1
|
Piscopo L, Zampella E, Volpe F, Gaudieri V, Nappi C, Di Donna E, Clemente S, Varallo A, Scaglione M, Cuocolo A, Klain M. The Safety and Efficacy of Peptide Receptor Radionuclide Therapy for Gastro-Entero-Pancreatic Neuroendocrine Tumors: A Single Center Experience. Curr Oncol 2024; 31:5617-5629. [PMID: 39330044 PMCID: PMC11431754 DOI: 10.3390/curroncol31090416] [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: 07/23/2024] [Revised: 09/16/2024] [Accepted: 09/18/2024] [Indexed: 09/28/2024] Open
Abstract
The aim of the present study was to evaluate the safety and efficacy of radionuclide therapy with [177Lu]Lu-DOTA-TATE according to our single center experience at the University of Naples Federico II. For the present analysis, we considered 21 patients with progressive, advanced, well-differentiated G1 and G2 in patients with gastro-entero-pancreatic (GEP) neuroendocrine tumors (NETs) treated with [177Lu]Lu-DOTA-TATE according to the decisions of a multidisciplinary team. All patients underwent four cycles of 7-8 GBq of [177Lu]Lu-DOTA-TATE every 8 weeks. A whole-body scan (WBS) was performed 4, 48, and 168 h after each treatment. The dosimetry towards the organ at risk and target lesions was calculated. For each patient, renal and bone marrow parameters were evaluated before, during, and 3 months after the end of the treatment. Follow-up data were obtained and RECIST criteria were considered as the endpoint. Among 21 patients enrolled (mean age 65 ± 9 years); 17 (81%) were men and the small intestine was the most frequent location of disease (n = 12). A mild albeit significant variation (p < 0.05) in both platelets and white blood cell counts among all time points was observed, despite it disappearing 3 months after the end of the therapy. According to the RECIST criteria, 11 (55%) patients had a partial response to therapy and 8 (40%) had stable disease. Only one (5%) patient had disease progression 4 months after treatment. Our data confirm that [177Lu]Lu-DOTA is safe and effective in controlling the burden disease of G1/G2 GEP-NETs patients.
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
| | - Fabio Volpe
- 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
| | - Carmela Nappi
- Department of Advanced Biomedical Sciences, University of Naples, Federico II, 80131 Naples, Italy
| | - Erica Di Donna
- Department of Advanced Biomedical Sciences, University of Naples, Federico II, 80131 Naples, Italy
| | - Stefania Clemente
- Department of Advanced Biomedical Sciences, University of Naples, Federico II, 80131 Naples, Italy
| | - Antonio Varallo
- Department of Advanced Biomedical Sciences, University of Naples, Federico II, 80131 Naples, Italy
| | - Mariano Scaglione
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, 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
|
2
|
Bongiovanni A, Liverani C, Foca F, Bergamo F, Leo S, Pusceddu S, Gelsomino F, Brizzi MP, Di Meglio G, Spada F, Tamberi S, Lolli I, Cives M, Marconcini R, Pucci F, Berardi R, Antonuzzo L, Badalamenti G, Santini D, Recine F, Vanni S, Tebaldi M, Severi S, Rudnas B, Nanni O, Ranallo N, Crudi L, Calabrò L, Ibrahim T. A randomized phase II trial of Captem or Folfiri as second-line therapy in neuroendocrine carcinomas. Eur J Cancer 2024; 208:114129. [PMID: 39002347 DOI: 10.1016/j.ejca.2024.114129] [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: 02/08/2024] [Revised: 05/10/2024] [Accepted: 05/18/2024] [Indexed: 07/15/2024]
Abstract
BACKGROUND Neuroendocrine Carcinomas (NECs) prognosis is poor.No standard second-line therapy is currently recognized after failure of platinum-based first-line treatment. FOLFIRI and CAPTEM regimens have shown promising activity in preliminary studies. We aimed to evaluate these regimens in metastatic NEC patients. METHODS This is an open-label, multicenter, randomized non-comparative phase II trial to evaluate the activity and safety of FOLFIRI or CAPTEM in metastatic NEC patients. Primary endpoints were the 12 weeks-Disease Control Rate (12w-DCR) by investigator assessment per RECIST v1.1 and safety per CTCAE v5.0. Additional endpoints included overall response rate (ORR), progression-free survival (PFS) and overall survival (OS). Patients' serum samples were subject to NGS miRNome profiling in comparison with healthy donors to reveal differentially expressed miRNAs as candidate circulating biomarkers. RESULTS The study was halted for futility at interim analysis, as the minimum 12w-DCR threshold of 10 out of 25 patients required for the first step was not reached. From 06/03/2017 to 18/01/2021, 53 out of 112 patients were enrolled. Median follow-up was 22.6 months (range: 1.4-60.4). The 12w-DCR was 39.1 % in the FOLFIRI arm and 28.0 % in the CAPTEM arm. In the FOLFIRI subgroup the 12-months OS rate was 28.4 % (95 % CI: 12.7-46.5) while in the CAPTEM subgroup it was 32.4 % (95 % CI: 14.9-51.3). The most common G3-G4 side effects were neutropenia (n = 5, 18.5 %) and anemia (n = 2, 7.4 %) for FOLFIRI and G3-G4 thrombocytopenia (n = 2, 8.0 %), G4 nausea/vomiting (n = 1, 4.0 %) for CAPTEM. Three microRNAs emerged as NEC independent predictors. High expression values were found to be significantly associated with decreased PFS and OS. CONCLUSION The safety profile of FOLFIRI and CAPTEM was manageable. FOLFIRI and CAPTEM chemotherapy showed comparable activity in the second-line setting after progression on etoposide/platinum. CLINICALTRIALS GOV IDENTIFIER NCT03387592.
Collapse
Affiliation(s)
- Alberto Bongiovanni
- Osteoncology and Rare Tumor Center (CDO-TR), IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Chiara Liverani
- Bioscience Laboratory, Preclinic and Osteoncology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy.
