1
|
Di Franco M, Fortunati E, Zanoni L, Fanti S, Ambrosini V. The role of combined FDG and SST PET/CT in neuroendocrine tumors. J Neuroendocrinol 2025; 37:e13474. [PMID: 39575824 PMCID: PMC11919475 DOI: 10.1111/jne.13474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 10/08/2024] [Accepted: 11/10/2024] [Indexed: 01/06/2025]
Abstract
Somatostatin receptor (SST) positron emission tomography with computed tomography (PET/CT) is the gold standard for functional imaging of neuroendocrine tumors (NETs), but FDG PET/CT is increasingly recognized for its prognostic value, particularly for higher-grade NETs and to detect disease heterogeneity. Despite the established role of pathological grading, clinical heterogeneity within the tumor burden often complicates accurate prognostication. Evidence suggests FDG PET/CT can outperform WHO grading in predicting outcomes by identifying aggressive, undifferentiated tumor clones that influence long-term prognosis and treatment decisions. Several grading systems integrating both SST and FDG PET/CT have been proposed to better capture tumor heterogeneity and guide clinical management. Studies demonstrate that FDG PET/CT can influence management in a significant subset of patients, although variably reported. Its use remains variable across centers, also affected by different reimbursement policies and local clinical practices. This review explores the indications to FDG PET/CT in NET and the clinical impact of combined SST and FDG PET/CT imaging.
Collapse
Affiliation(s)
- Martina Di Franco
- Nuclear MedicineAlma Mater Studiorum, University of BolognaBolognaItaly
| | - Emilia Fortunati
- Nuclear MedicineIRCCS, Azienda Ospedaliero‐Universitaria di BolognaBolognaItaly
| | - Lucia Zanoni
- Nuclear MedicineIRCCS, Azienda Ospedaliero‐Universitaria di BolognaBolognaItaly
| | - Stefano Fanti
- Nuclear MedicineAlma Mater Studiorum, University of BolognaBolognaItaly
- Nuclear MedicineIRCCS, Azienda Ospedaliero‐Universitaria di BolognaBolognaItaly
| | - Valentina Ambrosini
- Nuclear MedicineAlma Mater Studiorum, University of BolognaBolognaItaly
- Nuclear MedicineIRCCS, Azienda Ospedaliero‐Universitaria di BolognaBolognaItaly
| |
Collapse
|
2
|
Lun T, Rosty C, Palamuthusingam P. The first case of combined pancreatic neuroendocrine tumour and neuroendocrine carcinoma. J Surg Case Rep 2024; 2024:rjae746. [PMID: 39678483 PMCID: PMC11645528 DOI: 10.1093/jscr/rjae746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 11/12/2024] [Indexed: 12/17/2024] Open
Abstract
Pancreatic neuroendocrine neoplasms are currently thought to originate from distinct progenitor cells that cannot differentiate into each other. We present the first reported case of a combined pancreatic neuroendocrine tumour and neuroendocrine carcinoma in a 58-year-old man who was investigated for abdominal pain and constipation. Imaging revealed a large left upper quadrant mass infiltrating the pancreatic body and tail, splenic hilum, and posterior stomach wall, with five hepatic metastases. This was treated with neoadjuvant and adjuvant chemotherapy, debulking surgery, and lutetium-177-DOTATATE peptide receptor radionuclide therapy. This case emphasises the importance of molecular imaging, meticulous microscopic examination, and multidisciplinary discussion for accurate diagnoses, improved prognostication, and efficacious treatment.
Collapse
Affiliation(s)
- Tegan Lun
- Department of Surgery, Townsville University Hospital, Townsville, 4814, QLD, Australia
| | - Christophe Rosty
- Envoi Specialist Pathologists, Brisbane, 4059, QLD, Australia
- The University of Queensland, Brisbane, 4072, QLD, Australia
| | - Pranavan Palamuthusingam
- Department of Surgery, Royal Brisbane and Women’s Hospital, Brisbane, 4006, QLD, Australia
- Department of Surgery, The Wesley Hospital, Brisbane, 4066, QLD, Australia
| |
Collapse
|
3
|
Metser U, Nunez JE, Chan D, Kulanthaivelu R, Murad V, Santiago AT, Singh S. Dual Somatostatin Receptor/ 18F-FDG PET/CT Imaging in Patients with Well-Differentiated, Grade 2 and 3 Gastroenteropancreatic Neuroendocrine Tumors. J Nucl Med 2024; 65:1591-1596. [PMID: 39266292 DOI: 10.2967/jnumed.124.267982] [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: 04/22/2024] [Accepted: 08/02/2024] [Indexed: 09/14/2024] Open
Abstract
Our purpose was to prospectively assess the distribution of NETPET scores in well-differentiated (WD) grade 2 and 3 gastroenteropancreatic (GEP) neuroendocrine tumors (NETs) and to determine the impact of the NETPET score on clinical management. Methods: This single-arm, institutional ethics review board-approved prospective study included 40 patients with histologically proven WD GEP NETs. 68Ga-DOTATATE PET and 18F-FDG PET were performed within 21 d of each other. NETPET scores were evaluated qualitatively by 2 reviewers, with up to 10 marker lesions selected for each patient. The quantitative parameters that were evaluated included marker lesion SUVmax for each tracer; 18F-FDG/68Ga-DOTATATE SUVmax ratios; functional tumor volume (FTV) and metabolic tumor volume (MTV) on 68Ga-DOTATATE and 18F-FDG PET, respectively; and FTV/MTV ratios. The treatment plan before and after 18F-FDG PET was recorded. Results: There were 22 men and 18 women (mean age, 60.8 y) with grade 2 (n = 24) or grade 3 (n = 16) tumors and a mean Ki-67 index of 16.1%. NETPET scores of P0, P1, P2A, P2B, P3B, P4B, and P5 were documented in 2 (5%), 5 (12.5%), 5 (12.5%) 20 (50%), 2 (5%), 4 (10%), and 2 (5%) patients, respectively. No association was found between the SUVmax of target lesions on 68Ga-DOTATATE and the SUVmax of target lesions on 18F-FDG PET (P = 0.505). 18F-FDG/68Ga-DOTATATE SUVmax ratios were significantly lower for patients with low (P1-P2) primary NETPET scores than for those with high (P3-P5) primary NETPET scores (mean ± SD, 0.20 ± 0.13 and 1.68 ± 1.44, respectively; P < 0.001). MTV on 18F-FDG PET was significantly lower for low primary NETPET scores than for high ones (mean ± SD, 464 ± 601 cm3 and 66 ± 114 cm3, respectively; P = 0.005). A change in the type of management was observed in 42.5% of patients after 18F-FDG PET, with the most common being a change from systemic therapy to peptide receptor radionuclide therapy and from debulking surgery to systemic therapy. Conclusion: There was a heterogeneous distribution of NETPET scores in patients with WD grade 2 and 3 GEP NETs, with more than 1 in 5 patients having a high NETPET score and a frequent change in management after 18F-FDG PET. Quantitative parameters including 18F-FDG/68Ga-DOTATATE SUVmax ratios in target lesions and FTV/MTV ratios can discriminate between patients with high and low NETPET scores.
Collapse
Affiliation(s)
- Ur Metser
- University Medical Imaging Toronto; University Health Network, Sinai Health Systems, Women's College Hospital; and University of Toronto, Toronto, Ontario, Canada;
| | - Jose E Nunez
- Division of Medical Oncology, University of Toronto; and Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - David Chan
- Department of Medical Oncology, Royal North Shore Hospital, St. Leonards, New South Wales, Australia; and Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, University of Sydney, Sydney, New South Wales, Australia; and
| | - Roshini Kulanthaivelu
- University Medical Imaging Toronto; University Health Network, Sinai Health Systems, Women's College Hospital; and University of Toronto, Toronto, Ontario, Canada
| | - Vanessa Murad
- University Medical Imaging Toronto; University Health Network, Sinai Health Systems, Women's College Hospital; and University of Toronto, Toronto, Ontario, Canada
| | - Anna T Santiago
- Department of Biostatistics, University Health Network, Toronto, Ontario, Canada
| | - Simron Singh
- Division of Medical Oncology, University of Toronto; and Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| |
Collapse
|
4
|
Padmanabhan Nair Sobha R, Jensen CT, Waters R, Calimano-Ramirez LF, Virarkar MK. Appendiceal Neuroendocrine Neoplasms: A Comprehensive Review. J Comput Assist Tomogr 2024; 48:545-562. [PMID: 37574653 DOI: 10.1097/rct.0000000000001528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
ABSTRACT Appendiceal neuroendocrine neoplasm (NEN) is the most common adult appendiceal malignant tumor, constituting 16% of gastrointestinal NENs. They are versatile tumors with varying morphology, immunohistochemistry, secretory properties, and cancer genomics. They are slow growing and clinically silent, to begin with, or present with features of nonspecific vague abdominal pain. Most acute presentations are attributed clinically to appendicitis, with most cases detected incidentally on pathology after an appendectomy. Approximately 40% of them present clinically with features of hormonal excess, which is likened to the functional secretory nature of their parent cell of origin. The symptoms of carcinoid syndrome render their presence clinically evident. However, slow growing and symptomatically silent in its initial stages, high-grade neuroendocrine tumors and neuroendocrine carcinomas of the appendix are aggressive and usually have hepatic and lymph node metastasis at presentation. This review article focuses on imaging characteristics, World Health Organization histopathological classification and grading, American Joint Committee on Cancer/Union or International Cancer Control, European Neuroendocrine Tumor Society staging, European Neuroendocrine Tumor Society standardized guidelines for reporting, data interpretation, early-stage management protocols, and advanced-stage appendiceal NENs. Guidelines are also set for the follow-up and reassessment. The role of targeted radiotherapy, chemotherapy, and high-dose somatostatin analogs in treating advanced disease are discussed, along with types of ablative therapies and liver transplantation for tumor recurrence. The search for newer location-specific biomarkers in NEN is also summarized. Regarding the varying aggressiveness of the tumor, there is a scope for research in the field, with plenty of data yet to be discovered.
Collapse
Affiliation(s)
| | - Corey T Jensen
- From the Department of Radiology, University of Texas MD Anderson Cancer Center
| | - Rebecca Waters
- Department of Pathology and Lab Medicine MD Anderson Cancer Center, Houston, TX
| | | | - Mayur K Virarkar
- Department of Radiology, University of Florida College of Medicine, Jacksonville, FL
| |
Collapse
|
5
|
Massironi S, Franchina M, Ippolito D, Elisei F, Falco O, Maino C, Pagni F, Elvevi A, Guerra L, Invernizzi P. Improvements and future perspective in diagnostic tools for neuroendocrine neoplasms. Expert Rev Endocrinol Metab 2024; 19:349-366. [PMID: 38836602 DOI: 10.1080/17446651.2024.2363537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 05/30/2024] [Indexed: 06/06/2024]
Abstract
INTRODUCTION Neuroendocrine neoplasms (NENs) represent a complex group of tumors arising from neuroendocrine cells, characterized by heterogeneous behavior and challenging diagnostics. Despite advancements in medical technology, NENs present a major challenge in early detection, often leading to delayed diagnosis and variable outcomes. This review aims to provide an in-depth analysis of current diagnostic methods as well as the evolving and future directions of diagnostic strategies for NENs. AREA COVERED The review extensively covers the evolution of diagnostic tools for NENs, from traditional imaging and biochemical tests to advanced genomic profiling and next-generation sequencing. The emerging role of technologies such as artificial intelligence, machine learning, and liquid biopsies could improve diagnostic precision, as could the integration of imaging modalities such as positron emission tomography (PET)/magnetic resonance imaging (MRI) hybrids and innovative radiotracers. EXPERT OPINION Despite progress, there is still a significant gap in the early diagnosis of NENs. Bridging this diagnostic gap and integrating advanced technologies and precision medicine are crucial to improving patient outcomes. However, challenges such as low clinical awareness, limited possibility of noninvasive diagnostic tools and funding limitations for rare diseases like NENs are acknowledged.
Collapse
Affiliation(s)
- Sara Massironi
- Division of Gastroenterology, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Marianna Franchina
- Division of Gastroenterology, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Davide Ippolito
- Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
- Department of Diagnostic Radiology, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Federica Elisei
- Division of Nuclear Medicine, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Olga Falco
- Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Cesare Maino
- Department of Diagnostic Radiology, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Fabio Pagni
- Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
- Division of Pathology, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Alessandra Elvevi
- Division of Gastroenterology, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Luca Guerra
- Division of Nuclear Medicine, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Pietro Invernizzi
- Division of Gastroenterology, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| |
Collapse
|
6
|
Santo G, Di Santo G, Virgolini I. Peptide Receptor Radionuclide Therapy of Neuroendocrine Tumors: Agonist, Antagonist and Alternatives. Semin Nucl Med 2024; 54:557-569. [PMID: 38490913 DOI: 10.1053/j.semnuclmed.2024.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 02/14/2024] [Accepted: 02/14/2024] [Indexed: 03/17/2024]
Abstract
Peptide receptor radionuclide therapy (PRRT) today is a well-established treatment strategy for patients with neuroendocrine tumors (NET). First performed already more than 30 years ago, PRRT was incorporated only in recent years into the major oncology guidelines, based on its proven efficacy and safety in clinical trials. Following the phase 3 NETTER-1 trial, which led to the final registration of the radiopharmaceutical Luthatera® for G1/G2 NET patients in 2017, the long-term results of the phase 3 NETTER-2 trial may pave the way for a new treatment option also for advanced G2/G3 patients as first-line therapy. The growing knowledge about the synergistic effect of combined therapies could also allow alternative (re)treatment options for NET patients, in order to create a tailored treatment strategy. The evolving thera(g)nostic concept could be applied for the identification of patients who might benefit from different image-guided treatment strategies. In this scenario, the use of dual tracer PET/CT in NET patients, using both [18F]F-FDG/[68Ga]Ga-DOTA-somatostatin analog (SSA) for diagnosis and follow-up, is under discussion and could also result in a powerful prognostic tool. In addition, alternative strategies based on different metabolic pathways, radioisotopes, or combinations of different medical approaches could be applied. A number of different promising "doors" could thus open in the near future for the treatment of NET patients - and the "key" will be thera(g)nostic!
Collapse
Affiliation(s)
- Giulia Santo
- Department of Nuclear Medicine, Medical University of Innsbruck, Innsbruck, Austria; Department of Experimental and Clinical Medicine, "Magna Graecia" University of Catanzaro, Catanzaro, Italy
| | - Gianpaolo Di Santo
- Department of Nuclear Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Irene Virgolini
- Department of Nuclear Medicine, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
7
|
Bodei L, Jayaprakasam VS, Ying Wong BZ, Aparici CM. Neuroendocrine Tumors: Beta Labeled Radiopeptides. PET Clin 2024; 19:e1-e11. [PMID: 40199623 DOI: 10.1016/j.cpet.2024.06.003] [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] [Indexed: 04/10/2025]
Abstract
Peptide receptor radionuclide therapy (PRRT) consists of administrating a radiolabeled octreotide derivative that targets somatostatin receptors present on the cell membrane of neuroendocrine tumor cells. Although PRRT was initially performed with 90Y-peptides, currently 177Lu-peptides represent the predominant form of treatment. PRRT results in significant tumor and symptomatic control in patients. Like with other available systemic therapies, responses are relatively short-lived. Several new peptides and strategies to improve the efficacy and tolerability of PRRT have been proposed. A critical step is individualizing treatments based on specific dosimetric estimates for the tumor and normal organs, and determining tissue radiosensitivity.
