expression pattern, biological function in PC. In our study, we measured the expression levels of six lncRNAs (LINC00346, LINC00578, LINC00673, LINC00671, LINC00261, and SNHG9) in PC tissues and serums. The results showed that LINC00346, LINC00578, and LINC00673 were highly expressed, whereas LINC00671, LINC00261, and SNHG9 were lowly expressed in PC tissues and serums, and their expression levels were correlated with clinical stages. Results from receiver operating characteristic (ROC) curve analysis showed that the area under the curve (AUC) of six lncRNAs was 0.7073, 0.7837, 0.6093, 0.6057, 0.5712, and 0.5983, respectively. Survival analysis indicated that patients with high expression of LINC00346, LINC00578, or LINC00673 had significantly lower survival rate, while patients with high expression of LINC00671, LINC00261, and SNHG9 had significantly higher survival rate. In addition, we also found that silence of LINC00346, LINC00578 and LINC00673 inhibited PC cell proliferation, and silence of LINC00671, LINC00261, and SNHG9 promoted PC cell proliferation. Therefore, we suggested that LINC00346, LINC00578, LINC00673, LINC00671, LINC00261, and SNHG9 may be novel prognostic markers for PC.
Collapse
Affiliation(s)
- Baogang Zhang
- Department of Endoscopy, China-Japan Union Hospital of Jilin UniversityChangchun 130031, Jilin, China
| | - Changfeng Li
- Department of Endoscopy, China-Japan Union Hospital of Jilin UniversityChangchun 130031, Jilin, China
| | - Zhixia Sun
- Department of Ultrasound, China-Japan Union Hospital of Jilin UniversityChangchun 130031, Jilin, China
| |
Collapse
|
25
|
Clinical application of 99mTc-HYNIC-TOC SPECT/CT in diagnosing and monitoring of pancreatic neuroendocrine neoplasms. Ann Nucl Med 2018; 32:446-452. [PMID: 29926342 DOI: 10.1007/s12149-018-1266-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 05/22/2018] [Indexed: 01/20/2023]
Abstract
OBJECTIVE Our aim of this research was to determine the value of SPECT/CT with 99mTc-HYNIC-TOC for evaluation of the pancreatic masses which were suspected as neuroendocrine neoplasms and follow-up of patients with pancreatic neuroendocrine neoplasms. METHODS We retrospectively analyzed 184 patients who performed 99mTc-HYNIC-TOC SPECT/CT. All the patients were divided into two groups: one for assessment of diagnostic efficiency for pancreatic suspected masses (n = 140) and another for monitoring recurrence after surgery (n = 44). The image findings acquired at 2 h postinjection were compared to final diagnoses from pathological results and clinical follow-up. Then, the correlation between ratios of tumor-to-background (TBR) and tumor grade was analyzed. RESULTS In group 1, 95/140 (67.9%) patients were confirmed as neuroendocrine neoplasms including 85 neuroendocrine tumors and 10 neuroendocrine carcinomas. Patient-based analysis showed that the sensitivity, specificity and accuracy of diagnosing neuroendocrine neoplasms with SPECT/CT were 81.1, 84.4 and 82.1%. There was significant difference of TBRs among G1, G2 and G3 (F = 3.175, P = 0.048). In group 2, 22/44 (50.0%) patients occurred metastasis mainly in liver. The sensitivity, specificity and accuracy of monitoring recurrence were 87.0, 100 and 93.2%. CONCLUSIONS 99mTc-HYNIC-TOC SPECT/CT is a reliable method of diagnosing and monitoring of pancreatic neuroendocrine neoplasms, especially neuroendocrine tumors.
Collapse
|
26
|
Varas-Lorenzo MJ, Cugat E, Capdevila J, Sánchez-Vizcaíno Mengual E. Detection of pancreatic neuroendocrine tumors: 23 years of experience. REVISTA DE GASTROENTEROLOGÍA DE MÉXICO 2018; 84:18-25. [PMID: 29858120 DOI: 10.1016/j.rgmx.2018.02.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 02/14/2018] [Accepted: 02/20/2018] [Indexed: 11/16/2022]
Abstract
INTRODUCTION AND AIMS Neuroendocrine tumors are of great scientific interest, given that they are difficult to diagnose and treat. Despite being relatively rare (< 1/100,000 individuals, 1-2% of the gastrointestinal neoplasias) and indolent, their potential malignancy must not be forgotten. An increase in the number of diagnosed tumors has been observed in recent years. The aim of the present study was to update a published case series of 19 patients suspected of presenting with pancreatic neuroendocrine tumor with 51 current cases, to study and compare the new results with those of the previous case series, as well as with other recent publications from Spain, the United States, China, and India. MATERIALS AND METHODS A retrospective, multicenter case series was conducted on 70 patients (19 cases published in 2011), whose data has been collected over a period of 23 years. The variables analyzed were: age, sex, symptomatology, tumor size, location, metastasis, final diagnosis, and surgery, among others. RESULTS Mean patient age was 55 years and 60% of the patients were men. Disease location was the pancreatic head in 28.5% of the patients and the tail in 27.1%, mean tumor size was 3.9cm (0.2-10cm), 71.4% of the patients had non-functioning tumors, 32.8% had metastases (100% to the liver), 74.2% of the patients were operated on, and actuarial survival was 75%. CONCLUSIONS Differences were observed between the previously published case series and the current results. There was an increase in incidentalomas and non-functioning tumors, but no variation in the overall survival rate. The differences with other case series (age, sex, and tumor location) were dependent on the country where the cases were compiled. The increase in tumors could be related to a higher number of diagnoses made through imaging studies and to the greater sensitivity of the devices employed.