| | - Flavia Foca
- Unit of Biostatistics and Clinical Trials, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Francesca Bergamo
- Medical Oncology Unit 1, IOV-Veneto Institute of Oncology IRCCS, Padua, Italy
| | - Silvana Leo
- Department of Medical Oncology, "Vito Fazzi" Hospital, Lecce, Italy
| | - Sara Pusceddu
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, ENETS Center of Excellence, Milan, Italy
| | - Fabio Gelsomino
- Department of Oncology and Hematology, Division of Oncology, University Hospital of Modena, Modena, Italy
| | - Maria Pia Brizzi
- Department of Oncology, "San Luigi Gonzaga" University Hospital, University of Turin, Orbassano, Italy
| | | | - Francesca Spada
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology (IEO) IRCCS, Milan, Italy
| | - Stefano Tamberi
- Medical Oncology Unit, "Degli Infermi" Hospital, Faenza, Italy
| | - Ivan Lolli
- Medical Oncology Unit, National Institute of Gastroenterology, IRCCS "Saverio De Bellis", Castellana Grotte, Italy
| | - Mauro Cives
- Department of Interdisciplinary Medicine, "Aldo Moro" University of Bari, Bari, Italy; Division of Medical Oncology, Azienda Ospedaliero Universitaria Consorziale Policlinico di Bari, Bari, Italy
| | - Riccardo Marconcini
- Medical Oncology Unit, Azienda Ospedaliero Universitaria Pisana, Pisa, Italy
| | - Francesca Pucci
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Rossana Berardi
- Department of Medical Oncology, Università Politecnica delle Marche, Azienda Ospedaliero Universitaria Ospedali Riuniti di Ancona, Ancona, Italy
| | - Lorenzo Antonuzzo
- Clinical Oncology Unit, Careggi University Hospital, Florence, Italy; Department of Experimental and Clinical Medicine, University of Florence, Italy
| | - Giuseppe Badalamenti
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Daniele Santini
- Department of Medical Oncology, University Campus Bio-Medico, Rome, Italy
| | - Federica Recine
- Medical Oncology Unit, Azienda Ospedaliera "San Giovanni Addolorata", Rome, Italy
| | - Silvia Vanni
- Bioscience Laboratory, Preclinic and Osteoncology Unit, 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
| | - Stefano Severi
- Nuclear Medicine and Radiometabolic Units, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Britt Rudnas
- Unit of Biostatistics and Clinical Trials, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Oriana Nanni
- Unit of Biostatistics and Clinical Trials, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Nicoletta Ranallo
- Osteoncology and Rare Tumor Center (CDO-TR), IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Laura Crudi
- Department of Oncology, University Hospital of Ferrara, Cona, Italy
| | - Luana Calabrò
- Oncology Pharmacy Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori", 47014 Meldola, Italy; Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Toni Ibrahim
- Osteoncology and Rare Tumor Center (CDO-TR), IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy; Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies Unit, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| |
Collapse
|
3
|
Partelli S, Landoni L, Bartolomei M, Zerbi A, Grana CM, Boggi U, Butturini G, Casadei R, Salvia R, Falconi M. Neoadjuvant 177Lu-DOTATATE for non-functioning pancreatic neuroendocrine tumours (NEOLUPANET): multicentre phase II study. Br J Surg 2024; 111:znae178. [PMID: 39213395 PMCID: PMC11364141 DOI: 10.1093/bjs/znae178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 06/23/2024] [Accepted: 07/02/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Resection of non-functioning pancreatic neuroendocrine tumours (NF-PanNETs) is curative in most patients. The potential benefits of neoadjuvant treatments have, however, never been explored. The primary aim of this study was to evaluate the safety of neoadjuvant 177Lu-labelled DOTA0-octreotate (177Lu-DOTATATE) followed by surgery in patients with NF-PanNETs. METHODS NEOLUPANET was a multicentre, single-arm, phase II trial of patients with sporadic, resectable or potentially resectable NF-PanNETs at high-risk of recurrence; those with positive 68Ga-labelled DOTA PET were eligible. All patients were candidates for neoadjuvant 177Lu-DOTATATE followed by surgery. A sample size of 30 patients was calculated to test postoperative complication rates against predefined cut-offs. The primary endpoint was safety, reflected by postoperative morbidity and mortality within 90 days. Secondary endpoints included rate of objective radiological response and quality of life. RESULTS From March 2020 to February 2023, 31 patients were enrolled, of whom 26 completed 4 cycles of 177Lu-DOTATATE. A partial radiological response was observed in 18 of 31 patients, and 13 patients had stable disease. Disease progression was not observed. Twenty-four R0 resections and 4 R1 resections were performed in 29 patients who underwent surgery. One tumour was unresectable owing to vascular involvement. There was no postoperative death. Postoperative complications occurred in 21 of 29 patients. Severe complications were observed in seven patients. Quality of life remained stable after 177Lu-DOTATATE and decreased after surgery. CONCLUSION Neoadjuvant treatment with 177Lu-DOTATATE is safe and effective for patients with NF-PanNETs.
Collapse
Affiliation(s)
- Stefano Partelli
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Pancreas Translational and Clinical Research Centre, Pancreatic and Transplant Surgery Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Luca Landoni
- General and Pancreatic Surgery Unit, Pancreas Institute, University of Verona Hospital Trust, Verona, Italy
| | - Mirco Bartolomei
- Nuclear Medicine Unit, Oncological Medical and Specialists Department, University Hospital of Ferrara, Ferrara, Italy
| | - Alessandro Zerbi
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- Pancreatic Surgery Unit, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Chiara Maria Grana
- Radiometabolic Therapy Unit, Division of Nuclear Medicine, IRCCS European Institute of Oncology, Milan, Italy
| | - Ugo Boggi
- Division of General and Transplant Surgery, University of Pisa, Pisa, Italy
| | | | - Riccardo Casadei
- Division of Pancreatic Surgery, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Internal Medicine and Surgery (DIMEC), Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Roberto Salvia
- General and Pancreatic Surgery Unit, Pancreas Institute, University of Verona Hospital Trust, Verona, Italy
| | - Massimo Falconi
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Pancreas Translational and Clinical Research Centre, Pancreatic and Transplant Surgery Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
4
|
Ursprung S, Zhang ML, Asmundo L, Hesami M, Najmi Z, Cañamaque LG, Shenoy-Bhangle AS, Pierce TT, Mojtahed A, Blake MA, Cochran R, Nikolau K, Harisinghani MG, Catalano OA. An Illustrated Review of the Recent 2019 World Health Organization Classification of Neuroendocrine Neoplasms: A Radiologic and Pathologic Correlation. J Comput Assist Tomogr 2024; 48:601-613. [PMID: 38438338 DOI: 10.1097/rct.0000000000001593] [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: 03/06/2024]
Abstract
ABSTRACT Recent advances in molecular pathology and an improved understanding of the etiology of neuroendocrine neoplasms (NENs) have given rise to an updated World Health Organization classification. Since gastroenteropancreatic NENs (GEP-NENs) are the most common forms of NENs and their incidence has been increasing constantly, they will be the focus of our attention. Here, we review the findings at the foundation of the new classification system, discuss how it impacts imaging research and radiological practice, and illustrate typical and atypical imaging and pathological findings. Gastroenteropancreatic NENs have a highly variable clinical course, which existing classification schemes based on proliferation rate were unable to fully capture. While well- and poorly differentiated NENs both express neuroendocrine markers, they are fundamentally different diseases, which may show similar proliferation rates. Genetic alterations specific to well-differentiated neuroendocrine tumors graded 1 to 3 and poorly differentiated neuroendocrine cancers of small cell and large-cell subtype have been identified. The new tumor classification places new demands and creates opportunities for radiologists to continue providing the clinically most relevant report and on researchers to design projects, which continue to be clinically applicable.
Collapse
Affiliation(s)
- Stephan Ursprung
- From the Department of Radiology, University Hospital Tuebingen, Tuebingen, Germany
| | - M Lisa Zhang
- Department of Pathology, Massachusetts General Hospital, Boston, MA
| | | | - Mina Hesami
- Department of Radiology, Massachusetts General Hospital, Boston, MA
| | - Zahra Najmi
- Department of Radiology, Massachusetts General Hospital, Boston, MA
| | | | | | | | | | - Michael A Blake
- Department of Radiology, Massachusetts General Hospital, Boston, MA
| | - Rory Cochran
- Department of Radiology, Massachusetts General Hospital, Boston, MA
| | - Konstantin Nikolau
- From the Department of Radiology, University Hospital Tuebingen, Tuebingen, Germany
| | | | | |
Collapse
|
5
|
Di Franco M, Zanoni L, Fortunati E, Fanti S, Ambrosini V. Radionuclide Theranostics in Neuroendocrine Neoplasms: An Update. Curr Oncol Rep 2024; 26:538-550. [PMID: 38581469 PMCID: PMC11063107 DOI: 10.1007/s11912-024-01526-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2024] [Indexed: 04/08/2024]
Abstract
PURPOSE OF REVIEW This paper aims to address the latest findings in neuroendocrine tumor (NET) theranostics, focusing on new evidence and future directions of combined diagnosis with positron emission tomography (PET) and treatment with peptide receptor radionuclide therapy (PRRT). RECENT FINDINGS Following NETTER-1 trial, PRRT with [177Lu]Lu-DOTATATE was approved by FDA and EMA and is routinely employed in advanced G1 and G2 SST (somatostatin receptor)-expressing NET. Different approaches have been proposed so far to improve the PRRT therapeutic index, encompassing re-treatment protocols, combinations with other therapies and novel indications. Molecular imaging holds a potential added value in characterizing disease biology and heterogeneity using different radiopharmaceuticals (e.g., SST and FDG) and may provide predictive and prognostic parameters. Response assessment criteria are still an unmet need and new theranostic pairs showed preliminary encouraging results. PRRT for NET has become a paradigm of modern theranostics. PRRT holds a favorable toxicity profile, and it is associated with a prolonged time to progression, reduction of symptoms, and improved patients' quality of life. In light of further optimization, different new strategies have been investigated, along with the development of new radiopharmaceuticals.