Collapse
Affiliation(s)
- Lisa Bodei
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Radiology, Weill Cornell Medical College of Cornell University, New York, NY, USA.
| | - Vetri Sudar Jayaprakasam
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Radiology, Weill Cornell Medical College of Cornell University, New York, NY, USA
| | | | - Carina Mari Aparici
- Division of Nuclear Medicine, Department of Radiology, University of Stanford, Stanford, CA, USA
| |
Collapse
|
8
|
Mileva M, Marin G, Levillain H, Artigas C, Van Bogaert C, Marin C, Danieli R, Deleporte A, Picchia S, Stathopoulos K, Jungels C, Vanderlinden B, Paesmans M, Ameye L, Critchi G, Taraji-Schiltz L, Velghe C, Wimana Z, Bali M, Hendlisz A, Flamen P, Karfis I. Prediction of 177Lu-DOTATATE PRRT Outcome Using Multimodality Imaging in Patients with Gastroenteropancreatic Neuroendocrine Tumors: Results from a Prospective Phase II LUMEN Study. J Nucl Med 2024; 65:236-244. [PMID: 38164576 DOI: 10.2967/jnumed.123.265987] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 10/25/2023] [Indexed: 01/03/2024] Open
Abstract
Our objective was to predict the outcome of peptide receptor radionuclide therapy (PRRT) using multimodality imaging and tumor dosimetry on gastroenteropancreatic neuroendocrine tumor (GEP-NET) lesions and patients. Methods: This prospective study included patients with progressive GEP-NETs. Treatment consisted of 4 cycles of 7.4 GBq of 177Lu-DOTATATE. Imaging parameters were measured on 68Ga-DOTATATE PET/CT (SUVmax/mean, somatostatin receptor [SSTR] tumor volume [TV], total lesion SSTR expression, and tumor-to-blood and tumor-to-spleen ratios), 18F-FDG PET/CT (SUVmax/mean, metabolically active TV, and total lesion glycolysis), and diffusion-weighted MRI (apparent diffusion coefficient) in a maximum of 5 target lesions per patient at approximately 10 wk after each injection. Tumor dosimetry was performed using SPECT/CT at 3 time points for every cycle. Baseline imaging parameters, their relative changes after PRRT cycle 1 (C1), and the tumor-absorbed dose at C1 were correlated with lesion morphologic outcome. The average values of the imaging parameters and the minimal, maximal, and mean C1 tumor-absorbed dose in each patient were tested for association with progression-free survival (PFS) and best objective response (RECIST 1.1). Results: In the 37 patients, the median PFS was 28 mo. Eleven of the 37 (30%) achieved a partial response (RECIST 1.1). After a median follow-up of 57 mo, the median time to lesion progression had not been reached in 84 morphologically evaluable lesions, with only 12 (14%) progressing (size increase ≥ 20% from baseline). Patients receiving a minimal C1 dose of 35 Gy in all target lesions exhibited a significantly longer PFS (48.1 vs. 26.2 mo; hazard ratio, 0.37; 95% CI, 0.17-0.82; P = 0.02). Volumetric 68Ga-DOTATATE PET parameters correlated with lesion and patient outcome: patients with an SSTR TV decrease of more than 10% after C1 had a longer PFS (51.3 vs. 22.8 mo; hazard ratio, 0.35; 95% CI, 0.16-0.75; P = 0.003). There was no statistical evidence of an association between other dosimetric or imaging parameters and the lesion or patient outcome. Conclusion: Minimal tumor-absorbed dose at C1 is predictive of outcome in patients with GEP-NETs treated with PRRT, providing a basis for personalized dosimetry-guided treatment strategies. An SSTR TV decrease after C1 could be used for early therapy response assessment as a predictor of PRRT outcome.
Collapse
Affiliation(s)
- Magdalena Mileva
- Nuclear Medicine Department, Institut Jules Bordet, ENETS Centre of Excellence, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| | - Gwennaëlle Marin
- Medical Physics Department, Institut Jules Bordet, ENETS Centre of Excellence, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| | - Hugo Levillain
- Medical Physics Department, Institut Jules Bordet, ENETS Centre of Excellence, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| | - Carlos Artigas
- Nuclear Medicine Department, Institut Jules Bordet, ENETS Centre of Excellence, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| | - Camille Van Bogaert
- Nuclear Medicine Department, CUB-Hôpital Erasme, ENETS Centre of Excellence, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| | - Clémentine Marin
- Medical Physics Department, Institut Jules Bordet, ENETS Centre of Excellence, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| | - Rachele Danieli
- Medical Physics Department, Institut Jules Bordet, ENETS Centre of Excellence, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| | - Amelie Deleporte
- Medical Oncology Department, Institut Jules Bordet, ENETS Centre of Excellence, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| | - Simona Picchia
- Radiology Department, Institut Jules Bordet, ENETS Centre of Excellence, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| | - Konstantinos Stathopoulos
- Radiology Department, Institut Jules Bordet, ENETS Centre of Excellence, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| | - Christiane Jungels
- Medical Oncology Department, Institut Jules Bordet, ENETS Centre of Excellence, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| | - Bruno Vanderlinden
- Medical Physics Department, Institut Jules Bordet, ENETS Centre of Excellence, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| | - Marianne Paesmans
- Data Center, Institut Jules Bordet, ENETS Centre of Excellence, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium; and
| | - Lieveke Ameye
- Data Center, Institut Jules Bordet, ENETS Centre of Excellence, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium; and
| | - Gabriela Critchi
- Nuclear Medicine Department, Institut Jules Bordet, ENETS Centre of Excellence, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| | - Loubna Taraji-Schiltz
- Nuclear Medicine Department, Institut Jules Bordet, ENETS Centre of Excellence, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| | - Chloe Velghe
- Data Center, Institut Jules Bordet, ENETS Centre of Excellence, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium; and
| | - Zéna Wimana
- Nuclear Medicine Department, Institut Jules Bordet, ENETS Centre of Excellence, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
- Radiopharmacy Department, Institut Jules Bordet, ENETS Centre of Excellence, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| | - Maria Bali
- Radiology Department, Institut Jules Bordet, ENETS Centre of Excellence, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| | - Alain Hendlisz
- Medical Oncology Department, Institut Jules Bordet, ENETS Centre of Excellence, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| | - Patrick Flamen
- Nuclear Medicine Department, Institut Jules Bordet, ENETS Centre of Excellence, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| | - Ioannis Karfis
- Nuclear Medicine Department, Institut Jules Bordet, ENETS Centre of Excellence, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium;
| |
Collapse
|
9
|
Piscopo L, Zampella E, Pellegrino S, Volpe F, Nappi C, Gaudieri V, Fonti R, Vecchio SD, Cuocolo A, Klain M. Diagnosis, Management and Theragnostic Approach of Gastro-Entero-Pancreatic Neuroendocrine Neoplasms. Cancers (Basel) 2023; 15:3483. [PMID: 37444593 DOI: 10.3390/cancers15133483] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/23/2023] [Accepted: 06/30/2023] [Indexed: 07/15/2023] Open
Abstract
Gastro-entero-pancreatic neuroendocrine neoplasms (GEP-NENs) constitute an ideal target for radiolabeled somatostatin analogs. The theragnostic approach is able to combine diagnosis and therapy by the identification of a molecular target that can be diagnosed and treated with the same radiolabeled compound. During the last years, advances in functional imaging with the introduction of somatostatin analogs and peptide receptor radionuclide therapy, have improved the diagnosis and treatment of GEP-NENs. Moreover, PET/CT imaging with 18F-FDG represents a complementary tool for prognostic evaluation of patients with GEP-NENs. In the field of personalized medicine, the theragnostic approach has emerged as a promising tool in diagnosis and management of patients with GEP-NENs. The aim of this review is to summarize the current evidence on diagnosis and management of patients with GEP-NENs, focusing on the theragnostic approach.
Collapse
Affiliation(s)
- Leandra Piscopo
- Department of Advanced Biomedical Sciences, University of Naples, Federico II, 80131 Naples, Italy
| | - Emilia Zampella
- Department of Advanced Biomedical Sciences, University of Naples, Federico II, 80131 Naples, Italy
| | - Sara Pellegrino
- Department of Advanced Biomedical Sciences, University of Naples, Federico II, 80131 Naples, Italy
| | - Fabio Volpe
- Department of Advanced Biomedical Sciences, University of Naples, Federico II, 80131 Naples, Italy
| | - Carmela Nappi
- Department of Advanced Biomedical Sciences, University of Naples, Federico II, 80131 Naples, Italy
| | - Valeria Gaudieri
- Department of Advanced Biomedical Sciences, University of Naples, Federico II, 80131 Naples, Italy
| | - Rosa Fonti
- Department of Advanced Biomedical Sciences, University of Naples, Federico II, 80131 Naples, Italy
| | - Silvana Del Vecchio
- Department of Advanced Biomedical Sciences, University of Naples, Federico II, 80131 Naples, Italy
| | - Alberto Cuocolo
- Department of Advanced Biomedical Sciences, University of Naples, Federico II, 80131 Naples, Italy
| | - Michele Klain
- Department of Advanced Biomedical Sciences, University of Naples, Federico II, 80131 Naples, Italy
| |
Collapse
|
10
|
Haidar M, Al Mahmasani L, Chehade L, Elias C, El Jebai M, Temraz S, Charafeddine M, Al Darazi M, Shamseddine A. Well-differentiated gastro-entero-pancreatic neuroendocrine tumors with positive FDG-PET/CT: a retrospective chart review. Nucl Med Commun 2023; 44:471-479. [PMID: 36897058 DOI: 10.1097/mnm.0000000000001683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
Abstract
PURPOSE Rarely, well-differentiated gastro-entero-pancreatic neuroendocrine tumors (GEP NETs) can have positive uptake on 18F-fluorodeoxyglucose-PET/computerized tomography ( 18 F-FDG-PET/CT), with or without a positive 68 Ga-PET/CT. We aim to evaluate the diagnostic role of 18 F-FDG-PET/CT in patients with well-differentiated GEP NETs. METHODS We retrospectively reviewed a chart of patients diagnosed with GEP NETs between 2014 and 2021, at the American University of Beirut Medical Center, who have low (G1; Ki-67 ≤2) or intermediate (G2; and Ki-67 >2-≤20) well-differentiated tumors with positive findings on FDG-PET/CT. The primary endpoint is progression-free survival (PFS) compared to historical control, and the secondary outcome is to describe their clinical outcome. RESULTS In total 8 out of 36 patients with G1 or G2 GEP NET met the inclusion criteria for this study. The median age was 60 years (range 51-75 years) and 75% were male. One patient (12.5%) had a G1 tumor whereas 7 (87.5%) had G2, and seven patients were stage IV. The primary tumor was intestinal in 62.5% of the patients and pancreatic in 37.5%. Seven patients had both 18 F-FDG-PET/CT and 68 Ga-PET/CT positive and one patient had a positive 18 F-FDG-PET/CT and negative 68 Ga-PET/CT. Median and mean PFS in patients positive for both 68 Ga-PET/CT and 18 F-FDG-PET/CT were 49.71 months and 37.5 months (95% CI, 20.7-54.3), respectively. PFS in these patients is lower than that reported in the literature for G1/G2 NETs with positive 68 Ga-PET/CT and negative FDG-PET/CT (37.5 vs. 71 months; P = 0.0217). CONCLUSION A new prognostic score that includes 18 F-FDG-PET/CT in G1/G2 GEP NETs could identify more aggressive tumors.
Collapse
Affiliation(s)
| | - Layal Al Mahmasani
- Department of Internal Medicine, Naef K. Basile Cancer Institute, American University of Beirut Medical Center, Beirut, Lebanon
| | - Laudy Chehade
- Department of Internal Medicine, Naef K. Basile Cancer Institute, American University of Beirut Medical Center, Beirut, Lebanon
| | - Charbel Elias
- Department of Internal Medicine, Naef K. Basile Cancer Institute, American University of Beirut Medical Center, Beirut, Lebanon
| | | | - Sally Temraz
- Department of Internal Medicine, Naef K. Basile Cancer Institute, American University of Beirut Medical Center, Beirut, Lebanon
| | - Maya Charafeddine
- Department of Internal Medicine, Naef K. Basile Cancer Institute, American University of Beirut Medical Center, Beirut, Lebanon
| | - Monita Al Darazi
- Department of Internal Medicine, Naef K. Basile Cancer Institute, American University of Beirut Medical Center, Beirut, Lebanon
| | - Ali Shamseddine
- Department of Internal Medicine, Naef K. Basile Cancer Institute, American University of Beirut Medical Center, Beirut, Lebanon
| |
Collapse
|
11
|
Bocchini M, Tazzari M, Ravaioli S, Piccinini F, Foca F, Tebaldi M, Nicolini F, Grassi I, Severi S, Calogero RA, Arigoni M, Schrader J, Mazza M, Paganelli G. Circulating hsa-miR-5096 predicts 18F-FDG PET/CT positivity and modulates somatostatin receptor 2 expression: a novel miR-based assay for pancreatic neuroendocrine tumors. Front Oncol 2023; 13:1136331. [PMID: 37287922 PMCID: PMC10242108 DOI: 10.3389/fonc.2023.1136331] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 04/25/2023] [Indexed: 06/09/2023] Open
Abstract
Gastro-entero-pancreatic neuroendocrine tumors (GEP-NETs) are rare diseases encompassing pancreatic (PanNETs) and ileal NETs (SINETs), characterized by heterogeneous somatostatin receptors (SSTRs) expression. Treatments for inoperable GEP-NETs are limited, and SSTR-targeted Peptide Receptor Radionuclide Therapy (PRRT) achieves variable responses. Prognostic biomarkers for the management of GEP-NET patients are required. 18F-FDG uptake is a prognostic indicator of aggressiveness in GEP-NETs. This study aims to identify circulating and measurable prognostic miRNAs associated with 18F-FDG-PET/CT status, higher risk and lower response to PRRT. Methods Whole miRNOme NGS profiling was conducted on plasma samples obtained from well-differentiated advanced, metastatic, inoperable G1, G2 and G3 GEP-NET patients enrolled in the non-randomized LUX (NCT02736500) and LUNET (NCT02489604) clinical trials prior to PRRT (screening set, n= 24). Differential expression analysis was performed between 18F-FDG positive (n=12) and negative (n=12) patients. Validation was conducted by Real Time quantitative PCR in two distinct well-differentiated GEP-NET validation cohorts, considering the primary site of origin (PanNETs n=38 and SINETs n=30). The Cox regression was applied to assess independent clinical parameters and imaging for progression-free survival (PFS) in PanNETs. In situ RNA hybridization combined with immunohistochemistry was performed to simultaneously detect miR and protein expression in the same tissue specimens. This novel semi-automated miR-protein protocol was applied in PanNET FFPE specimens (n=9). In vitro functional experiments were performed in PanNET models. Results While no miRNAs emerged to be deregulated in SINETs, hsa-miR-5096, hsa-let-7i-3p and hsa-miR-4311 were found to correlate with 18F-FDG-PET/CT in PanNETs (p-value:<0.005). Statistical analysis has shown that, hsa-miR-5096 can predict 6-month PFS (p-value:<0.001) and 12-month Overall Survival upon PRRT treatment (p-value:<0.05), as well as identify 18F-FDG-PET/CT positive PanNETs with worse prognosis after PRRT (p-value:<0.005). In addition, hsa-miR-5096 inversely correlated with both SSTR2 expression in PanNET tissue and with the 68Gallium-DOTATOC captation values (p-value:<0.05), and accordingly it was able to decrease SSTR2 when ectopically expressed in PanNET cells (p-value:<0.01). Conclusions hsa-miR-5096 well performs as a biomarker for 18F-FDG-PET/CT and as independent predictor of PFS. Moreover, exosome-mediated delivery of hsa-miR-5096 may promote SSTR2 heterogeneity and thus resistance to PRRT.