Collapse
Affiliation(s)
- M J Varas-Lorenzo
- Servicio de Aparato Digestivo, Hospital Sanitas CIMA, Barcelona, España; Unidad de Gastroenterología y Hepatología, Hospital Universitario Teknon Quirón Salud, Barcelona, España; Facultat de Ciencias de la Salud, Universitat Oberta de Catalunya (UOC), Barcelona, España
| | - E Cugat
- Servicio de Cirugía General-Aparato Digestivo, Hospital Universitario Teknon Quirón Salud, Barcelona, España
| | - J Capdevila
- Instituto Oncológico, Hospital Universitario Teknon Quirón Salud, Barcelona, España
| | | |
Collapse
|
27
|
A meta-analysis of Prognostic factor of Pancreatic neuroendocrine neoplasms. Sci Rep 2018; 8:7271. [PMID: 29739948 PMCID: PMC5940798 DOI: 10.1038/s41598-018-24072-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 03/26/2018] [Indexed: 02/07/2023] Open
Abstract
Pancreatic neuroendocrine neoplasms (pNENs) are a group of clinically rare and heterogeneous diseases of the pancreas. However, the prognostic factors for this disease in patients still remain controversial. The purpose of our study is to evaluate the predictive roles of those prognostic factors for pNENs. All related articles published until Sep 17, 2017 were identified via PubMed, EMBASE, Web of Science, Ovid and the Cochrane Library. Studies that examined the prognostic factors of pNENs were enrolled. 17 articles (2822 patients) were finally included in this study. The pooled data suggested that patients with positive surgical resection margin and lymph node, advanced G stage and TMN stage, organ metastasis, vascular invasion and the necrosis of specimens had a decreased overall survival for pNENs. Similarly, patients with functional tumors might have a poor prognosis. However, age, gender, surgical type and size of tumor could not be regarded as prognostic factors for pNENs. Our analytic data demonstrated that surgical resection margin, G stage, TMN stage, lymph node, metastasis, vascular invasion and the necrosis could be prognostic factors for pNENs. Our study may assist doctors to screen patients with different prognosis more efficiently during follow-up and select appropriate treatment measures.
Collapse
|
28
|
Yusuf S, Alsadik S, AL-Nahhas A. Peptide receptor radionuclide therapy for neuroendocrine tumours. Clin Transl Imaging 2018. [DOI: 10.1007/s40336-018-0267-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
29
|
Lykoudis PM, Partelli S, Muffatti F, Caplin M, Falconi M, Fusai GK. Treatment challenges in and outside a specialist network setting: Pancreatic neuroendocrine tumours. Eur J Surg Oncol 2017; 45:46-51. [PMID: 29126671 DOI: 10.1016/j.ejso.2017.08.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 08/18/2017] [Indexed: 01/14/2023] Open
Abstract
Pancreatic Neuroendocrine Neoplasms comprise a group of rare tumours with special biology, an often indolent behaviour and particular diagnostic and therapeutic requirements. The specialized biochemical tests and radiological investigations, the complexity of surgical options and the variety of medical treatments that require individual tailoring, mandate a multidisciplinary approach that can be optimally achieved through an organized network. The present study describes currents concepts in the management of these tumours as well as an insight into the challenges of delivering the pathway in and outside a Network.
Collapse
Affiliation(s)
- Panagis M Lykoudis
- Department of Hepato-Pancreato-Biliary Surgery & Liver Transplantation, Royal Free Hospital, London, UK.
| | - Stefano Partelli
- Pancreatic Surgery Unit, Pancreas Translational & Research Institute, Scientific Institute San Raffaele Hospital & University "Vita e Salute", Milan, Italy
| | - Francesca Muffatti
- Pancreatic Surgery Unit, Pancreas Translational & Research Institute, Scientific Institute San Raffaele Hospital & University "Vita e Salute", Milan, Italy
| | - Martyn Caplin
- Department of Gastroenterology and G.I. & Tumour Neuroendocrinology, Royal Free Hospital, London, UK
| | - Massimo Falconi
- Pancreatic Surgery Unit, Pancreas Translational & Research Institute, Scientific Institute San Raffaele Hospital & University "Vita e Salute", Milan, Italy
| | - Giuseppe K Fusai
- Department of Hepato-Pancreato-Biliary Surgery & Liver Transplantation, Royal Free Hospital, London, UK
| | | |
Collapse
|