Collapse
Affiliation(s)
- Martina Di Franco
- Nuclear Medicine, Alma Mater Studiorum, University of Bologna, Via Massarenti 9, 40138, Bologna, Italy.
| | - Lucia Zanoni
- Nuclear Medicine, IRCCS, Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
| | - Emilia Fortunati
- Nuclear Medicine, IRCCS, Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
| | - Stefano Fanti
- Nuclear Medicine, Alma Mater Studiorum, University of Bologna, Via Massarenti 9, 40138, Bologna, Italy
- Nuclear Medicine, IRCCS, Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
| | - Valentina Ambrosini
- Nuclear Medicine, Alma Mater Studiorum, University of Bologna, Via Massarenti 9, 40138, Bologna, Italy
- Nuclear Medicine, IRCCS, Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
| |
Collapse
|
6
|
Davis CH, Laird AM, Libutti SK. Resistant gastroenteropancreatic neuroendocrine tumors: a definition and guideline to medical and surgical management. Proc AMIA Symp 2023; 37:104-110. [PMID: 38174011 PMCID: PMC10761146 DOI: 10.1080/08998280.2023.2284039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 11/07/2023] [Indexed: 01/05/2024] Open
Abstract
Gastroenteropancreatic neuroendocrine tumors (NETs), also historically known as carcinoids, are tumors derived of hormone-secreting enteroendocrine cells. Carcinoids may be found in the esophagus, stomach, small intestine, appendix, colon, rectum, or pancreas. The biologic behavior of carcinoids differs based on their location, with gastric and appendiceal NETs among the least aggressive and small intestinal and pancreatic NETs among the most aggressive. Ultimately, however, biologic behavior is most heavily influenced by tumor grade. The incidence of NETs has increased by 6.4 times over the past 40 years. Surgery remains the mainstay for management of most carcinoids. Medical management, however, is a useful adjunct and/or definitive therapy in patients with symptomatic functional carcinoids, in patients with unresectable or incompletely resected carcinoids, in some cases of recurrent carcinoid, and in postoperative patients to prevent recurrence. Functional tumors with persistent symptoms or progressive metastatic carcinoids despite therapy are called "resistant" tumors. In patients with unresectable disease and/or carcinoid syndrome, an array of medical therapies is available, mainly including somatostatin analogues, molecular-targeted therapy, and peptide receptor radionuclide therapy. Active research is ongoing to identify additional targeted therapies for patients with resistant carcinoids.
Collapse
Affiliation(s)
- Catherine H. Davis
- Division of Surgical Oncology, Baylor University Medical Center, Dallas, Texas, USA
- Texas A&M University School of Medicine, Dallas, Texas, USA
| | - Amanda M. Laird
- Division of Surgical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA
- Rutgers Robert Wood Johnson University Medical School, New Brunswick, New Jersey, USA
| | - Steven K. Libutti
- Division of Surgical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA
- Rutgers Robert Wood Johnson University Medical School, New Brunswick, New Jersey, USA
| |
Collapse
|
7
|
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: 3] [Impact Index Per Article: 3.0] [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
|
8
|
Alsadik S, Gnanasegaran G, Chen L, Quigley AM, Mandair D, Toumpanakis C, Caplin M, Navalkissoor S. Safety and Efficacy of 177Lu-DOTATATE in Neuroendocrine Tumor Patients With Extensive Bone Disease. Clin Nucl Med 2023:00003072-990000000-00548. [PMID: 37167406 DOI: 10.1097/rlu.0000000000004671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
AIM The aim of this study was to assess the efficacy and safety of 177Lu-DOTATATE in patients with neuroendocrine tumors (NETs) and extensive bone metastases, that is, more than 50% of the skeleton involved. METHOD A single-center retrospective analysis was performed in 30 patients (13 women and 17 men, mean age, 60 years; range, 35-77 years) undergoing 177Lu-DOTATATE therapy. Patients had progressive metastatic NETs with extensive skeletal metastases (>50% skeletal involvement seen on baseline 68Ga-DOTATATE PET/CT). The average administered activity was 7.308 (SD, 0.02) GBq per cycle with average treatment interval of 15 weeks. Survival analyses (progression-free survival [PFS], overall survival), radiological response assessment, toxicity assessment, and health-related quality of life (QoL) was performed. RESULTS Overall, 26 patients completed 4 cycles, and 4 patients had less than 4 cycles of 177Lu-DOTATATE. One patient (3%) did not complete treatment because of hematological toxicity. The estimated median PFS and median overall survival were calculated at 27 and 35 months, respectively. End-of-treatment radiological assessment showed partial response in 5 patients (17%), stable disease in 20 patients (66%), and radiological progressive disease in 3 patients (10%). Clinical progression was seen in a further 2 patients (7%).The incidence of grade 3/4 bone marrow toxicity was 10%. No patient had grade 3/4 peptide receptor radionuclide therapy-related nephrotoxicity. There was overall improvement in global QoL score (European Organization for Research and Treatment of Cancer Quality of Life Questionnaire-Gastrointestinal NET-21) (P = 0.019). CONCLUSION 177Lu-DOTATATE seems to have satisfactory therapeutic outcome in patients with advanced metastatic NET with extensive bone disease, with reasonable PFS and significant improvement in the global health-related QoL. The bone marrow toxicity was within the accepted range. Increasing the interval between cycles does not seem to reduce efficacy and may reduce toxicity, ensuring the bone marrow has sufficient time to recover between cycles.
Collapse
Affiliation(s)
| | | | | | | | - Dalvinder Mandair
- Neuroendocrine Tumour Unit, Royal Free London NHS Foundation Trust, London, England
| | - Christos Toumpanakis
- Neuroendocrine Tumour Unit, Royal Free London NHS Foundation Trust, London, England
| | - Martyn Caplin
- Neuroendocrine Tumour Unit, Royal Free London NHS Foundation Trust, London, England
| | | |
Collapse
|
9
|
Ladrière T, Faudemer J, Levigoureux E, Peyronnet D, Desmonts C, Vigne J. Safety and Therapeutic Optimization of Lutetium-177 Based Radiopharmaceuticals. Pharmaceutics 2023; 15:pharmaceutics15041240. [PMID: 37111725 PMCID: PMC10145759 DOI: 10.3390/pharmaceutics15041240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/24/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
Peptide receptor radionuclide therapy (PRRT) using Lutetium-177 (177Lu) based radiopharmaceuticals has emerged as a therapeutic area in the field of nuclear medicine and oncology, allowing for personalized medicine. Since the first market authorization in 2018 of [¹⁷⁷Lu]Lu-DOTATATE (Lutathera®) targeting somatostatin receptor type 2 in the treatment of gastroenteropancreatic neuroendocrine tumors, intensive research has led to transfer innovative 177Lu containing pharmaceuticals to the clinic. Recently, a second market authorization in the field was obtained for [¹⁷⁷Lu]Lu-PSMA-617 (Pluvicto®) in the treatment of prostate cancer. The efficacy of 177Lu radiopharmaceuticals are now quite well-reported and data on the safety and management of patients are needed. This review will focus on several clinically tested and reported tailored approaches to enhance the risk-benefit trade-off of radioligand therapy. The aim is to help clinicians and nuclear medicine staff set up safe and optimized procedures using the approved 177Lu based radiopharmaceuticals.