Collapse
Affiliation(s)
- Martine Bocchini
- Immunotherapy, Cell Therapy and Biobank (ITCB), IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Marcella Tazzari
- Immunotherapy, Cell Therapy and Biobank (ITCB), IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Sara Ravaioli
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Filippo Piccinini
- Scientific Directorate, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Flavia Foca
- Unit of Biostatistics and Clinical Trials, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Michela Tebaldi
- Unit of Biostatistics and Clinical Trials, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Fabio Nicolini
- Immunotherapy, Cell Therapy and Biobank (ITCB), IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Ilaria Grassi
- Nuclear Medicine and Radiometabolic Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Stefano Severi
- Nuclear Medicine and Radiometabolic Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Raffaele Adolfo Calogero
- Molecular Biotechnology Center, Department of Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Maddalena Arigoni
- Molecular Biotechnology Center, Department of Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Joerg Schrader
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Massimiliano Mazza
- Immunotherapy, Cell Therapy and Biobank (ITCB), IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Giovanni Paganelli
- Nuclear Medicine and Radiometabolic Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| |
Collapse
|
12
|
Neuroendocrine Tumor Therapy Response Assessment. PET Clin 2023; 18:267-286. [PMID: 36858748 DOI: 10.1016/j.cpet.2022.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
Peptide receptor radionuclide therapy has become an integral part of management of neuroendocrine neoplasms. Gallium-68- and lutetium-177-labeled somatostatin receptor analogues have replaced yttrium-90- and 111-indium-based tracers. Several newer targeted therapies are also being used in clinical and research settings. It is imperative to accurately evaluate the response to these agents. The characteristics of NENs and the response patterns of the targeted therapies make response assessment in this group challenging. This article provides an overview of the strengths and weaknesses of the various biomarkers available for response assessment.
Collapse
|
13
|
Khatri W, Spiro E, Henderson A, Rowe SP, Solnes LB. Gastro-Entero-Pancreatic Tumors: FDG Positron Emission Tomography/Computed Tomography. PET Clin 2023; 18:243-250. [PMID: 36707371 DOI: 10.1016/j.cpet.2022.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Gastro-entero-pancreatic tumors comprise a group of heterogenous neoplasms, with medical imaging being paramount in the diagnosis, staging, and treatment planning of these tumors. Moreover, with the advent of newer radiopharmaceuticals, such as 68 Ga-labeled and 64 Cu-labeled somatostatin analogs (eg, 68 Ga-DOTATOC, 68 Ga-DOTATATE, 68 Ga-DOTANOC, and 64Cu-DOTATATE) that bind to the somatostatin receptor (SSTR), molecular imaging plays an increasing and critical role in the diagnosis, staging, and treatment planning of these neoplasms. Dual-tracer imaging with 18F-FDG PET/CT and SSTR agents may play a significant role in treatment planning and predicting patient outcomes in the setting of high-grade or poorly differentiated neuroendocrine tumors.
Collapse
Affiliation(s)
- Wajahat Khatri
- Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 601 North Caroline Street, JHOC 3, Baltimore, MD 21287, USA
| | - Ergi Spiro
- Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 601 North Caroline Street, JHOC 3, Baltimore, MD 21287, USA
| | - Amanda Henderson
- Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 601 North Caroline Street, JHOC 3, Baltimore, MD 21287, USA
| | - Steven P Rowe
- Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 601 North Caroline Street, JHOC 3, Baltimore, MD 21287, USA
| | - Lilja B Solnes
- Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 601 North Caroline Street, JHOC 3, Baltimore, MD 21287, USA.
| |
Collapse
|
14
|
Urso L, Nieri A, Uccelli L, Castello A, Artioli P, Cittanti C, Marzola MC, Florimonte L, Castellani M, Bissoli S, Porto F, Boschi A, Evangelista L, Bartolomei M. Lutathera® Orphans: State of the Art and Future Application of Radioligand Therapy with 177Lu-DOTATATE. Pharmaceutics 2023; 15:pharmaceutics15041110. [PMID: 37111596 PMCID: PMC10142322 DOI: 10.3390/pharmaceutics15041110] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/28/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023] Open
Abstract
Lutathera® is the first EMA- and FDA-approved radiopharmaceutical for radioligand therapy (RLT). Currently, on the legacy of the NETTER1 trial, only adult patients with progressive unresectable somatostatin receptor (SSTR) positive gastroenteropancreatic (GEP) neuroendocrine neoplasms (NET) can be treated with Lutathera®. Conversely, patients with SSTR-positive disease arising from outside the gastroenteric region do not currently have access to Lutathera® treatment despite several papers in the literature reporting the effectiveness and safety of RLT in these settings. Moreover, patients with well-differentiated G3 GEP-NET are also still “Lutathera orphans”, and retreatment with RLT in patients with disease relapse is currently not approved. The aim of this critical review is to summarize current literature evidence assessing the role of Lutathera® outside the approved indications. Moreover, ongoing clinical trials evaluating new possible applications of Lutathera® will be considered and discussed to provide an updated picture of future investigations.
Collapse
Affiliation(s)
- Luca Urso
- Department of Translational Medicine, University of Ferrara, Via Aldo Moro 8, 44124 Ferrara, Italy; (L.U.); (C.C.); (F.P.)
- Department of Nuclear Medicine, PET/CT Centre, S. Maria della Misericordia Hospital, 45100 Rovigo, Italy;
| | - Alberto Nieri
- Nuclear Medicine Unit, Oncological Medical and Specialist Department, University Hospital of Ferrara, 44124 Cona, Italy; (A.N.); (M.B.)
| | - Licia Uccelli
- Department of Translational Medicine, University of Ferrara, Via Aldo Moro 8, 44124 Ferrara, Italy; (L.U.); (C.C.); (F.P.)
- Nuclear Medicine Unit, Oncological Medical and Specialist Department, University Hospital of Ferrara, 44124 Cona, Italy; (A.N.); (M.B.)
- Correspondence: ; Tel.: +39-053-232-6387
| | - Angelo Castello
- Nuclear Medicine Unit, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.C.); (L.F.); (M.C.)
| | - Paolo Artioli
- Nuclear Medicine Unit, AULSS1 Dolomiti, San Martino Hospital, 32100 Belluno, Italy; (P.A.); (S.B.)
| | - Corrado Cittanti
- Department of Translational Medicine, University of Ferrara, Via Aldo Moro 8, 44124 Ferrara, Italy; (L.U.); (C.C.); (F.P.)
- Nuclear Medicine Unit, Oncological Medical and Specialist Department, University Hospital of Ferrara, 44124 Cona, Italy; (A.N.); (M.B.)
| | - Maria Cristina Marzola
- Department of Nuclear Medicine, PET/CT Centre, S. Maria della Misericordia Hospital, 45100 Rovigo, Italy;
| | - Luigia Florimonte
- Nuclear Medicine Unit, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.C.); (L.F.); (M.C.)
| | - Massimo Castellani
- Nuclear Medicine Unit, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.C.); (L.F.); (M.C.)
| | - Sergio Bissoli
- Nuclear Medicine Unit, AULSS1 Dolomiti, San Martino Hospital, 32100 Belluno, Italy; (P.A.); (S.B.)
| | - Francesca Porto
- Department of Translational Medicine, University of Ferrara, Via Aldo Moro 8, 44124 Ferrara, Italy; (L.U.); (C.C.); (F.P.)
| | - Alessandra Boschi
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy;
| | - Laura Evangelista
- Department of Medicine DIMED, University of Padua, 35128 Padua, Italy;
| | - Mirco Bartolomei
- Nuclear Medicine Unit, Oncological Medical and Specialist Department, University Hospital of Ferrara, 44124 Cona, Italy; (A.N.); (M.B.)
| |
Collapse
|
15
|
Pellegrino F, Granata V, Fusco R, Grassi F, Tafuto S, Perrucci L, Tralli G, Scaglione M. Diagnostic Management of Gastroenteropancreatic Neuroendocrine Neoplasms: Technique Optimization and Tips and Tricks for Radiologists. Tomography 2023; 9:217-246. [PMID: 36828370 PMCID: PMC9958666 DOI: 10.3390/tomography9010018] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/22/2023] [Accepted: 01/23/2023] [Indexed: 01/31/2023] Open
Abstract
Gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs) comprise a heterogeneous group of neoplasms, which derive from cells of the diffuse neuroendocrine system that specializes in producing hormones and neuropeptides and arise in most cases sporadically and, to a lesser extent, in the context of complex genetic syndromes. Furthermore, they are primarily nonfunctioning, while, in the case of insulinomas, gastrinomas, glucagonomas, vipomas, and somatostatinomas, they produce hormones responsible for clinical syndromes. The GEP-NEN tumor grade and cell differentiation may result in different clinical behaviors and prognoses, with grade one (G1) and grade two (G2) neuroendocrine tumors showing a more favorable outcome than grade three (G3) NET and neuroendocrine carcinoma. Two critical issues should be considered in the NEN diagnostic workup: first, the need to identify the presence of the tumor, and, second, to define the primary site and evaluate regional and distant metastases. Indeed, the primary site, stage, grade, and function are prognostic factors that the radiologist should evaluate to guide prognosis and management. The correct diagnostic management of the patient includes a combination of morphological and functional evaluations. Concerning morphological evaluations, according to the consensus guidelines of the European Neuroendocrine Tumor Society (ENETS), computed tomography (CT) with a contrast medium is recommended. Contrast-enhanced magnetic resonance imaging (MRI), including diffusion-weighted imaging (DWI), is usually indicated for use to evaluate the liver, pancreas, brain, and bones. Ultrasonography (US) is often helpful in the initial diagnosis of liver metastases, and contrast-enhanced ultrasound (CEUS) can solve problems in characterizing the liver, as this tool can guide the biopsy of liver lesions. In addition, intraoperative ultrasound is an effective tool during surgical procedures. Positron emission tomography (PET-CT) with FDG for nonfunctioning lesions and somatostatin analogs for functional lesions are very useful for identifying and evaluating metabolic receptors. The detection of heterogeneity in somatostatin receptor (SSTR) expression is also crucial for treatment decision making. In this narrative review, we have described the role of morphological and functional imaging tools in the assessment of GEP-NENs according to current major guidelines.
Collapse
Affiliation(s)
| | - Vincenza Granata
- Division of Radiology, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| | - Roberta Fusco
- Medical Oncology Division, Igea SpA, 80013 Naples, Italy
| | - Francesca Grassi
- Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, Via della Signora 2, 20122 Milan, Italy
- Division of Radiology, Università degli Studi della Campania Luigi Vanvitelli, 80127 Naples, Italy
| | - Salvatore Tafuto
- S.C. Sarcomi e Tumori Rari, Istituto Nazionale Tumori, IRCCS, Fondazione “G. Pascale”, 80131 Naples, Italy
| | - Luca Perrucci
- Ferrara Department of Interventional and Diagnostic Radiology, Ospedale di Lagosanto, Azienda AUSL, 44023 Ferrara, Italy
| | - Giulia Tralli
- Department of Radiology, Ospedale Santa Maria della Misericordia, 45100 Rovigo, Italy
| | - Mariano Scaglione
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, 07100 Sassari, Italy
| |
Collapse
|
16
|
Veenstra EB, Noordzij W, Erba PA. Neuroendocrine Neoplasm Imaging and Image-Guided Therapies. MULTIMODALITY IMAGING AND INTERVENTION IN ONCOLOGY 2023:419-439. [DOI: 10.1007/978-3-031-28524-0_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
17
|
Rectal neuroendocrine neoplasms: what the radiologists should know. ABDOMINAL RADIOLOGY (NEW YORK) 2022; 47:4016-4031. [PMID: 35288791 DOI: 10.1007/s00261-022-03474-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/18/2022] [Accepted: 02/21/2022] [Indexed: 02/06/2023]
Abstract
Neuroendocrine neoplasms of the rectum (R-NENs) are rare; however, their incidence has increased almost threefold in the last few decades. Imaging of R-NENs includes two primary categories: anatomic/morphologic imaging comprised of endoscopic ultrasound (EUS), computed tomography (CT), magnetic resonance imaging (MRI), and functional/molecular imaging comprising of planar scintigraphy, single-photon emission computed tomography (SPECT), and positron emission tomography (PET). The management depends on stage, dimension, atypical features, histological grade, and lymphovascular invasion (LVI). Low-risk local R-NENs can be resected endoscopically, and high-risk or locally advanced neoplasms can be treated with radical surgery and lymphadenectomy and/or chemoradiation. The review article focuses on imaging illustrations and discusses applications of different imaging modalities in diagnosing and managing R-NENs.
Collapse
|
18
|
Jiang Y, Liu Q, Wang G, Sui H, Wang R, Wang J, Zhu Z. A prospective head-to-head comparison of 68 Ga-NOTA-3P-TATE-RGD and 68 Ga-DOTATATE in patients with gastroenteropancreatic neuroendocrine tumours. Eur J Nucl Med Mol Imaging 2022; 49:4218-4227. [PMID: 35657429 DOI: 10.1007/s00259-022-05852-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/24/2022] [Indexed: 11/30/2022]
Abstract
PURPOSE The aim of this study was to compare 68 Ga-NOTA-3P-TATE-RGD, a dual somatostatin receptor 2- and integrin αVβ3-targeting tracer, to 68 Ga-DOTATATE in a single group of patients with gastroenteropancreatic (GEP)-neuroendocrine tumours (NETs). METHODS Thirty-five patients with histologically confirmed GEP-NETs (5 grade 1, 28 grade 2, and 2 grade 3 tumours) were prospectively enrolled with informed consent. The primary tumour mainly originated from the pancreas and rectum. All patients were scanned with both 68 Ga-NOTA-3P-TATE-RGD PET/CT and 68 Ga-DOTATATE PET/CT within a week and compared on a head-to-head basis. Sixteen patients also had conventional 18F-FDG PET/CT. Images were evaluated semi-quantitatively using maximum standardized uptake values (SUVmax) of tumour and tumour-to-background ratio. RESULTS All patients had at least one positive lesion on each of the two scans. A total of 1190 and 1106 lesions were detected on 68 Ga-NOTA-3P-TATE-RGD images and 68 Ga-DOTATATE images, respectively (P = 0.152). 68 Ga-NOTA-3P-TATE-RGD PET/CT revealed significantly more lesions in the liver than 68 Ga-DOTATATE PET/CT (634 vs. 532, P = 0.021). Both tracers produced comparable results for detecting primary tumours (20 vs. 20, P = 1.000), lymph node metastases (101 vs. 102, P = 0.655), and bone metastases (381 vs. 398, P = 0.244). The tumour SUVmax in 12 patients was significantly higher for 68 Ga-NOTA-3P-TATE-RGD than for 68 Ga-DOTATATE (27.2 ± 13.6 vs. 19.5 ± 10.0, P < 0.001); among them, 9 had 18F-FDG PET/CT and all were found to be FDG-positive. The remaining 23 patients had significantly higher 68 Ga-DOTATATE uptake than 68 Ga-NOTA-3P-TATE-RGD uptake (22.3 ± 16.4 vs. 11.9 ± 7.5, P < 0.001); among them, 7 had 18F-FDG PET/CT and 6 were FDG-negative. Generally, 68 Ga-DOTATATE demonstrated higher tumour SUVmax than 68 Ga-NOTA-3P-TATE-RGD (20.8 ± 16.0 vs. 14.2 ± 8.9, P < 0.001), including primary tumours, liver lesions, lymph node lesions, and bone lesions. However, the tumour-to-background ratio of liver lesions was significantly higher when using 68 Ga-NOTA-3P-TATE-RGD compared with that when using 68 Ga-DOTATATE (8.4 ± 5.5 vs. 4.7 ± 3.7, P < 0.001). CONCLUSION 68 Ga-NOTA-3P-TATE-RGD performed better than 68 Ga-DOTATATE in detection of liver metastases with a higher tumour-to-background ratio. Moreover, 68 Ga-NOTA-3P-TATE-RGD tended to demonstrate higher uptake over 68 Ga-DOTATATE in FDG-avid NETs. TRIAL REGISTRATION Dual SSTR2 and Integrin αvβ3 Targeting PET/CT Imaging (NCT02817945, registered 5 November 2018). URL OF REGISTRY: https://clinicaltrials.gov/ct2/show/NCT02817945.