Collapse
Affiliation(s)
- Typhanie Ladrière
- Department of Nuclear Medicine, CHU de Caen Normandie, Normandie Université, UNICAEN, 14000 Caen, France
- Department of Pharmacy, CHU de Caen Normandie, Normandie Université, UNICAEN, 14000 Caen, France
| | - Julie Faudemer
- Department of Nuclear Medicine, CHU de Caen Normandie, Normandie Université, UNICAEN, 14000 Caen, France
| | - Elise Levigoureux
- Hospices Civils de Lyon, Groupement Hospitalier Est, 69677 Bron, France
- Lyon Neuroscience Research Center, CNRS UMR5292, INSERM U1028, Université Claude Bernard Lyon 1, 69677 Bron, France
| | - Damien Peyronnet
- Department of Nuclear Medicine, CHU de Caen Normandie, Normandie Université, UNICAEN, 14000 Caen, France
- Department of Pharmacy, CHU de Caen Normandie, Normandie Université, UNICAEN, 14000 Caen, France
| | - Cédric Desmonts
- Department of Nuclear Medicine, CHU de Caen Normandie, Normandie Université, UNICAEN, 14000 Caen, France
- INSERM U1086, ANTICIPE, Normandy University, UNICAEN, 14000 Caen, France
| | - Jonathan Vigne
- Department of Nuclear Medicine, CHU de Caen Normandie, Normandie Université, UNICAEN, 14000 Caen, France
- Department of Pharmacy, CHU de Caen Normandie, Normandie Université, UNICAEN, 14000 Caen, France
- PhIND, Centre Cyceron, Institut Blood and Brain @ Caen-Normandie, INSERM U1237, Normandie Université, UNICAEN, 14000 Caen, France
| |
Collapse
|
10
|
Sanli Y, Denizmen D, Subramaniam RM. Gastro-Enteric-Pancreatic Neuroendocrine Tumor Treatment: 177Lu-DOTATATE. PET Clin 2023; 18:201-214. [PMID: 36858745 DOI: 10.1016/j.cpet.2022.11.002] [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: 03/02/2023]
Abstract
177Lu-DOTA-TATE therapy is a highly effective therapy in metastatic, well-differentiated, somatostatin receptor-positive GEP-neuroendocrine tumors (NETs) with mostly tolerable adverse effects. Guidelines generally refer to peptide receptor radionuclide therapy as a second-line therapy after SSA in gastroenteric and second- or third-line therapy in pancreatic NETs to improve survival rates and quality of life. Although we do not have sufficient data, 177Lu-DOTA-TATE therapy may also have a role in high-grade NET therapy, mostly in combination with other treatments such as chemotherapy.
Collapse
Affiliation(s)
- Yasemin Sanli
- Department of Nuclear Medicine, Istanbul Faculty of Medicine, Istanbul University, Millet Caddesi, Istanbul 34390, Turkey.
| | - Dilara Denizmen
- Department of Nuclear Medicine, Istanbul Faculty of Medicine, Istanbul University, Millet Caddesi, Istanbul 34390, Turkey
| | - Rathan M Subramaniam
- Dean's Office, Otago Medical School, University of Otago, Dunedin 9016, New Zealand; Department of Radiology, Duke University, Durham, NC, USA
| |
Collapse
|
11
|
Rubira L, Deshayes E, Santoro L, Kotzki PO, Fersing C. 225Ac-Labeled Somatostatin Analogs in the Management of Neuroendocrine Tumors: From Radiochemistry to Clinic. Pharmaceutics 2023; 15:1051. [PMID: 37111537 PMCID: PMC10146019 DOI: 10.3390/pharmaceutics15041051] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/18/2023] [Accepted: 03/22/2023] [Indexed: 04/29/2023] Open
Abstract
The widespread use of peptide receptor radionuclide therapy (PRRT) represents a major therapeutic breakthrough in nuclear medicine, particularly since the introduction of 177Lu-radiolabeled somatostatin analogs. These radiopharmaceuticals have especially improved progression-free survival and quality of life in patients with inoperable metastatic gastroenteropancreatic neuroendocrine tumors expressing somatostatin receptors. In the case of aggressive or resistant disease, the use of somatostatin derivatives radiolabeled with an alpha-emitter could provide a promising alternative. Among the currently available alpha-emitting radioelements, actinium-225 has emerged as the most suitable candidate, especially regarding its physical and radiochemical properties. Nevertheless, preclinical and clinical studies on these radiopharmaceuticals are still few and heterogeneous, despite the growing momentum for their future use on a larger scale. In this context, this report provides a comprehensive and extensive overview of the development of 225Ac-labeled somatostatin analogs; particular emphasis is placed on the challenges associated with the production of 225Ac, its physical and radiochemical properties, as well as the place of 225Ac-DOTATOC and 225Ac-DOTATATE in the management of patients with advanced metastatic neuroendocrine tumors.
Collapse
Affiliation(s)
- Léa Rubira
- Nuclear Medicine Department, Institut Régional du Cancer de Montpellier (ICM), University Montpellier, 34090 Montpellier, France
| | - Emmanuel Deshayes
- Nuclear Medicine Department, Institut Régional du Cancer de Montpellier (ICM), University Montpellier, 34090 Montpellier, France
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, University Montpellier, Institut Régional du Cancer de Montpellier (ICM), 34298 Montpellier, France
| | - Lore Santoro
- Nuclear Medicine Department, Institut Régional du Cancer de Montpellier (ICM), University Montpellier, 34090 Montpellier, France
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, University Montpellier, Institut Régional du Cancer de Montpellier (ICM), 34298 Montpellier, France
| | - Pierre Olivier Kotzki
- Nuclear Medicine Department, Institut Régional du Cancer de Montpellier (ICM), University Montpellier, 34090 Montpellier, France
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, University Montpellier, Institut Régional du Cancer de Montpellier (ICM), 34298 Montpellier, France
| | - Cyril Fersing
- Nuclear Medicine Department, Institut Régional du Cancer de Montpellier (ICM), University Montpellier, 34090 Montpellier, France
- IBMM, University Montpellier, CNRS, ENSCM, 34293 Montpellier, France
| |
Collapse
|
12
|
Minczeles NS, de Herder WW, Feelders RA, Verburg FA, Hofland J, Brabander T. Long-Term Outcomes of Submaximal Activities of Peptide Receptor Radionuclide Therapy with 177Lu-DOTATATE in Neuroendocrine Tumor Patients. J Nucl Med 2023; 64:40-46. [PMID: 35680417 DOI: 10.2967/jnumed.122.263856] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 05/25/2022] [Accepted: 05/25/2022] [Indexed: 01/13/2023] Open
Abstract
In the literature, up to 45% of neuroendocrine tumor (NET) patients who are treated with 177Lu-DOTATATE peptide receptor radionuclide therapy (PRRT) do not receive the intended cumulative activity of 29.6 GBq (800 mCi). The aim of this study was to analyze the efficacy of submaximal activities of PRRT in patients who discontinued treatment for non-disease-related reasons. Methods: We retrospectively included patients with well-differentiated and advanced NETs who underwent PRRT from 2000 until 2019 and did not receive 29.6 GBq of 177Lu-DOTATATE. For comparison, we selected control NET patients who received the intended cumulative activity of 29.6 GBq of 177Lu-DOTATATE between 2000 and 2012. Primary outcomes were progression-free survival (PFS) and tumor response, and the secondary outcome was overall survival (OS). Results: In total, 243 patients received 3.7-27.8 GBq. In 130 patients, the submaximal activity was unrelated to disease (e.g., bone marrow and renal toxicity in 48% and maximal renal absorbed dose in 23%), and they were included. Patients receiving a reduced activity had more bone metastases, a lower body mass index and albumin level, a higher alkaline phosphatase level, and fewer grade 1 tumors than the 350 patients included in the control group. The disease control rate in the reduced-activity group was 85%, compared with 93% for the control group (P = 0.011). The median PFS (95% CI) was 23 mo (range, 21-26 mo) for the reduced-activity group and 31 mo (range, 27-35 mo) for the control group (P = 0.001), and the median OS (95% CI) was 34 mo (range, 28-40 mo) and 60 mo (range, 53-67 mo), respectively (P < 0.0001). With adjustment for relevant confounders in the multivariable Cox regression analyses, cumulative activity was an independent predictor of both PFS and OS. Conclusion: In NET patients treated with a cumulative activity of less than 29.6 GBq of 177Lu-DOTATATE, PRRT was less efficacious in tumor response and survival than in patients who received 29.6 GBq.