Collapse
Affiliation(s)
- Yuanyuan Jiang
- Department of Nuclear Medicine, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- State Key Laboratory of Complex Severe and Rare Diseases, Beijing, 100730, China
| | - Qingxing Liu
- Department of Nuclear Medicine, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- State Key Laboratory of Complex Severe and Rare Diseases, Beijing, 100730, China
| | - Guochang Wang
- Department of Nuclear Medicine, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- State Key Laboratory of Complex Severe and Rare Diseases, Beijing, 100730, China
| | - Huimin Sui
- Department of Nuclear Medicine, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- State Key Laboratory of Complex Severe and Rare Diseases, Beijing, 100730, China
| | - Rongxi Wang
- Department of Nuclear Medicine, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- State Key Laboratory of Complex Severe and Rare Diseases, Beijing, 100730, China
| | - Jiarou Wang
- Department of Nuclear Medicine, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- State Key Laboratory of Complex Severe and Rare Diseases, Beijing, 100730, China
| | - Zhaohui Zhu
- Department of Nuclear Medicine, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
- State Key Laboratory of Complex Severe and Rare Diseases, Beijing, 100730, China.
| |
Collapse
|
19
|
Glucose Metabolism Modification Induced by Radioligand Therapy with [ 177Lu]Lu/[ 90Y]Y-DOTATOC in Advanced Neuroendocrine Neoplasms: A Prospective Pilot Study within FENET-2016 Trial. Pharmaceutics 2022; 14:pharmaceutics14102009. [PMID: 36297443 PMCID: PMC9612170 DOI: 10.3390/pharmaceutics14102009] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 12/13/2022] Open
Abstract
[18F]F-FDG (FDG) PET is emerging as a relevant diagnostic and prognostic tool in neuroendocrine neoplasms (NENs), as a simultaneous decrease in [68Ga]Ga-DOTA peptides and increase in FDG uptake (the “flip-flop” phenomenon) occurs during the natural history of these tumors. The aim of this study was to evaluate the variations on FDG PET in NEN patients treated with two different schemes of radioligand therapy (RLT) and to correlate them with clinical−pathologic variables. A prospective evaluation of 108 lesions in 56 patients (33 males and 23 females; median age, 64.5 years) affected by NENs of various primary origins (28 pancreatic, 13 gastrointestinal, 9 bronchial, 6 unknown primary (CUP-NENs) and 1 pheochromocytoma) and grades (median Ki-67 = 9%) was performed. The patients were treated with RLT within the phase II clinical trial FENET-2016 (CTID: NCT04790708). RLT was offered for 32 patients with the MONO scheme (five cycles of [177Lu]Lu-DOTATOC) and for 24 with the DUO scheme (three cycles of [177Lu]Lu-DOTATOC alternated with two cycles of [90Y]Y-DOTATOC). Variations in terms of the ΔSUVmax of a maximum of three target lesions per patient (58 for MONO and 50 for DUO RLT) were assessed between baseline and 3 months post-RLT FDG PET. In patients with negative baseline FDG PET, the three most relevant lesions on [68Ga]Ga-DOTA-peptide PET were assessed and matched on post-RLT FDG PET, to check for any possible changes in FDG avidity. Thirty-five patients (62.5%) had at least one pathological FDG uptake at the baseline scans, but the number was reduced to 29 (52%) after RLT. In the patients treated with DUO-scheme RLT, 20 out of 50 lesions were FDG positive before therapy, whereas only 14 were confirmed after RLT (p = 0.03). Moreover, none of the 30 FDG-negative lesions showed an increased FDG uptake after RLT. The lesions of patients with pancreatic and CUP-NENs treated with the DUO scheme demonstrated a significant reduction in ΔSUVmax in comparison to those treated with MONO RLT (p = 0.03 and p = 0.04, respectively). Moreover, we found a mild positive correlation between the grading and ΔSUVmax in patients treated with the MONO scheme (r = 0.39, p < 0.02), while no evidence was detected for patients treated with the DUO scheme. Our results suggest that RLT, mostly with the DUO scheme, could be effective in changing NEN lesions’ glycometabolism, in particular, in patients affected by pancreatic and CUP-NENs, regardless of their Ki-67 index. Probably, associating [90Y]Y-labelled peptides, which have high energy emission and a crossfire effect, and [177Lu]Lu ones, characterized by a longer half-life and a safer profile for organs at risk, might represent a valid option in FDG-positive NENs addressed to RLT. Further studies are needed to validate our preliminary findings. In our opinion, FDG PET/CT should represent a potent tool for fully assessing a patient’s disease characteristics, both before and after RLT.
Collapse
|
20
|
Refardt J, Hofland J, Wild D, Christ E. Molecular Imaging of Neuroendocrine Neoplasms. J Clin Endocrinol Metab 2022; 107:e2662-e2670. [PMID: 35380158 DOI: 10.1210/clinem/dgac207] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Indexed: 12/17/2022]
Abstract
The key for molecular imaging is the use of a radiotracer with a radioactive and a functional component. While the functional component targets a specific feature of the tumor, the radioactive component makes the target visible. Neuroendocrine neoplasms (NEN) are a diverse group of rare tumors that arise from neuroendocrine cells found mainly in the gastroenteropancreatic system, lung, thyroid, and adrenal glands. They are characterized by the expression of specific hormone receptors on the tumor cell surface, which makes them ideal targets for radiolabeled peptides. The most commonly expressed hormone receptors on NEN cells are the somatostatin receptors. They can be targeted for molecular imaging with various radiolabeled somatostatin analogs, but also with somatostatin antagonists, which have shown improved imaging quality. 18F-DOPA imaging has become a second-line imaging modality in NENs, with the exception of the evaluation of advanced medullary thyroid carcinoma. Alternatives for NENs with insufficient somatostatin receptor expression due to poor differentiation involve targeting glucose metabolism, which can also be used for prognosis. For the localization of the often-small insulinoma, glucagon-like peptide-1 (GLP-1) receptor imaging has become the new standard. Other alternatives involve metaiodobenzylguanidine and the molecular target C-X-C motif chemokine receptor-4. In addition, new radiopeptides targeting the fibroblast activation protein, the glucose-dependent insulinotropic polypeptide receptor and cholecystokinin-2 receptors have been identified in NENs and await further evaluation. This mini-review aims to provide an overview of the major molecular imaging modalities currently used in the field of NENs, and also to provide an outlook on future developments.
Collapse
Affiliation(s)
- Julie Refardt
- Department of Internal Medicine, Section of Endocrinology, ENETS Center of Excellence, Erasmus Medical Center, Rotterdam, the Netherlands
- ENETS Center of Excellence for Neuroendocrine and Endocrine Tumors, University Hospital Basel, Basel, Switzerland
- Department of Endocrinology, Diabetology and Metabolism, University Hospital Basel, Basel, Switzerland
| | - Johannes Hofland
- Department of Internal Medicine, Section of Endocrinology, ENETS Center of Excellence, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Damian Wild
- ENETS Center of Excellence for Neuroendocrine and Endocrine Tumors, University Hospital Basel, Basel, Switzerland
- Division of Nuclear Medicine, University Hospital Basel, Basel, Switzerland
| | - Emanuel Christ
- ENETS Center of Excellence for Neuroendocrine and Endocrine Tumors, University Hospital Basel, Basel, Switzerland
- Department of Endocrinology, Diabetology and Metabolism, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
21
|
Laffi A, Spada F, Bagnardi V, Frassoni S, Pisa E, Rubino M, Barberis M, Fazio N. Gastroenteropancreatic grade 3 neuroendocrine tumors: a single entity or a heterogeneous group? A retrospective analysis. J Endocrinol Invest 2022; 45:317-325. [PMID: 34282554 DOI: 10.1007/s40618-021-01642-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 07/14/2021] [Indexed: 10/20/2022]
Abstract
PURPOSE Grade 3 neuroendocrine tumor (NET G3) is a novel pathologic category within gastro-entero-pancreatic (GEP) neuroendocrine neoplasms (NENs) but its clinical behavior and therapeutic management still remain challenging. Prognostic and predictive factors aiding NET G3 management are needed. PATIENTS AND METHODS We performed a retrospective analysis from 2015 to 2020 of all patients with > 20% Ki-67, well-differentiated NETs evaluated within our NEN-dedicated multidisciplinary team. We divided the sample according the timing of NET G3 diagnosis, the radiotracers distribution and Ki-67. We analyzed the correlation between these NET G3 features and clinical outcomes. RESULTS Among 3238 multidisciplinary discussion reports, we selected 55 patients, 48 from GEP and 7 from an occult GEP origin. In 45 patients, NET G3 diagnosis occurred at the beginning of clinical history (upfront-NET G3), whereas in 10, during the NET G1-G2 clinical history (late-NET G3). Patients with ≤ 30% (34/55) vs. > 30% Ki-67 (21/55) had a better overall survival (OS) (p = 0.042); patients with a homogeneous vs. inhomogeneous/negative 68Gallium(68Ga)-DOTA-Peptide Positron Emission Tomography (PET)/computed tomography (CT) showed a trend to a better OS, and a significant better progression-free survival (PFS) (p = 0.033). A better OS was observed for negative/inhomogeneous vs. homogeneous 18-fluorodeoxyglucose (18FDG)-PET/CT (p = 0.027). A trend to a better OS was reported in late- vs. upfront-NET G3, while the latter showed a significantly better response rate (RR) (p = 0.048). CONCLUSION Our findings suggested that Ki-67 cutoff, functional imaging and the timing to NET G3 diagnosis may help clinicians in more accurate selection of NET G3 management. Prospective studies are needed.
Collapse
Affiliation(s)
- A Laffi
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology (IEO), IRCCS, Via Ripamonti 435, 20141, Milan, Italy.
| | - F Spada
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology (IEO), IRCCS, Via Ripamonti 435, 20141, Milan, Italy
| | - V Bagnardi
- Department of Statistics and Quantitative Methods, University of Milano-Bicocca, Piazza dell'Ateneo Nuovo 1, 20126, Milan, Italy
| | - S Frassoni
- Department of Statistics and Quantitative Methods, University of Milano-Bicocca, Piazza dell'Ateneo Nuovo 1, 20126, Milan, Italy
| | - E Pisa
- Division of Pathology and Laboratory Medicine, IEO, European Institute of Oncology IRCCS, Via Ripamonti 435, 20141, Milan, Italy
| | - M Rubino
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology (IEO), IRCCS, Via Ripamonti 435, 20141, Milan, Italy
| | - M Barberis
- Division of Pathology and Laboratory Medicine, IEO, European Institute of Oncology IRCCS, Via Ripamonti 435, 20141, Milan, Italy
| | - N Fazio
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology (IEO), IRCCS, Via Ripamonti 435, 20141, Milan, Italy
| |
Collapse
|
22
|
Kaewput C, Vinjamuri S. Role of Combined 68Ga DOTA-Peptides and 18F FDG PET/CT in the Evaluation of Gastroenteropancreatic Neuroendocrine Neoplasms. Diagnostics (Basel) 2022; 12:diagnostics12020280. [PMID: 35204371 PMCID: PMC8871217 DOI: 10.3390/diagnostics12020280] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 12/12/2022] Open
Abstract
This review article summarizes the role of combined 68Ga DOTA-peptides and 18F-fluorodeoxyglucose (FDG) positron emission tomography/computed tomography (PET/CT) in the evaluation of gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs). Patients with GEP-NENs can initially present themselves to a gastroenterologist or endocrinologist rather than cancer specialist; hence, it is vital for a wider group of clinicians to be familiar with the range of tests available for the evaluation of these patients. The role of PET scanning by using 68Ga DOTA-peptides has a high sensitivity in the diagnosis of GEP-NENs and to guide patient selection for treatment with somatostatin analogues (SSA) and/or peptide receptor radionuclide therapy (PRRT). The loss of somatostatin receptor (SSTR) expression was found to be associated with an increased glucose metabolism in cells. However, the routine use of SSTR targeted radiotracers in combination with 18F-FDG to evaluate glucose utilization in GEP-NENs is still debatable. In our opinion, in patients with NENs, 18F-FDG PET should be performed in the case of a negative or slightly positive 68Ga DOTA-peptides PET scan for assessing the dedifferentiation status, to guide correct therapeutic strategy and to evaluate the prognosis. The approach of combined receptor and metabolic imaging can improve diagnostic accuracy, especially considering the heterogeneity of these lesions. Therefore, 68Ga DOTA-peptides and 18F-FDG PET should be considered complementary in patients with GEP-NENs.
Collapse
Affiliation(s)
- Chalermrat Kaewput
- Division of Nuclear Medicine, Department of Radiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
- Correspondence: ; Tel.: +66-2419-6220
| | - Sobhan Vinjamuri
- Department of Nuclear Medicine, Royal Liverpool University Hospital, Liverpool L7 8XP, UK;
| |
Collapse
|
23
|
Bodei L, Jayaprakasam VS, Kidd M, Gilardi L, Volterrani D, Paganelli G, Grana CM, Modlin IM. Diagnostic Applications of Nuclear Medicine: Neuroendocrine Tumors. NUCLEAR ONCOLOGY 2022:933-974. [DOI: 10.1007/978-3-031-05494-5_18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
24
|
Kabasakal L, Demirci E, Selçuk NA. Radionuclide Therapy in Neuroendocrine Tumors. RADIONUCLIDE THERAPY 2022:173-186. [DOI: 10.1007/978-3-030-97220-2_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
25
|
Bodei L, Kidd M, Modlin IM. Clinical and scientific considerations of genomics and metabolomics in radionuclide therapy. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00198-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
26
|
Fanti S, Ambrosini V. PET Imaging in Neuro-Endocrine Neoplasms (NEN). Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00034-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
27
|
Tabacchi E, Nanni C, Bossert I, Maffione AM, Fanti S. Diagnostic Applications of Nuclear Medicine: Pancreatic Cancer. NUCLEAR ONCOLOGY 2022:891-917. [DOI: 10.1007/978-3-031-05494-5_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
28
|
Peptide Receptor Radionuclide Therapy Targeting the Somatostatin Receptor: Basic Principles, Clinical Applications and Optimization Strategies. Cancers (Basel) 2021; 14:cancers14010129. [PMID: 35008293 PMCID: PMC8749814 DOI: 10.3390/cancers14010129] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/13/2021] [Accepted: 12/22/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Peptide receptor radionuclide therapy (PRRT) is a systemic treatment consisting of the administration of a tumor-targeting radiopharmaceutical into the circulation of a patient. The radiopharmaceutical will bind to a specific peptide receptor leading to tumor-specific binding and retention. This will subsequently cause lethal DNA damage to the tumor cell. The only target that is currently used in widespread clinical practice is the somatostatin receptor, which is overexpressed on a range of tumor cells, including neuroendocrine tumors and neural-crest derived tumors. Academia played an important role in the development of PRRT, which has led to heterogeneous literature over the last two decades, as no standard radiopharmaceutical or regimen has been available for a long time. This review focuses on the basic principles and clinical applications of PRRT, and discusses several PRRT-optimization strategies. Abstract Peptide receptor radionuclide therapy (PRRT) consists of the administration of a tumor-targeting radiopharmaceutical into the circulation of a patient. The radiopharmaceutical will bind to a specific peptide receptor leading to tumor-specific binding and retention. The only target that is currently used in clinical practice is the somatostatin receptor (SSTR), which is overexpressed on a range of tumor cells, including neuroendocrine tumors and neural-crest derived tumors. Academia played an important role in the development of PRRT, which has led to heterogeneous literature over the last two decades, as no standard radiopharmaceutical or regimen has been available for a long time. This review provides a summary of the treatment efficacy (e.g., response rates and symptom-relief), impact on patient outcome and toxicity profile of PRRT performed with different generations of SSTR-targeting radiopharmaceuticals, including the landmark randomized-controlled trial NETTER-1. In addition, multiple optimization strategies for PRRT are discussed, i.e., the dose–effect concept, dosimetry, combination therapies (i.e., tandem/duo PRRT, chemoPRRT, targeted molecular therapy, somatostatin analogues and radiosensitizers), new radiopharmaceuticals (i.e., SSTR-antagonists, Evans-blue containing vector molecules and alpha-emitters), administration route (intra-arterial versus intravenous) and response prediction via molecular testing or imaging. The evolution and continuous refinement of PRRT resulted in many lessons for the future development of radionuclide therapy aimed at other targets and tumor types.
Collapse
|
29
|
Marini I, Sansovini M, Bongiovanni A, Nicolini S, Grassi I, Ranallo N, Monti M, DI Iorio V, Germanò L, Caroli P, Sarnelli A, Paganelli G, Severi S. Theragnostic in neuroendocrine tumors. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF... 2021; 65:342-352. [PMID: 34881852 DOI: 10.23736/s1824-4785.21.03426-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
In the last few decades, the incidence and prevalence of neuroendocrine tumors has been increasing. The theragnostic approach, that allows the diagnosis and treatment of different neoplasms with the same ligand, is a typical nuclear medicine tool. Applied for years, is also pivotal in neuroendocrine tumors (NETs) where it has improved the diagnostic accuracy and the therapeutic efficacy with impact on patient's survival. Theragnostic also allows the identification of important prognostic factors such as tumor location and burden, presence of liver metastases and intensity of somatostatin receptors (SSTR) expression to consider in new and possibly combined studies to ameliorate patient's outcome. Moreover, the possibility to evaluate receptor expression even in non-NET malignancies has de facto widened the possible indications for PRRT. We believe that this innovative therapeutic approach will be implemented in next years by radiomics and biological tumors characterization to better address PRRT applications.
Collapse
Affiliation(s)
- Irene Marini
- Unit of Nuclear Medicine, IRCCS Istituto Romagnolo per lo Studio dei Tumori - IRST Dino Amadori, Meldola, Forlì-Cesena, Italy
| | - Maddalena Sansovini
- Unit of Nuclear Medicine, IRCCS Istituto Romagnolo per lo Studio dei Tumori - IRST Dino Amadori, Meldola, Forlì-Cesena, Italy
| | - Alberto Bongiovanni
- Osteoncology and Rare Tumors Center - CDO-TR, IRCCS Istituto Romagnolo per lo Studio dei Tumori - IRST Dino Amadori, Meldola, Forlì-Cesena, Italy
| | - Silvia Nicolini
- Unit of Nuclear Medicine, IRCCS Istituto Romagnolo per lo Studio dei Tumori - IRST Dino Amadori, Meldola, Forlì-Cesena, Italy
| | - Ilaria Grassi
- Unit of Nuclear Medicine, IRCCS Istituto Romagnolo per lo Studio dei Tumori - IRST Dino Amadori, Meldola, Forlì-Cesena, Italy
| | - Nicoletta Ranallo
- Osteoncology and Rare Tumors Center - CDO-TR, IRCCS Istituto Romagnolo per lo Studio dei Tumori - IRST Dino Amadori, Meldola, Forlì-Cesena, Italy
| | - Manuela Monti
- Unit of Biostatistics and Clinical Trials, IRCCS Istituto Romagnolo per lo Studio dei Tumori - IRST Dino Amadori, Meldola, Forlì-Cesena, Italy
| | - Valentina DI Iorio
- Unit of Oncological Pharmacy, IRCCS Istituto Romagnolo per lo Studio dei Tumori - IRST Dino Amadori, Meldola, Forlì-Cesena, Italy
| | - Luca Germanò
- Unit of Nuclear Medicine, IRCCS Istituto Romagnolo per lo Studio dei Tumori - IRST Dino Amadori, Meldola, Forlì-Cesena, Italy
| | - Paola Caroli
- Unit of Nuclear Medicine, IRCCS Istituto Romagnolo per lo Studio dei Tumori - IRST Dino Amadori, Meldola, Forlì-Cesena, Italy
| | - Anna Sarnelli
- Unit of Medical Physics, IRCCS Istituto Romagnolo per lo Studio dei Tumori - IRST Dino Amadori, Meldola, Forlì-Cesena, Italy
| | - Giovanni Paganelli
- Unit of Nuclear Medicine, IRCCS Istituto Romagnolo per lo Studio dei Tumori - IRST Dino Amadori, Meldola, Forlì-Cesena, Italy
| | - Stefano Severi
- Unit of Nuclear Medicine, IRCCS Istituto Romagnolo per lo Studio dei Tumori - IRST Dino Amadori, Meldola, Forlì-Cesena, Italy -
| |
Collapse
|
30
|
Combined use of peptide receptor radionuclide therapy and metronomic chemotherapy in neuroendocrine tumors: a possible choice driven by nuclear medicine molecular imaging. Eur J Nucl Med Mol Imaging 2021; 48:3041-3042. [PMID: 33851244 DOI: 10.1007/s00259-021-05330-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
31
|
Pirasteh A, Lovrec P, Bodei L. Imaging of neuroendocrine tumors: A pictorial review of the clinical value of different imaging modalities. Rev Endocr Metab Disord 2021; 22:539-552. [PMID: 33783695 DOI: 10.1007/s11154-021-09631-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/25/2021] [Indexed: 02/07/2023]
Abstract
Neuroendocrine tumors (NETs) are multifaceted tumors occurring in a variety of organs and often present as metastatic at the time of diagnosis. Accurate staging is the most significant factor in therapy planning, but it remains a challenge. Imaging is established as the cornerstone for disease detection/diagnosis, staging, and follow up. To accurately assess and monitor tumor burden in patients with NETs, various imaging techniques have been developed and optimized. Current recommendations for the imaging of patients with NETs include a combination of both morphologic (or anatomic) and molecular imaging, but a final choice can be puzzling for clinicians. Recognizing that there is no uniform sequence consensus on the "best" imaging test, and the heterogeneity of technologic availability at different centers, we hope to provide a pictorial review of the different imaging techniques and their role and utility in management of patients with NETs, aimed to provide a practical guide for all clinicians.
Collapse
Affiliation(s)
- Ali Pirasteh
- Department of Radiology, University of Wisconsin-Madison, Madison, WI, United States.
| | - Petra Lovrec
- Department of Radiology, University of Wisconsin-Madison, Madison, WI, United States
| | - Lisa Bodei
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| |
Collapse
|
32
|
Albertelli M, Dotto A, Di Dato C, Malandrino P, Modica R, Versari A, Colao A, Ferone D, Faggiano A. PRRT: identikit of the perfect patient. Rev Endocr Metab Disord 2021; 22:563-579. [PMID: 32978685 PMCID: PMC8346456 DOI: 10.1007/s11154-020-09581-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Peptide receptor radionuclide therapy (PRRT) has been strengthened since the publication of NETTER-1. Nevertheless, the correct positioning in the therapeutic algorithm is debated, and no optimal sequence has yet been standardized. Possible criteria to predict the response to PRRT in neuroendocrine tumors (NET) have been proposed. The aim of this review is to define the perfect identity of the eligible patient who can mostly benefit from this therapy. Possible predictive criteria which have been analysed were: primary tumor site, grading, tumor burden, FDG PET and 68Ga-PET uptake. Primary tumor site and 68Ga-PET uptake do not play a pivotal role in predicting the response, while tumor burden, FDG PET uptake and grading seem to represent predictive/prognostic factors for response to PRRT. The heterogeneity in trial designs, patient populations, type of radionuclides, previous therapies and measurement of outcomes, inevitably limits the strength of our conclusions, therefore care must be taken in applying these results to clinical practice. In conclusion, the perfect patient, selected by 68Ga-PET uptake, will likely have a relatively limited liver tumor burden, a ki67 index <20% and will respond to PRRT irrespective to primary tumor. Nevertheless, we have mostly prognostic than predictive factors to predict the efficacy of PRRT in individual patients, while a promising tool could be the NETest. However, to date, the identikit of the perfect patient for PRRT is a puzzle without some pieces and still we cannot disregard a multidisciplinary discussion of the individual case to select the patients who will mostly benefit from PRRT.
Collapse
Affiliation(s)
- M Albertelli
- Endocrinology Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Endocrinology Unit, Department of Internal Medicine and Medical Specialties (DiMI) and Center of Excellence for Biomedical Research, University of Genova, Genova, Italy
| | - A Dotto
- Endocrinology Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Endocrinology Unit, Department of Internal Medicine and Medical Specialties (DiMI) and Center of Excellence for Biomedical Research, University of Genova, Genova, Italy
| | - C Di Dato
- Endocrinology, Department of Experimental Medicine, "Sapienza", University of Rome, Rome, Italy
| | - P Malandrino
- Endocrinology, Department of Clinical and Experimental Medicine, Garibaldi-Nesima Medical Center, University of Catania, Catania, Italy
| | - R Modica
- Endocrinology, Department of Clinical Medicine and Surgery, "Federico II" University of Napoli, Napoli, Italy
| | - A Versari
- Nuclear Medicine, Azienda Ospedaliera Santa Maria Nuova-IRCCS Reggio Emilia, Reggio Emilia, Italy
| | - A Colao
- Endocrinology, Department of Clinical Medicine and Surgery, "Federico II" University of Napoli, Napoli, Italy
| | - D Ferone
- Endocrinology Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Endocrinology Unit, Department of Internal Medicine and Medical Specialties (DiMI) and Center of Excellence for Biomedical Research, University of Genova, Genova, Italy
| | - A Faggiano
- Endocrinology, Department of Experimental Medicine, "Sapienza", University of Rome, Rome, Italy.
- Depart. of Experimental Medicine, Division of Medical Physiopathology Sapienza University of Rome Viale del Policlinico 155, 00161, Rome, Italy.
| | | |
Collapse
|
33
|
Rao YY, Zhang HJ, Wang XJ, Li MF. Primary hepatic neuroendocrine tumor — 18F-fluorodeoxyglucose positron emission tomography/computed tomography findings: A case report. World J Clin Cases 2021; 9:6450-6456. [PMID: 34435011 PMCID: PMC8362580 DOI: 10.12998/wjcc.v9.i22.6450] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 05/11/2021] [Accepted: 06/01/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Primary hepatic neuroendocrine tumors (PHNETs) are rare hepatic tumors. Their diagnosis, which is based on radiological findings, is difficult.
CASE SUMMARY We present a case of PHNET in a 79-year-old man with no clinical symptoms. Computed tomography (CT) and 2-Deoxy-2-[fluorine-18] fluorodeoxyglucose positron emission tomography/CT (18F-FDG PET/CT) were performed for further evaluation. A hypoattenuating mass with rim-like enhancement in segment 6 of the liver was detected on contrast-enhanced CT imaging. Increased uptake was also observed on 18F-FDG PET/CT. Histopathological and immunohistochemical examinations, which revealed a grade 2 neuroendocrine tumor (NET), confirmed the diagnosis.
CONCLUSION Diagnosing PHNET is challenging, and must be distinguished from other liver tumors. Metastatic NETs should be excluded.
Collapse
Affiliation(s)
- Yan-Ying Rao
- Department of Radiology, Fujian Cancer Hospital and Fujian Medical University Cancer Hospital, Fuzhou 350000, Fujian Province, China
| | - He-Jun Zhang
- Department of Pathology, Fujian Cancer Hospital and Fujian Medical University Cancer Hospital, Fuzhou 350000, Fujian Province, China
| | - Xiao-Jiang Wang
- Department of Pathology, Fujian Cancer Hospital and Fujian Medical University Cancer Hospital, Fuzhou 350000, Fujian Province, China
| | - Min-Feng Li
- Department of Radiology, Fujian Cancer Hospital and Fujian Medical University Cancer Hospital, Fuzhou 350000, Fujian Province, China
| |
Collapse
|
34
|
Peptide receptor radionuclide therapy for GEP-NET: consolidated knowledge and innovative applications. Clin Transl Imaging 2021. [DOI: 10.1007/s40336-021-00443-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
35
|
Roll W, Weckesser M, Seifert R, Bodei L, Rahbar K. Imaging and liquid biopsy in the prediction and evaluation of response to PRRT in neuroendocrine tumors: implications for patient management. Eur J Nucl Med Mol Imaging 2021; 48:4016-4027. [PMID: 33903926 PMCID: PMC8484222 DOI: 10.1007/s00259-021-05359-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 04/05/2021] [Indexed: 12/16/2022]
Abstract
Purpose The aim of this narrative review is to give an overview on current and emerging imaging methods and liquid biopsy for prediction and evaluation of response to PRRT. Current limitations and new perspectives, including artificial intelligence, are discussed. Methods A literature review of PubMed/Medline was performed with representative keywords. The search included articles published online through August 31, 2020. All searches were restricted to English language manuscripts. Results Peptide radio receptor therapy (PRRT) is a prospectively evaluated and approved therapy option in neuroendocrine tumors (NETs). Different ligands targeting the somatostatin receptor (SSTR) are used as theranostic pairs for imaging NET and for PRRT. Response assessment in prospective trials often relies on the morphological RECIST 1.1 criteria, based on lesion size in CT or MRI. The role of SSTR-PET and quantitative uptake parameters and volumetric data is still not defined. Monoanalyte tumor marker chromogranin A has a limited value for response assessment after PRRT. New emerging liquid biopsy techniques are offering prediction of response to PRRT and prognostic value. Conclusions New response criteria for NET patients undergoing PRRT will comprise multiparametric hybrid imaging and blood-based multianalyte markers. This represents tumor biology and heterogeneity.
Collapse
Affiliation(s)
- Wolfgang Roll
- Department of Nuclear Medicine, University Hospital Muenster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany.,West German Cancer Center, Muenster and Essen, Essen, Germany
| | - Matthias Weckesser
- Department of Nuclear Medicine, University Hospital Muenster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany.,West German Cancer Center, Muenster and Essen, Essen, Germany
| | - Robert Seifert
- Department of Nuclear Medicine, University Hospital Muenster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany.,West German Cancer Center, Muenster and Essen, Essen, Germany.,Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
| | - Lisa Bodei
- Department of Nuclear Medicine, Radiology, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Kambiz Rahbar
- Department of Nuclear Medicine, University Hospital Muenster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany. .,West German Cancer Center, Muenster and Essen, Essen, Germany.
| |
Collapse
|
36
|
Duclos V, Iep A, Gomez L, Goldfarb L, Besson FL. PET Molecular Imaging: A Holistic Review of Current Practice and Emerging Perspectives for Diagnosis, Therapeutic Evaluation and Prognosis in Clinical Oncology. Int J Mol Sci 2021; 22:4159. [PMID: 33923839 PMCID: PMC8073681 DOI: 10.3390/ijms22084159] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 04/14/2021] [Accepted: 04/15/2021] [Indexed: 02/06/2023] Open
Abstract
PET/CT molecular imaging has been imposed in clinical oncological practice over the past 20 years, driven by its two well-grounded foundations: quantification and radiolabeled molecular probe vectorization. From basic visual interpretation to more sophisticated full kinetic modeling, PET technology provides a unique opportunity to characterize various biological processes with different levels of analysis. In clinical practice, many efforts have been made during the last two decades to standardize image analyses at the international level, but advanced metrics are still under use in practice. In parallel, the integration of PET imaging with radionuclide therapy, also known as radiolabeled theranostics, has paved the way towards highly sensitive radionuclide-based precision medicine, with major breakthroughs emerging in neuroendocrine tumors and prostate cancer. PET imaging of tumor immunity and beyond is also emerging, emphasizing the unique capabilities of PET molecular imaging to constantly adapt to emerging oncological challenges. However, these new horizons face the growing complexity of multidimensional data. In the era of precision medicine, statistical and computer sciences are currently revolutionizing image-based decision making, paving the way for more holistic cancer molecular imaging analyses at the whole-body level.