Collapse
Affiliation(s)
- Noémie S Minczeles
- Section of Endocrinology, Department of Internal Medicine, ENETS Centre of Excellence Rotterdam, Erasmus MC and Erasmus MC Cancer Centre, Rotterdam, The Netherlands; and .,Department of Radiology and Nuclear Medicine, ENETS Centre of Excellence Rotterdam, Erasmus MC and Erasmus MC Cancer Centre, Rotterdam, The Netherlands
| | - Wouter W de Herder
- Section of Endocrinology, Department of Internal Medicine, ENETS Centre of Excellence Rotterdam, Erasmus MC and Erasmus MC Cancer Centre, Rotterdam, The Netherlands; and
| | - Richard A Feelders
- Section of Endocrinology, Department of Internal Medicine, ENETS Centre of Excellence Rotterdam, Erasmus MC and Erasmus MC Cancer Centre, Rotterdam, The Netherlands; and
| | - Frederik A Verburg
- Department of Radiology and Nuclear Medicine, ENETS Centre of Excellence Rotterdam, Erasmus MC and Erasmus MC Cancer Centre, Rotterdam, The Netherlands
| | - Johannes Hofland
- Section of Endocrinology, Department of Internal Medicine, ENETS Centre of Excellence Rotterdam, Erasmus MC and Erasmus MC Cancer Centre, Rotterdam, The Netherlands; and
| | - Tessa Brabander
- Department of Radiology and Nuclear Medicine, ENETS Centre of Excellence Rotterdam, Erasmus MC and Erasmus MC Cancer Centre, Rotterdam, The Netherlands
| |
Collapse
|
13
|
177Lu-DOTATATE Efficacy and Safety in Functioning Neuroendocrine Tumors: A Joint Analysis of Phase II Prospective Clinical Trials. Cancers (Basel) 2022; 14:cancers14246022. [PMID: 36551507 PMCID: PMC9776442 DOI: 10.3390/cancers14246022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 10/31/2022] [Accepted: 11/25/2022] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION Neuroendocrine tumors (NETs) are rare malignancies with different prognoses. At least 25% of metastatic patients have functioning neuroendocrine tumors (F-NETs) that secrete bioactive peptides, causing specific debilitating and occasionally life-threatening symptoms such as diarrhea and flushing. Somatostatin analogs (SSAs) are usually effective but beyond them few treatment options are available. We evaluated the clinical efficacy of 177 Lu-DOTATATE in patients with progressive metastatic F-NETs and SSA-refractory syndrome. PATIENTS AND METHODS A non-pre-planned joint analysis was conducted in patients enrolled in phase II clinical trials on metastatic NETs. We extrapolated data from F-NET patients with ≥1 refractory sign/symptom to octreotide, and ≥1 measurable lesion. Syndrome response (SR), overall survival (OS), progression-free survival (PFS), tolerance and disease response were analyzed. RESULTS Sixty-eight patients were enrolled, the majority (88.1%) with a SR. According to RECIST criteria, 1 (1.5%) patient showed a CR, 21 (32.3%) had a PR and 40 (61.5%) SD. At a median follow-up of 28.9 months (range 2.2-63.2) median PFS was 33.0 months (95%CI: 27.1-48.2). Median OS (mOS) had not been reached at the time of the analysis; the 2-year OS was 87.8% (95%CI: 76.1-94.1). Syndromic responders showed better survival than non-responders, with a 2-year OS of 93.9% (95%CI: 92.2-98.0) vs. 40.0% (95%CI: 6.6-73.4), respectively. A total of 233 adverse events were recorded. Grade 1-2 hematological toxicity was the most frequent. CONCLUSION The 177 Lu-DOTATATE improved symptoms and disease control in patients with F-NETs. Treatment was well tolerated. The syndrome had an impact on both quality of life and OS.
Collapse
|
14
|
Prosperi D, Gentiloni Silveri G, Panzuto F, Faggiano A, Russo VM, Caruso D, Polici M, Lauri C, Filice A, Laghi A, Signore A. Nuclear Medicine and Radiological Imaging of Pancreatic Neuroendocrine Neoplasms: A Multidisciplinary Update. J Clin Med 2022; 11:jcm11226836. [PMID: 36431313 PMCID: PMC9694730 DOI: 10.3390/jcm11226836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/14/2022] [Accepted: 11/16/2022] [Indexed: 11/22/2022] Open
Abstract
Pancreatic neuroendocrine neoplasms (panNENs) are part of a large family of tumors arising from the neuroendocrine system. PanNENs show low-intermediate tumor grade and generally high somatostatin receptor (SSTR) expression. Therefore, panNENs benefit from functional imaging with 68Ga-somatostatin analogues (SSA) for diagnosis, staging, and treatment choice in parallel with morphological imaging. This narrative review aims to present conventional imaging techniques and new perspectives in the management of panNENs, providing the clinicians with useful insight for clinical practice. The 68Ga-SSA PET/CT is the most widely used in panNENs, not only fr diagnosis and staging purpose but also to characterize the biology of the tumor and its responsiveness to SSAs. On the contrary, the 18F-Fluordeoxiglucose (FDG) PET/CT is not employed systematically in all panNEN patients, being generally preferred in G2-G3, to predict aggressiveness and progression rate. The combination of 68Ga-SSA PET/CT and 18F-FDG PET/CT can finally suggest the best therapeutic strategy. Other radiopharmaceuticals are 68Ga-exendin-4 in case of insulinomas and 18F-dopamine (DOPA), which can be helpful in SSTR-negative tumors. New promising but still-under-investigation radiopharmaceuticals include radiolabeled SSTR antagonists and 18F-SSAs. Conventional imaging includes contrast enhanced CT and multiparametric MRI. There are now enriched by radiomics, a new non-invasive imaging approach, very promising to early predict tumor response or progression.
Collapse
Affiliation(s)
- Daniela Prosperi
- Nuclear Medicine Unit, Department of Medical-Surgical Sciences and of Translational Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, 00189 Roma, Italy
| | - Guido Gentiloni Silveri
- Nuclear Medicine Unit, Department of Medical-Surgical Sciences and of Translational Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, 00189 Roma, Italy
| | - Francesco Panzuto
- Digestive Disease Unit, Department of Medical-Surgical Sciences and Translational Medicine, Sant’Andrea University Hospital, ENETS Center of Excellence, Sapienza University of Rome, 00189 Roma, Italy
| | - Antongiulio Faggiano
- Endocrinology Unit, Department of Clinical and Molecular Medicine, Sant’Andrea University Hospital, ENETS Center of Excellence, Sapienza University of Rome, 00189 Roma, Italy
| | - Vincenzo Marcello Russo
- Nuclear Medicine Unit, Department of Medical-Surgical Sciences and of Translational Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, 00189 Roma, Italy
| | - Damiano Caruso
- Radiology Unit, Department of Medical Surgical Sciences and Translational Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, 00189 Roma, Italy
| | - Michela Polici
- Radiology Unit, Department of Medical Surgical Sciences and Translational Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, 00189 Roma, Italy
| | - Chiara Lauri
- Nuclear Medicine Unit, Department of Medical-Surgical Sciences and of Translational Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, 00189 Roma, Italy
- Correspondence:
| | - Angelina Filice
- Nucler Medicine Unit, AUSL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy
| | - Andrea Laghi
- Radiology Unit, Department of Medical Surgical Sciences and Translational Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, 00189 Roma, Italy
| | - Alberto Signore
- Nuclear Medicine Unit, Department of Medical-Surgical Sciences and of Translational Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, 00189 Roma, Italy
| |
Collapse
|
15
|
Alsadik S, Gnanasegaran G, Chen L, Quigley AM, Mandair D, Toumpanakis C, Caplin M, Navalkissoor S. Single centre retrospective review of outcome of 177 Lu-DOTATATE peptide receptor radionuclide therapy in the treatment of progressive metastatic neuroendocrine tumours: Survival, toxicity, and prognostic factors. J Neuroendocrinol 2022; 34:e13210. [PMID: 36399420 DOI: 10.1111/jne.13210] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 10/17/2022] [Accepted: 10/18/2022] [Indexed: 11/30/2022]
Abstract
The aim of this study was to evaluate the efficacy and safety of 177 Lu-DOTATATE therapy in advanced metastatic disease. A retrospective analysis of 395 patients (180 female, 215 males, mean age 62) with progressive metastatic neuroendocrine tumours (NETs) who were treated with 177 Lu-DOTATATE was performed. Overall, 115 patients had less than four cycles and 280 completed four cycles of treatment. Progression-free survival (PFS) and overall survival (OS) was performed using Kaplan-Meier analysis. Analysis of survival predictors was performed using Cox regression model. Toxicity was defined using the Common Terminology Criteria for Adverse Events version 5 (CTCAE 5.0). The percentage of patients with liver and skeletal metastases were 91 and 57%, respectively. Median PFS and OS were calculated at 33 months (95% CI: 29-37 months) and 46 months (95% CI: 48-56 months), respectively. End of treatment response assessment was performed using cross sectional imaging demonstrated partial response in 22%, stable disease in 64% and progressive disease in 14% of patients. Overall, grade 3 and 4 bone marrow toxicity was seen in 8%. One patient (0.3%) developed irreversible grade 4 nephrotoxicity. Myelodysplastic disease was recorded in one patient (0.3%). Univariate analysis of PFS predictors showed that body mass index (BMI), baseline chromogranin A (CgA) >400 ng/l, baseline alkaline phosphatase (ALP) >130 mg/dl, liver tumour volume and overall tumour burden were significant. On multivariate analysis only Ki67, high CgA and low BMI retained significance. 177 Lu-DOTATATE is an effective treatment in advanced NETs with generally high-volume metastases. It is well-tolerated. Ki-67, CgA and BMI appear to be predictors for PFS.