Collapse
Affiliation(s)
- Valentin Duclos
- Department of Biophysics and Nuclear Medicine-Molecular Imaging, Hôpitaux Universitaires Paris Saclay, Assistance Publique-Hôpitaux de Paris, CHU Bicêtre, 94270 Le Kremlin-Bicêtre, France; (V.D.); (A.I.); (L.G.)
| | - Alex Iep
- Department of Biophysics and Nuclear Medicine-Molecular Imaging, Hôpitaux Universitaires Paris Saclay, Assistance Publique-Hôpitaux de Paris, CHU Bicêtre, 94270 Le Kremlin-Bicêtre, France; (V.D.); (A.I.); (L.G.)
| | - Léa Gomez
- Department of Biophysics and Nuclear Medicine-Molecular Imaging, Hôpitaux Universitaires Paris Saclay, Assistance Publique-Hôpitaux de Paris, CHU Bicêtre, 94270 Le Kremlin-Bicêtre, France; (V.D.); (A.I.); (L.G.)
| | - Lucas Goldfarb
- Service Hospitalier Frédéric Joliot-CEA, 91401 Orsay, France;
| | - Florent L. Besson
- Department of Biophysics and Nuclear Medicine-Molecular Imaging, Hôpitaux Universitaires Paris Saclay, Assistance Publique-Hôpitaux de Paris, CHU Bicêtre, 94270 Le Kremlin-Bicêtre, France; (V.D.); (A.I.); (L.G.)
- Université Paris Saclay, CEA, CNRS, Inserm, BioMaps, 91401 Orsay, France
- School of Medicine, Université Paris Saclay, 94720 Le Kremlin-Bicêtre, France
| |
Collapse
|
37
|
Clinical Utility of 18F-FDG PET in Neuroendocrine Tumors Prior to Peptide Receptor Radionuclide Therapy: A Systematic Review and Meta-Analysis. Cancers (Basel) 2021; 13:cancers13081813. [PMID: 33920195 PMCID: PMC8069875 DOI: 10.3390/cancers13081813] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/05/2021] [Accepted: 04/08/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Functional imaging with 18F-fluorodeoxyglucose positron emission tomography (18F-FDG PET) has evolved into a major clinical tool in cancer diagnosis and management for many malignancies in diverse clinical settings, providing valuable information on tumor behavior and aggressiveness. In the field of neuroendocrine tumors (NETs), recent advances in molecular imaging and targeted treatments with novel theranostic agents favor a more patient-tailored approach. Although peptide receptor radionuclide therapy (PRRT) has recently become an established therapy for progressive NETs, the role of 18F-FDG PET prior to PRRT in patients with NETs of different origins and grades remains to be determined. Herein, we provide a comprehensive summary of available evidence in contemporary literature by means of a systematic review and meta-analysis, demonstrating that dual-functional imaging with 68Ga-DOTA-peptides and 18F-FDG prior to PRRT appears to be a useful tool in NET management by delineating tumor somatostatin receptor expression and glycolytic metabolic activity, and predicting tumor response and survival outcomes. Abstract The role of 18F-FDG PET in patients with variable grades of neuroendocrine tumors (NETs) prior to peptide receptor radionuclide therapy (PRRT) has not been adequately elucidated. We aimed to evaluate the impact of 18F-FDG PET status on disease control rate (DCR), progression-free survival (PFS), and overall survival (OS) in neuroendocrine tumor (NET) patients receiving PRRT. We searched the MEDLINE, Embase, Cochrane Library, and Web of Science databases up to July 2020 and used the Newcastle-Ottawa scale (NOS) criteria to assess quality/risk of bias. A total of 5091 articles were screened. In 12 studies, 1492 unique patients with NETs of different origins were included. The DCR for patients with negative 18F-FDG PET status prior to PRRT initiation was 91.9%, compared to 74.2% in patients with positive 18F-FDG PET status (random effects odds ratio (OR): 4.85; 95% CI: 2.27–10.36). Adjusted analysis of pooled hazard ratios (HRs) confirmed longer PFS and OS in NET patients receiving PRRT with negative 18F-FDG PET (random effects HR:2.45; 95%CIs: 1.48–4.04 and HR:2.25; 95% CIs:1.55–3.28, respectively). In conclusion, 18F-FDG PET imaging prior to PRRT administration appears to be a useful tool in NET patients to predict tumor response and survival outcomes and a negative FDG uptake of the tumor is associated with prolonged PFS and OS.
Collapse
|
38
|
Rinzivillo M, Prosperi D, Mazzuca F, Magi L, Iannicelli E, Pilozzi E, Franchi G, Laghi A, Annibale B, Signore A, Panzuto F. [ 18F]FDG-PET/CT and long-term responses to everolimus in advanced neuroendocrine neoplasia. J Endocrinol Invest 2021; 44:811-818. [PMID: 32767279 DOI: 10.1007/s40618-020-01378-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 07/30/2020] [Indexed: 02/08/2023]
Abstract
PURPOSE This study aims to identify in patients with neuroendocrine neoplasia (NEN) the potential correlation between FDG-PET findings and responses to everolimus therapy to identify predictors of long-term efficacy. METHODS Retrospective analysis of patients with sporadic, advanced, progressive NEN treated with everolimus was performed based on the available data on FDG-PET patients obtained before commencing therapy. Data are expressed as the median (25-75th IQR). Risk factor analysis and survival analysis were performed by logistic regression and Cox proportional hazard regression and the determination of Kaplan-Meier curves, as appropriate. RESULTS Sixty-six patients were evaluated (NET G1 19.7%, NET G2 75.7%, and NET G3 4.6%), including 45.4% with positive FDG-PET findings. Overall, disease stabilization and a partial response were achieved for 71.2% and 6% of patients, respectively. A long-term response (> 24 months) was observed in 33% of patients. Ki67 was the only predictor of tumor progression (p = 0.03). No significant difference in clinical outcomes was observed between patients with positive or negative FDG-PET findings (median PFS was 24 months and 18 months, respectively, p = 0.337; the disease control rate was 83.3% and 70%, respectively, p = 0.245). CONCLUSIONS Everolimus is a valid therapeutic option for advanced, progressive, well-differentiated NEN, even in patients with positive FDG-PET findings.
Collapse
Affiliation(s)
- M Rinzivillo
- Digestive Disease Unit, ENETS Center of Excellence, Sant'Andrea University Hospital, Via di Grottarossa 1035, 00189, Rome, Italy
| | - D Prosperi
- Nuclear Medicine Unit, ENETS Center of Excellence, Sant'Andrea University Hospital, Rome, Italy
| | - F Mazzuca
- Medical Oncology Unit, ENETS Center of Excellence, Sant'Andrea University Hospital, Rome, Italy
- Department of Clinical and Molecular Medicine, "Sapienza" University of Rome, Rome, Italy
| | - L Magi
- Digestive Disease Unit, ENETS Center of Excellence, Sant'Andrea University Hospital, Via di Grottarossa 1035, 00189, Rome, Italy
| | - E Iannicelli
- Radiology Unit, ENETS Center of Excellence, Sant'Andrea University Hospital, Rome, Italy
- Department of Medical-Surgical Sciences and of Translational Medicine, "Sapienza" University of Rome, Rome, Italy
| | - E Pilozzi
- Department of Clinical and Molecular Medicine, "Sapienza" University of Rome, Rome, Italy
- Pathology Unit, ENETS Center of Excellence, Sant'Andrea University Hospital, Rome, Italy
| | - G Franchi
- Nuclear Medicine Unit, ENETS Center of Excellence, Sant'Andrea University Hospital, Rome, Italy
| | - A Laghi
- Radiology Unit, ENETS Center of Excellence, Sant'Andrea University Hospital, Rome, Italy
- Department of Medical-Surgical Sciences and of Translational Medicine, "Sapienza" University of Rome, Rome, Italy
| | - B Annibale
- Digestive Disease Unit, ENETS Center of Excellence, Sant'Andrea University Hospital, Via di Grottarossa 1035, 00189, Rome, Italy
- Department of Medical-Surgical Sciences and of Translational Medicine, "Sapienza" University of Rome, Rome, Italy
| | - A Signore
- Nuclear Medicine Unit, ENETS Center of Excellence, Sant'Andrea University Hospital, Rome, Italy
- Department of Medical-Surgical Sciences and of Translational Medicine, "Sapienza" University of Rome, Rome, Italy
| | - F Panzuto
- Digestive Disease Unit, ENETS Center of Excellence, Sant'Andrea University Hospital, Via di Grottarossa 1035, 00189, Rome, Italy.
| |
Collapse
|
39
|
Combined use of 177Lu-DOTATATE and metronomic capecitabine (Lu-X) in FDG-positive gastro-entero-pancreatic neuroendocrine tumors. Eur J Nucl Med Mol Imaging 2021; 48:3260-3267. [PMID: 33604690 DOI: 10.1007/s00259-021-05236-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 02/01/2021] [Indexed: 01/27/2023]
Abstract
PURPOSE FDG-positive neuroendocrine tumors (NETs) have a poorer prognosis and exhibit shorter response duration to peptide receptor radionuclide therapy (PRRT). The aim of this prospective phase II study was to evaluate the efficacy and toxicity of PRRT with 177Lu-DOTATATE associated with metronomic capecitabine as a radiosensitizer agent in patients with advanced progressive FDG-positive gastro-entero-pancreatic (GEP) NETs. PATIENTS AND METHODS Patients with advanced somatostatin receptor- and FDG-positive G1-G3 GEP-NETs (Ki67 < 55%) were treated with a cumulative activity of 27.5 GBq of 177Lu-DOTATATE divided in five cycles of 5.5 GBq each every 8 weeks. Capecitabine (1000-1500 mg daily) was administered orally in the inter-cycle period between 177Lu-DOTATATE treatments. Prior to commencing capecitabine, all patients were triaged with the dihydropyrimidine dehydrogenase (DPD) test. Only DPD-proficient individuals were enrolled. The primary objectives were disease control rate (DCR) and safety. Secondary aims included progression-free (PFS) and overall survival (OS). Treatment response was assessed per Response Evaluation Criteria in Solid Tumors, version 1.1 (RECIST 1.1). Toxicity was assessed by Common Terminology Criteria for Adverse Events (CTCAE) version 4.0. RESULTS From August 2015 to December 2016, 37 subjects were consecutively enrolled. A total of 25 (68%) were affected by pancreatic neuroendocrine tumors (P-NETs), and 12 (32%) had gastrointestinal neuroendocrine tumors (GI-NETs). By grading (WHO 2010 classification), 12 patients (32%) had G1 (Ki67 ≤ 2%), 22 (59%) had G2 (3% < Ki67 ≤ 20%), and 3 patients (9%) had G3 (Ki67 > 20%) NETs. Grade 3 (G3) or 4 (G4) hematological toxicity occurred in 16.2% of patients. Other G3-G4 adverse events were diarrhea in 5.4% of cases and asthenia in 5.4%. No renal toxicity was observed for the duration of follow-up. In 37 patients, 33 were evaluable for response. Objective responses included partial response (PR) in 10 patients (30%) and stable disease (SD) in 18 patients (55%), with a DCR of 85%. The median follow-up was 38 months (range 4.6-51.1 months). The median PFS was 31.4 months (17.6-45.4), and mOS was not reached. CONCLUSIONS This study demonstrated that the combination of PRRT with 177Lu-DOTATATE and metronomic capecitabine is active and well tolerated in patients with aggressive FDG-positive G1-G3 GEP-NETs. These data constitute the basis for a randomized study of PPRT alone vs. PRRT plus metronomic capecitabine.
Collapse
|
40
|
Ambrosini V, Kunikowska J, Baudin E, Bodei L, Bouvier C, Capdevila J, Cremonesi M, de Herder WW, Dromain C, Falconi M, Fani M, Fanti S, Hicks RJ, Kabasakal L, Kaltsas G, Lewington V, Minozzi S, Cinquini M, Öberg K, Oyen WJG, O'Toole D, Pavel M, Ruszniewski P, Scarpa A, Strosberg J, Sundin A, Taïeb D, Virgolini I, Wild D, Herrmann K, Yao J. Consensus on molecular imaging and theranostics in neuroendocrine neoplasms. Eur J Cancer 2021; 146:56-73. [PMID: 33588146 DOI: 10.1016/j.ejca.2021.01.008] [Citation(s) in RCA: 151] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/31/2020] [Accepted: 01/07/2021] [Indexed: 02/07/2023]
Abstract
Nuclear medicine plays an increasingly important role in the management neuroendocrine neoplasms (NEN). Somatostatin analogue (SSA)-based positron emission tomography/computed tomography (PET/CT) and peptide receptor radionuclide therapy (PRRT) have been used in clinical trials and approved by the European Medicines Agency (EMA) and the US Food and Drug Administration (FDA). European Association of Nuclear Medicine (EANM) Focus 3 performed a multidisciplinary Delphi process to deliver a balanced perspective on molecular imaging and radionuclide therapy in well-differentiated neuroendocrine tumours (NETs). NETs form in cells that interact with the nervous system or in glands that produce hormones. These cells, called neuroendocrine cells, can be found throughout the body, but NETs are most often found in the abdomen, especially in the gastrointestinal tract. These tumours may also be found in the lungs, pancreas and adrenal glands. In addition to being rare, NETs are also complex and may be difficult to diagnose. Most NETs are non-functioning; however, a minority present with symptoms related to hypersecretion of bioactive compounds. NETs often do not cause symptoms early in the disease process. When diagnosed, substantial number of patients are already found to have metastatic disease. Several societies' guidelines address Neuroendocrine neoplasms (NENs) management; however, many issues are still debated, due to both the difficulty in acquiring strong clinical evidence in a rare and heterogeneous disease and the different availability of diagnostic and therapeutic options across countries. EANM Focus 3 reached consensus on employing 68gallium-labelled somatostatin analogue ([68Ga]Ga-DOTA-SSA)-based PET/CT with diagnostic CT or magnetic resonance imaging (MRI) for unknown primary NET detection, metastatic NET, NET staging/restaging, suspected extra-adrenal pheochromocytoma/paraganglioma and suspected paraganglioma. Consensus was reached on employing 18fluorine-fluoro-2-deoxyglucose ([18F]FDG) PET/CT in neuroendocrine carcinoma, G3 NET and in G1-2 NET with mismatched lesions (CT-positive/[68Ga]Ga-DOTA-SSA-negative). Peptide receptor radionuclide therapy (PRRT) was recommended for second line treatment for gastrointestinal NET with [68Ga]Ga-DOTA-SSA uptake in all lesions, in G1/G2 NET at disease progression, and in a subset of G3 NET provided all lesions are positive at [18F]FDG and [68Ga]Ga-DOTA-SSA. PRRT rechallenge may be used for in patients with stable disease for at least 1 year after therapy completion. An international consensus is not only a prelude to a more standardised management across countries but also serves as a guide for the direction to follow when designing new research studies.
Collapse
Affiliation(s)
- Valentina Ambrosini
- IRCCS, Azienda Ospedaliero-Universitaria di Bologna, Italy; Nuclear Medicine, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Italy
| | - Jolanta Kunikowska
- Nuclear Medicine Department, Medical University of Warsaw, Warsaw, Poland
| | - Eric Baudin
- Endocrine Oncolgy Unit, Institut Gustave Roussy, Villejuif Cedex, France
| | - Lisa Bodei
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Catherine Bouvier
- International Neuroendocrine Cancer Alliance (INCA), Leamington Spa, UK
| | - Jaume Capdevila
- Medical Oncology Department, Vall Hebron University Hospital, Vall Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Marta Cremonesi
- Radiation Research Unit, Istituto Europeo di Oncologia, IRCCS, Milano, Italy
| | - Wouter W de Herder
- Erasmus MC & Erasmus MC Cancer Center, ENETS Center of Excellence Rotterdam, Rotterdam, the Netherlands
| | | | - Massimo Falconi
- Pancreas Translational & Research Institute, Scientific Institute San Raffaele Hospital and University Vita-Salute, Milan, Italy
| | - Melpomeni Fani
- Division of Radiopharmaceutical Chemistry, University Hospital Basel, Basel, Switzerland
| | - Stefano Fanti
- IRCCS, Azienda Ospedaliero-Universitaria di Bologna, Italy; Nuclear Medicine, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Italy
| | - Rodney J Hicks
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| | - Levent Kabasakal
- Istanbul University-Cerrahpaşa, Faculty of Medicine, Department of Nuclear Medicine, Turkey
| | - Gregory Kaltsas
- National and Kapodistrian University of Athens, Athens, Greece
| | | | - Silvia Minozzi
- Laboratory of Clinical Research Methodology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Michela Cinquini
- Laboratory of Clinical Research Methodology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Kjell Öberg
- Dept of Endocrine Oncology, University Hospital Uppsala, Sweden
| | - Wim J G Oyen
- Humanitas University and Humanitas Clinical and Research Center, Milan, Italy; Department of Radiology and Nuclear Medicine, Radboud University Medical Centre, Nijmegen, the Netherlands; Department of Radiology and Nuclear Medicine, Rijnstate Hospital Arnhem, the Netherlands
| | | | - Marianne Pavel
- Friedrich Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Philippe Ruszniewski
- Department of Pancreatology, Beaujon Hospital, Université de Paris, Clichy, France
| | - Aldo Scarpa
- ARC-NET Centre for Applied Research on Cancer and Department of Pathology, University of Verona, Italy
| | | | - Anders Sundin
- Department of Surgical Sciences, Uppsala University, University Hospital, Sweden
| | - David Taïeb
- Department of Nuclear Medicine, La Timone University Hospital, Aix-Marseille University, Marseille, France
| | - Irene Virgolini
- Department of Nuclear Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Damian Wild
- Division of Nuclear Medicine, University Hospital Basel, Basel, Switzerland
| | - Ken Herrmann
- Department of Nuclear Medicine, Universitätsklinikum, Essen, Germany.
| | - James Yao
- Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
41
|
Magi L, Rinzivillo M, Panzuto F. Tumor Heterogenity in Gastro-Entero-Pancreatic Neuroendocrine Neoplasia. ENDOCRINES 2021; 2:28-36. [DOI: 10.3390/endocrines2010003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025] Open
Abstract
Owing to the rarity and the biological and clinical heterogeneity of gastroenteropancreatic neuroendocrine neoplasia (GEP NEN), the management of these patients may be challenging for physicians. This review highlights the specific features of GEP NEN with particular attention on the role of Ki67 heterogeneity, the potential prognostic role of novel radiological techniques, and the clinical usefulness of functional imaging, including 68Ga-DOTA-SST PET/CT and 18F-FDG PET/CT. Understanding these specific features may help to plan proper and tailored follow-up programs and therapeutic approaches.
Collapse
Affiliation(s)
- Ludovica Magi
- SAIMLAL Department (Human Anatomy, Histology, Forensic Medicine and Orthopedics), Sapienza University of Rome, 00185 Rome, Italy
- Digestive Disease Unit, Sant’Andrea University Hospital—ENETS Center of Excellence of Via di Grottarossa, 1035, 00189 Rome, Italy
| | - Maria Rinzivillo
- Digestive Disease Unit, Sant’Andrea University Hospital—ENETS Center of Excellence of Via di Grottarossa, 1035, 00189 Rome, Italy
| | - Francesco Panzuto
- Digestive Disease Unit, Sant’Andrea University Hospital—ENETS Center of Excellence of Via di Grottarossa, 1035, 00189 Rome, Italy
| |
Collapse
|
42
|
Liberini V, Huellner MW, Grimaldi S, Finessi M, Thuillier P, Muni A, Pellerito RE, Papotti MG, Piovesan A, Arvat E, Deandreis D. The Challenge of Evaluating Response to Peptide Receptor Radionuclide Therapy in Gastroenteropancreatic Neuroendocrine Tumors: The Present and the Future. Diagnostics (Basel) 2020; 10:E1083. [PMID: 33322819 PMCID: PMC7763988 DOI: 10.3390/diagnostics10121083] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/09/2020] [Accepted: 12/09/2020] [Indexed: 02/07/2023] Open
Abstract
The NETTER-1 study has proven peptide receptor radionuclide therapy (PRRT) to be one of the most effective therapeutic options for metastatic neuroendocrine tumors (NETs), improving progression-free survival and overall survival. However, PRRT response assessment is challenging and no consensus on methods and timing has yet been reached among experts in the field. This issue is owed to the suboptimal sensitivity and specificity of clinical biomarkers, limitations of morphological response criteria in slowly growing tumors and necrotic changes after therapy, a lack of standardized parameters and timing of functional imaging and the heterogeneity of PRRT protocols in the literature. The aim of this article is to review the most relevant current approaches for PRRT efficacy prediction and response assessment criteria in order to provide an overview of suitable tools for safe and efficacious PRRT.
Collapse
Affiliation(s)
- Virginia Liberini
- Nuclear Medicine Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (S.G.); (M.F.); (P.T.); (D.D.)
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland;
| | - Martin W. Huellner
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland;
| | - Serena Grimaldi
- Nuclear Medicine Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (S.G.); (M.F.); (P.T.); (D.D.)
| | - Monica Finessi
- Nuclear Medicine Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (S.G.); (M.F.); (P.T.); (D.D.)
| | - Philippe Thuillier
- Nuclear Medicine Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (S.G.); (M.F.); (P.T.); (D.D.)
- Department of Endocrinology, University Hospital of Brest, 29200 Brest, France
| | - Alfredo Muni
- Department of Nuclear Medicine, S.S. Biagio e Antonio e C. Arrigo Hospital, 15121 Alessandria, Italy;
| | | | - Mauro G. Papotti
- Pathology Unit, City of Health and Science University Hospital, 10126 Turin, Italy;
- Department of Oncology, University of Turin at Molinette Hospital, 10126 Turin, Italy
| | - Alessandro Piovesan
- Department of Endocrinology, A. O. U. Città della Salute della Scienza of Turin, 10126 Turin, Italy;
| | - Emanuela Arvat
- Oncological Endocrinology, Department of Medical Sciences, University of Turin, 10126 Turin, Italy;
| | - Désirée Deandreis
- Nuclear Medicine Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (S.G.); (M.F.); (P.T.); (D.D.)
| |
Collapse
|
43
|
Calabrò D, Argalia G, Ambrosini V. Role of PET/CT and Therapy Management of Pancreatic Neuroendocrine Tumors. Diagnostics (Basel) 2020; 10:E1059. [PMID: 33297381 PMCID: PMC7762240 DOI: 10.3390/diagnostics10121059] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/02/2020] [Accepted: 12/04/2020] [Indexed: 12/12/2022] Open
Abstract
Pancreatic neuroendocrine neoplasms (panNENs) are heterogeneous neoplasms with neuroendocrine differentiation that show peculiar clinical and histomorphological features, with variable prognosis. In recent years, advances in knowledge regarding the pathophysiology and heterogeneous clinical presentation, as well as the availability of different diagnostic procedures for panNEN diagnosis and novel therapeutic options for patient clinical management, has led to the recognition of the need for an active multidisciplinary discussion for optimal patient care. Molecular imaging with positron emission tomography/computed tomography (PET/CT) has become indispensable for the management of panNENs. Several PET radiopharmaceuticals can be used to characterize either panNEN receptor expression or metabolism. The aim of this review is to offer an overview of all the currently used radiopharmaceuticals and of the new upcoming tracers for pancreatic neuroendocrine tumors (panNETs), and their clinical impact on therapy management. [68Ga]Ga-DOTA-peptide PET/CT (SSA-PET/CT) has high sensitivity, specificity, and accuracy and is recommended for the staging and restaging of any non-insulinoma well-differentiated panNEN cases to carry out detection of unknown primary tumor sites or early relapse and for evaluation of in vivo somatostatin receptors expression (SRE) to select patient candidates for peptide receptor radiometabolic treatment (PRRT) with 90Y or 177Lu and/or cold analogs. SSA-PET/CT also has a strong impact on clinical management, leading to a change in treatment in approximately a third of the cases. Its role for treatment response assessment is still under debate due to the lack of standardized criteria, even though some semiquantitative parameters seem to be able to predict response. [18F]FDG PET/CT generally shows low sensitivity in small growing and well-differentiated neuroendocrine tumors (NET; G1 and G2), while it is of utmost importance in the evaluation and management of high-grade NENs and also provides important prognostic information. When positive, [18F]FDG PET/CT impacts therapeutical management, indicating the need for a more aggressive treatment regime. Although FDG positivity does not exclude the patient from PRRT, several studies have demonstrated that it is certainly useful to predict response, even in this setting. The role of [18F]FDOPA for the study of panNET is limited by physiological uptake in the pancreas and is therefore not recommended. Moreover, it provides no information on SRE that has crucial clinical management relevance. Early acquisition of the abdomen and premedication with carbidopa may be useful to increase the accuracy, but further studies are needed to clarify its utility. GLP-1R agonists, such as exendin-4, are particularly useful for benign insulinoma detection, but their accuracy decreases in the case of malignant insulinomas. Being a whole-body imaging technique, exendin-PET/CT gives important preoperative information on tumor size and localization, which is fundamental for surgical planning as resection (enucleation of the lesion or partial pancreatic resection) is the only curative treatment. New upcoming tracers are under study, such as promising SSTR antagonists, which show a favorable biodistribution and higher tumor-to-background ratio that increases tumor detection, especially in the liver. [68Ga]pentixafor, an in vivo marker of CXCR4 expression associated with the behavior of more aggressive tumors, seems to only play a limited role in detecting well-differentiated NET since there is an inverse expression of SSTR2 and CXCR4 in G1 to G3 NETs with an elevation in CXCR4 and a decrease in SSTR2 expression with increasing grade. Other tracers, such as [68Ga]Ga-PSMA, [68Ga]Ga-DATA-TOC, [18F]SiTATE, and [18F]AlF-OC, are also under investigation.
Collapse
Affiliation(s)
- Diletta Calabrò
- Department of Nuclear Medicine, IRCCS Azienda Ospedaliera-Universitaria di Bologna, 40138 Bologna, Italy; (G.A.); (V.A.)
- Department of Nuclear Medicine, DIMES University of Bologna, 40138 Bologna, Italy
| | - Giulia Argalia
- Department of Nuclear Medicine, IRCCS Azienda Ospedaliera-Universitaria di Bologna, 40138 Bologna, Italy; (G.A.); (V.A.)
- Department of Nuclear Medicine, DIMES University of Bologna, 40138 Bologna, Italy
| | - Valentina Ambrosini
- Department of Nuclear Medicine, IRCCS Azienda Ospedaliera-Universitaria di Bologna, 40138 Bologna, Italy; (G.A.); (V.A.)
- Department of Nuclear Medicine, DIMES University of Bologna, 40138 Bologna, Italy
| |
Collapse
|
44
|
Binderup T, Knigge U, Johnbeck CB, Loft A, Berthelsen AK, Oturai P, Mortensen J, Federspiel B, Langer SW, Kjaer A. 18F-FDG PET is Superior to WHO Grading as a Prognostic Tool in Neuroendocrine Neoplasms and Useful in Guiding PRRT: A Prospective 10-Year Follow-up Study. J Nucl Med 2020; 62:808-815. [PMID: 33067340 DOI: 10.2967/jnumed.120.244798] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 09/17/2020] [Indexed: 12/12/2022] Open
Abstract
Accurate grading of patients with neuroendocrine neoplasms (NENs) is essential for risk stratification and optimal choice of therapy. Currently, grading is based on histologically assessed degree of tumor proliferation. The aim of the present study was to assess the long-term prognostic value of 18F-FDG PET imaging for risk stratification of NENs and compare it with tumor grading (World Health Organization 2010 classification). Methods: We conducted a prospective cohort study evaluating the prognostic value of 18F-FDG PET imaging and compared it with histologic grading. Enrolled were 166 patients of all grades and with histologically confirmed NENs of gastroenteropancreatic origin. The primary endpoint was overall survival (OS). Progression-free survival (PFS) was a secondary endpoint. In addition, OS in relation to peptide receptor radionuclide therapy (PRRT) was analyzed as an exploratory endpoint. The median follow-up time was 9.8 y. Results: Analysis of the whole cohort revealed that a positive 18F-FDG PET scan was associated with a shorter OS than a negative 18F-FDG PET scan (hazard ratio: 3.8; 95% CI: 2.4-5.9; P < 0.001). In G1 and G2 patients (n = 140), a positive 18F-FDG PET scan was the only identifier of high risk for death (hazard ratio: 3.6; 95% CI, 2.2-5.9; P < 0.001). In multivariate analysis, 18F-FDG PET, G3 tumor, ≥2 liver metastases, and ≥2 prior therapies were independent prognostic factors for OS, and 18F-FDG PET, G3 tumor, and ≥3 liver metastases were independent prognostic factors for PFS. For patients receiving PRRT, 18F-FDG-negative cases had a significantly longer survival than 18F-FDG-positive cases, whereas no difference was identified for tumor grading. 18F-FDG-positive patients receiving PRRT had a significantly longer median survival than patients not receiving PRRT (4.4 vs. 1.4 y, P = 0.001), whereas no difference was seen for 18F-FDG-negative patients. Conclusion: 18F-FDG PET is useful for risk stratification of all NEN grades and is superior to histologic grading. 18F-FDG PET could differentiate G1 and G2 tumors into low- and high-risk groups. In the selection of therapy and for risk stratification of NEN patients, 18F-FDG PET status should be considered.