Collapse
Affiliation(s)
- Shahad Alsadik
- Department of Nuclear Medicine, Royal Free Hospital, London, UK
| | | | - Luohai Chen
- Royal Free London NHS Foundation Trust; Department of Gastroenterology, London, United Kingdom. The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ann-Marie Quigley
- Department of Nuclear Medicine, Royal Free London NHS foundation Trust, London, UK
| | - Dalvinder Mandair
- Neuroendocrine Unit, Royal Free London NHS foundation Trust, London, UK
| | | | - Martyn Caplin
- Neuroendocrine Unit, Royal Free London NHS foundation Trust, London, UK
| | - Shaunak Navalkissoor
- Department of Nuclear Medicine, Royal Free London NHS foundation Trust, London, UK
| |
Collapse
|
16
|
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: 1] [Impact Index Per Article: 0.5] [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
|
17
|
Bartolomei M, Berruti A, Falconi M, Fazio N, Ferone D, Lastoria S, Pappagallo G, Seregni E, Versari A. Clinical Management of Neuroendocrine Neoplasms in Clinical Practice: A Formal Consensus Exercise. Cancers (Basel) 2022; 14:cancers14102501. [PMID: 35626105 PMCID: PMC9140035 DOI: 10.3390/cancers14102501] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 05/16/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Well-structured international guidelines are currently available regarding the management of patients with neuroendocrine neoplasms (NENs). However, in relation to the multiplicity of treatments and the relative rarity and heterogeneity of NENs, there are many controversial issues in which clinical evidence is insufficient and for which expert opinion can be of help. A group of experts selected 14 relevant topics and formulated relative statements concerning controversial issues in several areas on diagnosis, prognosis, therapeutic strategies, and patient follow-up. Specific statements have also been formulated regarding patient management on radioligand therapy (RLT), as well as in the presence of co-morbidities or bone metastases. All the statements were drafted, discussed, modified, and then approved. The Nominal Group Technique (NGT) method was used to obtain consensus. The results of this paper can facilitate the clinical approach of patients with NENs in daily practice in areas where there is scarcity or absence of clinical evidence. Abstract Many treatment approaches are now available for neuroendocrine neoplasms (NENs). While several societies have issued guidelines for diagnosis and treatment of NENs, there are still areas of controversy for which there is limited guidance. Expert opinion can thus be of support where firm recommendations are lacking. A group of experts met to formulate 14 statements relative to diagnosis and treatment of NENs and presented herein. The nominal group and estimate-talk-estimate techniques were used. The statements covered a broad range of topics from tools for diagnosis to follow-up, evaluation of response, treatment efficacy, therapeutic sequence, and watchful waiting. Initial prognostic characterization should be based on clinical information as well as histopathological analysis and morphological and functional imaging. It is also crucial to optimize RLT for patients with a NEN starting from accurate characterization of the patient and disease. Follow-up should be patient/tumor tailored with a shared plan about timing and type of imaging procedures to use to avoid safety issues. It is also stressed that patient-reported outcomes should receive greater attention, and that a multidisciplinary approach should be mandatory. Due to the clinical heterogeneity and relative lack of definitive evidence for NENs, personalization of diagnostic–therapeutic work-up is crucial.
Collapse
Affiliation(s)
- Mirco Bartolomei
- Azienda Ospedaliero-Universitaria di Ferrara, Presidio Ospedaliero Arcispedale Sant’Anna di Cona, 44124 Ferrara, Italy;
| | - Alfredo Berruti
- Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, Medical Oncology, University of Brescia, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
- Correspondence:
| | - Massimo Falconi
- Pancreas Surgical Unit, ENETS Center of Excellence, San Raffaele Hospital IRCCS, Vita Salute University, 20132 Milan, Italy;
| | - Nicola Fazio
- Division of Gastrointestinal Medical Oncologya and Neuroendocrine Tumors, European Institute of Oncology, 20132 Milan, Italy;
| | - Diego Ferone
- Endocrinology Unit, Department of Internal Medicine and Medical Specialties, IRCCS, Ospedale Policlinico San Martino, Università di Genova, 16132 Genova, Italy;
| | - Secondo Lastoria
- Nuclear Medicine Unit, Istituto Nazionale Tumori, Fondazione G. Pascale, 80131 Naples, Italy;
| | - Giovanni Pappagallo
- School of Clinical Methodology IRCCS “Sacred Heart–Don Calabria” Hospital; 37024 Negrar di Valpolicella, Italy;
| | - Ettore Seregni
- Nuclear Medicine Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, 20132 Milano, Italy;
| | - Annibale Versari
- Nuclear Medicine Unit, Azienda Unità Sanitaria Locale-IRCCS of Reggio Emilia, 42100 Reggio Emilia, Italy;
| |
Collapse
|
18
|
Fortunati E, Argalia G, Zanoni L, Fanti S, Ambrosini V. New PET Radiotracers for the Imaging of Neuroendocrine Neoplasms. Curr Treat Options Oncol 2022; 23:703-720. [PMID: 35325412 PMCID: PMC9001579 DOI: 10.1007/s11864-022-00967-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/23/2022] [Indexed: 12/18/2022]
Abstract
OPINION STATEMENT Neuroendocrine neoplasms (NEN) are a heterogeneous group of tumours derived from cells of neuroendocrine origin and can potentially arise everywhere in the human body. The diagnostic assessment of NEN can be performed using a variety of PET radiopharmaceuticals. Well-differentiated NEN (NET) present a high expression of SSTR (somatostatin receptors) and can therefore be studied with 68Ga-DOTA-peptides ([68Ga]Ga-DOTANOC, [68Ga]Ga-DOTATOC, [68Ga]Ga-DOTATATE). Current guidelines recommend the use of SSTR imaging to assess disease extension at staging/restaging, follow-up, assessment of response to therapy and selection of patients who may benefit from radionuclide therapy (PRRT). [18F]F-FDG is used for the assessment of high-grade tumours (high-grade G2, G3 and NEC) and in every case, there is one or more mismatched lesions between diagnostic CT (positive) and SSTR-PET/CT (negative). [18F]F-DOPA is currently used for the assessment of medullary thyroid carcinoma, neuroblastoma, primary pheochromocytoma and abdominal paraganglioma. In recent years, however, several new tracers were designed exploiting the many potential targets of the neuroendocrine cell and were employed in clinical trials for both imaging and therapy. Currently, the real-life clinical impact of these tracers is still mostly not known; however, the favourable biodistribution (e.g. [68Ga]Ga-FAPI, SSTR antagonists) and the possibility to use new theranostic pairs may provide novel diagnostic as well as therapeutic options (e.g. [68Ga]Ga-PSMA, [64Cu]Cu-SARTATE, [68Ga]Ga-CXCR4) for NEN patients.
Collapse
Affiliation(s)
- Emilia Fortunati
- Nuclear Medicine, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy.
| | - Giulia Argalia
- Nuclear Medicine, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Lucia Zanoni
- Nuclear Medicine, IRCCS, Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
| | - Stefano Fanti
- Nuclear Medicine, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
- Nuclear Medicine, IRCCS, Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
| | - Valentina Ambrosini
- Nuclear Medicine, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
- Nuclear Medicine, IRCCS, Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
| |
Collapse
|
19
|
Dose-Limiting Bone Marrow Toxicities After Peptide Receptor Radionuclide Therapy Are More Prevalent in Women Than in Men. Clin Nucl Med 2022; 47:599-605. [DOI: 10.1097/rlu.0000000000004203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
20
|
Malcolm JC, Falzone N, Gains JE, Aldridge MD, Mirando D, Lee BQ, Gaze MN, Vallis KA. Impact of cyclic changes in pharmacokinetics and absorbed dose in pediatric neuroblastoma patients receiving [ 177Lu]Lu-DOTATATE. EJNMMI Phys 2022; 9:24. [PMID: 35347483 PMCID: PMC8960523 DOI: 10.1186/s40658-022-00436-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 01/24/2022] [Indexed: 11/10/2022] Open
Abstract
PURPOSE Recent reports personalizing the administered activity (AA) of each cycle of peptide receptor radionuclide therapy based on the predicted absorbed dose (AD) to the kidneys (dose-limiting organ) have been promising. Assuming identical renal pharmacokinetics for each cycle is pragmatic, however it may lead to over- or under-estimation of the optimal AA. Here, we investigate the influence that earlier cycles of [177Lu]Lu-DOTATATE had on the biokinetics and AD of subsequent cycles in a recent clinical trial that evaluated the safety and activity of [177Lu]Lu-DOTATATE in pediatric neuroblastoma (NBL). We investigated whether predictions based on an assumption of unchanging AD per unit AA (Gy/GBq) prove robust to cyclical changes in biokinetics. METHODS A simulation study, based on dosimetry data from six children with NBL who received four-cycles of [177Lu]Lu-DOTATATE in the LuDO trial (ISRCTN98918118), was performed to explore the effect of variable biokinetics on AD. In the LuDO trial, AA was adapted to the patient's weight and SPECT/CT-based dosimetry was performed for the kidneys and tumour after each cycle. The largest tumour mass was selected for dosimetric analysis in each case. RESULTS The median tumour AD per cycle was found to decrease from 15.6 Gy (range 8.12-26.4) in cycle 1 to 11.4 Gy (range 9.67-28.8), 11.3 Gy (range 2.73-32.9) and 4.3 Gy (range 0.72-20.1) in cycles 2, 3 and 4, respectively. By the fourth cycle, the median of the ratios of the delivered AD (ADD) and the predicted (or "expected") AD (ADE) (which was based on an assumption of stable biokinetics from the first cycle onwards) were 0.16 (range 0.02-0.92, p = 0.013) for the tumour and 1.08 (range 0.84-1.76, p > 0.05) for kidney. None of the patients had an objective response at 1 month follow up. CONCLUSION This study demonstrates variability in Gy/GBq and tumour AD per cycle in children receiving four administrations of [177Lu]Lu-DOTATATE treatment for NBL. NBL is deemed a radiation sensitive tumour; therefore, dose-adaptive treatment planning schemes may be appropriate for some patients to compensate for decreasing tumour uptake as treatment progresses. Trial registration ISRCTN ISRCTN98918118. Registered 20 December 2013 (retrospectively registered).
Collapse
Affiliation(s)
- Javian C Malcolm
- Oxford Institute for Radiation Oncology, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ, UK
| | - Nadia Falzone
- Oxford Institute for Radiation Oncology, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ, UK
| | - Jennifer E Gains
- Department of Oncology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Matthew D Aldridge
- Institute of Nuclear Medicine, University College London Hospitals NHS Foundation Trust, London, UK
| | | | - Boon Q Lee
- Oxford Institute for Radiation Oncology, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ, UK
| | - Mark N Gaze
- Department of Oncology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Katherine A Vallis
- Oxford Institute for Radiation Oncology, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ, UK.
| |
Collapse
|
21
|
Lee L, Ramos-Alvarez I, Jensen RT. Predictive Factors for Resistant Disease with Medical/Radiologic/Liver-Directed Anti-Tumor Treatments in Patients with Advanced Pancreatic Neuroendocrine Neoplasms: Recent Advances and Controversies. Cancers (Basel) 2022; 14:cancers14051250. [PMID: 35267558 PMCID: PMC8909561 DOI: 10.3390/cancers14051250] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/08/2022] [Accepted: 02/23/2022] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Tumor resistance, both primary and acquired, is leading to increased complexity in the nonsurgical treatment of patients with advanced panNENs, which would be greatly helped by reliable prognostic/predictive factors. The importance in identifying resistance is being contributed to by the increased array of possible treatments available for treating resistant advanced disease; the variable clinical course as well as response to any given treatment approach of patients within one staging or grading system, the advances in imaging which are providing increasing promising results/parameters that correlate with grading/outcome/resistance, the increased understanding of the molecular pathogenesis providing promising prognostic markers, all of which can contribute to selecting the best treatment to overcome resistance disease. Several factors have been identified that have prognostic/predictive value for identifying development resistant disease and affecting overall survival (OS)/PFS with various nonsurgical treatments of patients with advanced panNENs. Prognostic factors identified for patients with advanced panNENs for both OS/PFSs include various clinically-related factors (clinical, laboratory/biological markers, imaging, treatment-related factors), pathological factors (histological, classification, grading) and molecular factors. Particularly important prognostic factors for the different treatment modalities studies are the recent grading systems. Most prognostic factors for each treatment modality for OS/PFS are not specific for a given treatment option. These advances have generated several controversies and new unanswered questions, particularly those related to their possible role in predicting the possible sequence of different anti-tumor treatments in patients with different presentations. Each of these areas is reviewed in this paper. Abstract Purpose: Recent advances in the diagnosis, management and nonsurgical treatment of patients with advanced pancreatic neuroendocrine neoplasms (panNENs) have led to an emerging need for sensitive and useful prognostic factors for predicting responses/survival. Areas covered: The predictive value of a number of reported prognostic factors including clinically-related factors (clinical/laboratory/imaging/treatment-related factors), pathological factors (histological/classification/grading), and molecular factors, on therapeutic outcomes of anti-tumor medical therapies with molecular targeting agents (everolimus/sunitinib/somatostatin analogues), chemotherapy, radiological therapy with peptide receptor radionuclide therapy, or liver-directed therapies (embolization/chemoembolization/radio-embolization (SIRTs)) are reviewed. Recent findings in each of these areas, as well as remaining controversies and uncertainties, are discussed in detail, particularly from the viewpoint of treatment sequencing. Conclusions: The recent increase in the number of available therapeutic agents for the nonsurgical treatment of patients with advanced panNENs have raised the importance of prognostic factors predictive for therapeutic outcomes of each treatment option. The establishment of sensitive and useful prognostic markers will have a significant impact on optimal treatment selection, as well as in tailoring the therapeutic sequence, and for maximizing the survival benefit of each individual patient. In the paper, the progress in this area, as well as the controversies/uncertainties, are reviewed.
Collapse
Affiliation(s)
- Lingaku Lee
- Digestive Diseases Branch, NIDDK, NIH, Bethesda, MD 20892-1804, USA; (L.L.); (I.R.-A.)
- National Kyushu Cancer Center, Department of Hepato-Biliary-Pancreatology, Fukuoka 811-1395, Japan
| | - Irene Ramos-Alvarez
- Digestive Diseases Branch, NIDDK, NIH, Bethesda, MD 20892-1804, USA; (L.L.); (I.R.-A.)
| | - Robert T. Jensen
- Digestive Diseases Branch, NIDDK, NIH, Bethesda, MD 20892-1804, USA; (L.L.); (I.R.-A.)