Collapse
Affiliation(s)
- Tina Binderup
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet & University of Copenhagen, Copenhagen, Denmark.,European Neuroendocrine Tumors Society Center of Excellence, Rigshospitalet, Copenhagen, Denmark
| | - Ulrich Knigge
- European Neuroendocrine Tumors Society Center of Excellence, Rigshospitalet, Copenhagen, Denmark.,Department of Surgical Gastroenterology, Rigshospitalet, Copenhagen, Denmark.,Department of Clinical Endocrinology, Rigshospitalet, Copenhagen, Denmark
| | - Camilla Bardram Johnbeck
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet & University of Copenhagen, Copenhagen, Denmark.,European Neuroendocrine Tumors Society Center of Excellence, Rigshospitalet, Copenhagen, Denmark
| | - Annika Loft
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet & University of Copenhagen, Copenhagen, Denmark.,European Neuroendocrine Tumors Society Center of Excellence, Rigshospitalet, Copenhagen, Denmark
| | - Anne Kiil Berthelsen
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet & University of Copenhagen, Copenhagen, Denmark.,European Neuroendocrine Tumors Society Center of Excellence, Rigshospitalet, Copenhagen, Denmark
| | - Peter Oturai
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet & University of Copenhagen, Copenhagen, Denmark.,European Neuroendocrine Tumors Society Center of Excellence, Rigshospitalet, Copenhagen, Denmark
| | - Jann Mortensen
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet & University of Copenhagen, Copenhagen, Denmark.,European Neuroendocrine Tumors Society Center of Excellence, Rigshospitalet, Copenhagen, Denmark
| | - Birgitte Federspiel
- European Neuroendocrine Tumors Society Center of Excellence, Rigshospitalet, Copenhagen, Denmark.,Department of Pathology, Rigshospitalet, Copenhagen, Denmark; and
| | - Seppo W Langer
- European Neuroendocrine Tumors Society Center of Excellence, Rigshospitalet, Copenhagen, Denmark.,Department of Oncology, Rigshospitalet, Copenhagen, Denmark
| | - Andreas Kjaer
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet & University of Copenhagen, Copenhagen, Denmark .,European Neuroendocrine Tumors Society Center of Excellence, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
45
|
Ostwal V, Basu S, Bhargava P, Shah M, Parghane RV, Srinivas S, Chaudhari V, Bhandare MS, Shrikhande SV, Ramaswamy A. Capecitabine-Temozolomide in Advanced Grade 2 and Grade 3 Neuroendocrine Neoplasms: Benefits of Chemotherapy in Neuroendocrine Neoplasms with Significant 18FDG Uptake. Neuroendocrinology 2020; 111:998-1004. [PMID: 33017827 DOI: 10.1159/000511987] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 09/29/2020] [Indexed: 01/27/2023]
Abstract
INTRODUCTION Capecitabine-temozolomide (CAPTEM) chemotherapy, alone or with concurrent peptide receptor radionuclide therapy (PRRT), has activity in advanced WHO grade 2 and grade 3 neuroendocrine neoplasms (NENs). The objective of this study was to evaluate the activity of the CAPTEM in patients with grade 2 and grade 3 NENs and identify prognostic factors. MATERIALS AND METHODS A retrospective analysis of patients with metastatic grade 2 and grade 3 NENs, who were having baseline significant dual uptake on 68Ga-DOTATATE/18F-fluorodeoxyglucose (FDG)-PET-CT scan and treated with CAPTEM chemotherapy between January 2014 and December 2019 at Tata Memorial Hospital, was conducted. The clinical variables and survival data were collected. Progression-free survival (PFS) was estimated using the Kaplan-Meier method. RESULTS A total of 68 patients received the CAPTEM regimen, of whom 29 patients (43%) received CAPTEM alone and 39 patients (57%) received concurrent PRRT. The primary sites were pancreas in 32 (47%) and small intestine in 12 (18%) patients. Mean Ki-67 index was 12.6% (range: 3-50). Forty-five patients (65%) were treatment naïve. There were no significant differences in baseline clinical variables between patients treated with CAPTEM alone or with CAPTEM-PRRT. Both regimens were well tolerated. With a median follow-up of 22.1 months, the median PFS for the entire cohort was 27.5 months. There was no statistical difference in the median PFS between patients receiving CAPTEM alone or CAPTEM-PRRT (33.7 vs. 22 months; p = 0.199). A Ki-67 index of >5% predicted for inferior PFS on multivariate analysis (24 versus 73.8 months; p = 0.04; hazard ratio -3.77; 95% confidence interval: 1.07-13.26). CONCLUSION CAPTEM, alone or concurrent with PRRT, has a significant activity in grade 2 and grade 3 NENs with dual SSTR and 18FDG expression. A Ki-67 index >5% predicts strongly for inferior outcomes and should be further explored as a prognostic cutoff in grade 2 NENs. Early initiation of CAPTEM should be considered in this group of tumors with significant baseline 18FDG expression.
Collapse
Affiliation(s)
- Vikas Ostwal
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Sandip Basu
- Radiation Medicine Centre, Babha Atomic Research Centre, Tata Memorial Hospital Annexe, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Prabhat Bhargava
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Minit Shah
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Rahul Vithalrao Parghane
- Radiation Medicine Centre, Babha Atomic Research Centre, Tata Memorial Hospital Annexe, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Sujay Srinivas
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Vikram Chaudhari
- GI and HPB Surgery, Tata Memorial Hospital (Homi Bhabha National Institute), Mumbai, India
| | - Manish S Bhandare
- GI and HPB Surgery, Tata Memorial Hospital (Homi Bhabha National Institute), Mumbai, India
| | - Shailesh V Shrikhande
- GI and HPB Surgery, Tata Memorial Hospital (Homi Bhabha National Institute), Mumbai, India
| | - Anant Ramaswamy
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, India,
| |
Collapse
|
46
|
Paganelli G, Bodei L, Modlin I. A metastatic tumor is no different to a viral pandemic: lessons learnt from COVID-19 may teach us to change the PRRT paradigm. Eur J Nucl Med Mol Imaging 2020; 47:2223-2226. [PMID: 32564170 PMCID: PMC7305056 DOI: 10.1007/s00259-020-04904-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Giovanni Paganelli
- Nuclear Medicine Operative Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014, Meldola, Italy
| | - Lisa Bodei
- Department Nuclear Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Irvin Modlin
- School of Medicine, Yale University, New Haven, CT, 06520, USA.
- Gastroenterological and Endoscopic Surgery, Yale University School of Medicine, New Haven, CT, 06520-8062, USA.
| |
Collapse
|
47
|
Tsoli M, Alexandraki K, Xanthopoulos C, Kassi E, Kaltsas G. Medical Treatment of Gastrointestinal Neuroendocrine Neoplasms. Horm Metab Res 2020; 52:614-620. [PMID: 32108932 DOI: 10.1055/a-1110-7251] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Neuroendocrine neoplasms (NENs) are rare tumours that arise mainly in the gastrointestinal or pulmonary system. Most NENs are well-differentiated and may obtain prolonged survival besides the presence of metastatic disease; however, a subset (poorly differentiated NENs) may display a truly aggressive behaviour exhibiting a poor prognosis. The recently developed classification systems along with advances in functional imaging have helped stratify patients to the administration of appropriate therapeutic options. Surgery is the mainstay of treatment of NENs, but in recent decades there has been a considerable evolution of medical treatments that are used for locally advanced or metastatic disease not amenable to surgical resection. Long acting somatostatin analogues are the main therapeutic modality for patients with functioning and well-differentiated low grade NENs exhibiting symptomatic control and mainly stabilisation of tumour growth. Other systemic treatments include chemotherapy, molecular targeted agents, interferon-α, peptide receptor radionuclide therapy (PRRT), and immunotherapy. In addition, new agents such as telotristat may be used for the control of symptoms of carcinoid syndrome. The choice and/or sequence of therapeutic agents should be individualized according to tumour origin and differentiation, disease burden, presence of clinical symptoms and patients' performance status in the context of a multidisciplinary approach. Recent advances in the molecular pathogenesis of NENs set the field for a more personalised treatment approach.
Collapse
Affiliation(s)
- Marina Tsoli
- 1st Department of Propaedeutic and Internal Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Krystallenia Alexandraki
- 1st Department of Propaedeutic and Internal Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Christos Xanthopoulos
- 1st Department of Propaedeutic and Internal Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Eva Kassi
- 1st Department of Propaedeutic and Internal Medicine, National and Kapodistrian University of Athens, Athens, Greece
- Department of Biological Chemistry, National and Kapodistrian University of Athens, Athens, Greece
| | - Gregory Kaltsas
- 1st Department of Propaedeutic and Internal Medicine, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
48
|
Evangelista L, Ravelli I, Bignotto A, Cecchin D, Zucchetta P. Ga-68 DOTA-peptides and F-18 FDG PET/CT in patients with neuroendocrine tumor: A review. Clin Imaging 2020; 67:113-116. [PMID: 32559681 DOI: 10.1016/j.clinimag.2020.05.035] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 05/18/2020] [Accepted: 05/29/2020] [Indexed: 01/17/2023]
Abstract
OBJECTIVES The aim of the present review was to assess the role of combined 18F-Fluorodeoxyglucose (F-18 FDG) and Ga-68 DOTA-peptides positron emission tomography (PET)-computed tomography (CT) in neuroendocrine tumors (NETs). METHODS We have searched MEDLINE databases, including PubMed and Scopus, for studies about the combined FDG and Ga-68 DOTA-peptides PET-CT or PET/Magnetic Resonance Imaging (MRI) in NETs in the last 15 years (from 2004 to November 2019). No limits were applied to the search strategy. Abstracts, reviews, letters to editors, and editorials were excluded. RESULTS Seven studies met the inclusion criteria. In total 236 patients received both 68Ga-DOTA-peptides and F-18 FDG PET-CT for the characterization of NETs. In particular, 84 patients had a neuroendocrine lung tumor while the others mainly a gastroenteropancreatic NET. The combined use of F-18 FDG and Ga-68 DOTA-peptides (mainly TOC) PET studies provides complementary information regarding different biological characteristics of the lesions, thus enabling a more accurate selection of patients for targeted radionuclide therapy and a better stratification of the prognosis. CONCLUSIONS Ga-68 DOTA-peptides and F-18 FDG PET should be considered complementary in patients with NETs. They should be both performed in the initial staging and during follow-up, with a specific selection of patients and in a multidisciplinary vision.
Collapse
Affiliation(s)
- Laura Evangelista
- Nuclear Medicine Unit, Department of Medicine - DIMED, University of Padua, Padua, Italy.
| | - Ilaria Ravelli
- Nuclear Medicine Unit, Department of Medicine - DIMED, University of Padua, Padua, Italy
| | - Antonio Bignotto
- Nuclear Medicine Unit, Department of Medicine - DIMED, University of Padua, Padua, Italy
| | - Diego Cecchin
- Nuclear Medicine Unit, Department of Medicine - DIMED, University of Padua, Padua, Italy
| | - Pietro Zucchetta
- Nuclear Medicine Unit, Department of Medicine - DIMED, University of Padua, Padua, Italy
| |
Collapse
|
49
|
Paganelli G, Sansovini M, Nicolini S, Grassi I, Ibrahim T, Amadori E, Di Iorio V, Monti M, Scarpi E, Bongiovanni A, Altini M, Urso L, Cittanti C, Matteucci F, Severi S. 177Lu-PRRT in advanced gastrointestinal neuroendocrine tumors: 10-year follow-up of the IRST phase II prospective study. Eur J Nucl Med Mol Imaging 2020; 48:152-160. [PMID: 32472437 DOI: 10.1007/s00259-020-04873-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 05/18/2020] [Indexed: 12/11/2022]
Abstract
PURPOSE In March 2014, we reported the activity and safety of 177Lu-DOTA-octreotate peptide receptor radionuclide therapy (Lu-PRRT) at two different dosages (18.5 GBq and 27.5 GBq in 5 cycles) in patients with progressive metastatic gastrointestinal neuroendocrine tumors (GI-NETs). Disease control rate (DCR) and toxicity were addressed. Herein, we report the late toxicity, progression-free survival (PFS), and overall survival (OS) in the same cohort after a 10-year follow-up. METHODS We conducted an open-label, disease-oriented prospective phase II trial. From March 2008 to June 2011, 43 patients received 3.7 GBq or 5.5 GBq of Lu-PRRT every 6 to 8 weeks, each cycle repeated 5 times. All patients showed 68Gallium-DOTA-peptide PET/Octreoscan® positivity (score 3-4 Rotterdam scale) in known lesions. Tumor burden was estimated radiologically. Time-to-event data (PFS and OS) were described using Kaplan-Meier curves and compared with the log-rank test. RESULTS Forty-three patients (28 males and 15 females) were evaluable and were monitored for a median period of 118 months (range 12.6-139.6). Median PFS in patients receiving 18.5 GBq was 59.8 months (95% confidence interval [95% CI] 14.3-79.6), identical to that of patients treated with 27.5 GBq (59.8 months, 95% CI 23.4-82.0). Median OS was 71.0 months (95% CI 46.1-107.3) in the group who received 18.5 GBq and 97.6 months (95% CI 64.3-not reached) in the group treated with 27.5 GBq (P = 0.22). Patients with progression limited to lymph nodes showed significantly longer median PFS and OS than those with hepatic lesions (P = 0.02 for PFS and P = 0.04 for OS). Age over 65 years at the time of PRRT was also significant for OS. Of note, no late hematological or renal toxicity was observed in either group. CONCLUSIONS The long-term follow-up of the IRST phase II study shows that Lu-PRRT is a safe and effective therapy for patients with advanced GI-NET, the most important prognostic factor being tumor burden, hepatic lesions, and age. We believe that Lu-PRRT should be offered to patients with early-stage disease.
Collapse
Affiliation(s)
- Giovanni Paganelli
- Nuclear Medicine and Radiometabolic Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via P. Maroncelli 40, 47014, Meldola, Italy
| | - Maddalena Sansovini
- Nuclear Medicine and Radiometabolic Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via P. Maroncelli 40, 47014, Meldola, Italy.
| | - Silvia Nicolini
- Nuclear Medicine and Radiometabolic Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via P. Maroncelli 40, 47014, Meldola, Italy
| | - Ilaria Grassi
- Nuclear Medicine and Radiometabolic Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via P. Maroncelli 40, 47014, Meldola, Italy
| | - Toni Ibrahim
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Elena Amadori
- Radiology Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Valentina Di Iorio
- Oncology Pharmacy, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Manuela Monti
- Unit of Biostatistics and Clinical Trials, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Emanuela Scarpi
- Unit of Biostatistics and Clinical Trials, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Alberto Bongiovanni
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Mattia Altini
- Healthcare Administration, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Luca Urso
- Nuclear Medicine Unit, University of Ferrara, Ferrara, Italy
| | | | - Federica Matteucci
- Nuclear Medicine and Radiometabolic Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via P. Maroncelli 40, 47014, Meldola, Italy
| | - Stefano Severi
- Nuclear Medicine and Radiometabolic Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via P. Maroncelli 40, 47014, Meldola, Italy
| |
Collapse
|
50
|
Bocchini M, Nicolini F, Severi S, Bongiovanni A, Ibrahim T, Simonetti G, Grassi I, Mazza M. Biomarkers for Pancreatic Neuroendocrine Neoplasms (PanNENs) Management-An Updated Review. Front Oncol 2020; 10:831. [PMID: 32537434 PMCID: PMC7267066 DOI: 10.3389/fonc.2020.00831] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Accepted: 04/28/2020] [Indexed: 12/14/2022] Open
Abstract
Pancreatic neuroendocrine tumors (PanNENs) are rare sporadic cancers or develop as part of hereditary syndromes. PanNENs can be both functioning and non-functioning based on whether they produce bioactive peptides. Some PanNENs are well differentiated while others-poorly. Symptoms, thus, depend on both oncological and hormonal causes. PanNEN diagnosis and treatment benefit from and in some instances are guided by biomarker monitoring. However, plasmatic monoanalytes are only suggestive of PanNEN pathological status and their positivity is typically followed by deepen diagnostic analyses through imaging techniques. There is a strong need for new biomarkers and follow-up modalities aimed to improve the outcome of PanNEN patients. Liquid biopsy follow-up, i.e., sequential analysis on tumor biomarkers in body fluids offers a great potential, that need to be substantiated by additional studies focusing on the specific markers and the timing of the analyses. This review provides the most updated panorama on PanNEN biomarkers.
Collapse
Affiliation(s)
- Martine Bocchini
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Fabio Nicolini
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Stefano Severi
- Nuclear Medicine and Radiometabolic Units, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Alberto Bongiovanni
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Toni Ibrahim
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Giorgia Simonetti
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Ilaria Grassi
- Nuclear Medicine and Radiometabolic Units, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Massimiliano Mazza
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| |
Collapse
|