- Correspondence: ; Tel.: +1-301-496-4201
| |
Collapse
|
22
|
Ambrosini V, Zanoni L, Filice A, Lamberti G, Argalia G, Fortunati E, Campana D, Versari A, Fanti S. Radiolabeled Somatostatin Analogues for Diagnosis and Treatment of Neuroendocrine Tumors. Cancers (Basel) 2022; 14:1055. [PMID: 35205805 PMCID: PMC8870358 DOI: 10.3390/cancers14041055] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/10/2022] [Accepted: 02/17/2022] [Indexed: 02/04/2023] Open
Abstract
Neuroendocrine neoplasms (NENs) are rare and heterogeneous tumors that require multidisciplinary discussion for optimal care. The theranostic approach (DOTA peptides labelled with 68Ga for diagnosis and with 90Y or 177Lu for therapy) plays a crucial role in the management of NENs to assess disease extension and as a criteria for peptide receptor radionuclide therapy (PRRT) eligibility based on somatostatin receptor (SSTR) expression. On the diagnostic side, [68Ga]Ga-DOTA peptides PET/CT (SSTR PET/CT) is the gold standard for imaging well-differentiated SSTR-expressing neuroendocrine tumors (NETs). [18F]FDG PET/CT is useful in higher grade NENs (NET G2 with Ki-67 > 10% and NET G3; NEC) for more accurate disease characterization and prognostication. Promising emerging radiopharmaceuticals include somatostatin analogues labelled with 18F (to overcome the limits imposed by 68Ga), and SSTR antagonists (for both diagnosis and therapy). On the therapeutic side, the evidence gathered over the past two decades indicates that PRRT is to be considered as an effective and safe treatment option for SSTR-expressing NETs, and is currently included in the therapeutic algorithms of the main scientific societies. The positioning of PRRT in the treatment sequence, as well as treatment personalization (e.g., tailored dosimetry, re-treatment, selection criteria, and combination with other alternative treatment options), is warranted in order to improve its efficacy while reducing toxicity. Although very preliminary (being mostly hampered by lack of methodological standardization, especially regarding feature selection/extraction) and often including small patient cohorts, radiomic studies in NETs are also presented. To date, the implementation of radiomics in clinical practice is still unclear. The purpose of this review is to offer an overview of radiolabeled SSTR analogues for theranostic use in NENs.
Collapse
Affiliation(s)
- Valentina Ambrosini
- Department of Experimental Diagnostic and Specialized Medicine, University of Bologna, 40138 Bologna, Italy; (V.A.); (G.L.); (G.A.); (E.F.); (D.C.); (S.F.)
- Nuclear Medicine Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Lucia Zanoni
- Nuclear Medicine Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Angelina Filice
- Nuclear Medicine Unit, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy; (A.F.); (A.V.)
| | - Giuseppe Lamberti
- Department of Experimental Diagnostic and Specialized Medicine, University of Bologna, 40138 Bologna, Italy; (V.A.); (G.L.); (G.A.); (E.F.); (D.C.); (S.F.)
- Division of Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Giulia Argalia
- Department of Experimental Diagnostic and Specialized Medicine, University of Bologna, 40138 Bologna, Italy; (V.A.); (G.L.); (G.A.); (E.F.); (D.C.); (S.F.)
| | - Emilia Fortunati
- Department of Experimental Diagnostic and Specialized Medicine, University of Bologna, 40138 Bologna, Italy; (V.A.); (G.L.); (G.A.); (E.F.); (D.C.); (S.F.)
| | - Davide Campana
- Department of Experimental Diagnostic and Specialized Medicine, University of Bologna, 40138 Bologna, Italy; (V.A.); (G.L.); (G.A.); (E.F.); (D.C.); (S.F.)
- Division of Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Annibale Versari
- Nuclear Medicine Unit, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy; (A.F.); (A.V.)
| | - Stefano Fanti
- Department of Experimental Diagnostic and Specialized Medicine, University of Bologna, 40138 Bologna, Italy; (V.A.); (G.L.); (G.A.); (E.F.); (D.C.); (S.F.)
- Nuclear Medicine Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| |
Collapse
|
23
|
Signore A, Prosperi D, Gentiloni G, Di Girolamo M, Lauri C, Filice A, Panzuto F. Therapy of NET with radiolabeled SST analogs. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00155-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
24
|
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: 17] [Impact Index Per Article: 5.7] [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
|
25
|
Park S, Parihar AS, Bodei L, Hope TA, Mallak N, Millo C, Prasad K, Wilson D, Zukotynski K, Mittra E. Somatostatin Receptor Imaging and Theranostics: Current Practice and Future Prospects. J Nucl Med 2021; 62:1323-1329. [PMID: 34301785 PMCID: PMC9364764 DOI: 10.2967/jnumed.120.251512] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 06/29/2021] [Indexed: 11/16/2022] Open
Abstract
A new era of precision diagnostics and therapy for patients with neuroendocrine neoplasms began with the approval of somatostatin receptor (SSTR) radiopharmaceuticals for PET imaging followed by peptide receptor radionuclide therapy (PRRT). With the transition from SSTR-based γ-scintigraphy to PET, the higher sensitivity of the latter raised questions regarding the direct application of the planar scintigraphy-based Krenning score for PRRT eligibility. Also, to date, the role of SSTR PET in response assessment and predicting outcome remains under evaluation. In this comprehensive review article, we discuss the current role of SSTR PET in all aspects of neuroendocrine neoplasms, including its relation to conventional imaging, selection of patients for PRRT, and the current understanding of SSTR PET-based response assessment. We also provide a standardized reporting template for SSTR PET with a brief discussion.
Collapse
Affiliation(s)
- Sonya Park
- Department of Nuclear Medicine, Seoul St. Mary's Hospital, Seoul, Korea
| | - Ashwin Singh Parihar
- Department of Nuclear Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Lisa Bodei
- Molecular Imaging and Therapy Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Thomas A Hope
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Nadine Mallak
- Department of Diagnostic Radiology, Oregon Health & Science University, Portland, Oregon
| | - Corina Millo
- Department of Nuclear Medicine, RAD&IS, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Kalpna Prasad
- Department of Nuclear Medicine, Walter Reed National Military Medical Center, Bethesda, Maryland
| | - Don Wilson
- BC Cancer, Vancouver, British Columbia, Canada
- Department of Radiology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Katherine Zukotynski
- Departments of Radiology and Medicine, McMaster University, Hamilton, Ontario, Canada
- Department of Radiology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Erik Mittra
- Department of Diagnostic Radiology, Oregon Health & Science University, Portland, Oregon;
| |
Collapse
|
26
|
Uccelli L, Boschi A, Cittanti C, Martini P, Panareo S, Tonini E, Nieri A, Urso L, Caracciolo M, Lodi L, Carnevale A, Giganti M, Bartolomei M. 90Y/ 177Lu-DOTATOC: From Preclinical Studies to Application in Humans. Pharmaceutics 2021; 13:1463. [PMID: 34575538 PMCID: PMC8469896 DOI: 10.3390/pharmaceutics13091463] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 08/31/2021] [Accepted: 09/10/2021] [Indexed: 12/30/2022] Open
Abstract
The PRRT (Peptide Receptor Radionuclide Therapy) is a promising modality treatment for patients with inoperable or metastatic neuroendocrine tumors (NETs). Progression-free survival (PFS) and overall survival (OS) of these patients are favorably comparable with standard therapies. The protagonist in this type of therapy is a somatostatin-modified peptide fragment ([Tyr3] octreotide), equipped with a specific chelating system (DOTA) capable of creating a stable bond with β-emitting radionuclides, such as yttrium-90 and lutetium-177. In this review, covering twenty five years of literature, we describe the characteristics and performances of the two most used therapeutic radiopharmaceuticals for the NETs radio-treatment: [90Y]Y-DOTATOC and [177Lu]Lu-DOTATOC taking this opportunity to retrace the most significant results that have determined their success, promoting them from preclinical studies to application in humans.
Collapse
Affiliation(s)
- Licia Uccelli
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (L.U.); (A.C.); (M.G.)
- Nuclear Medicine Unit, University Hospital, 44124 Ferrara, Italy; (S.P.); (A.N.); (L.U.); (M.C.); (L.L.); (M.B.)
| | - Alessandra Boschi
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy;
| | - Corrado Cittanti
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (L.U.); (A.C.); (M.G.)
- Nuclear Medicine Unit, University Hospital, 44124 Ferrara, Italy; (S.P.); (A.N.); (L.U.); (M.C.); (L.L.); (M.B.)
| | - Petra Martini
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (L.U.); (A.C.); (M.G.)
| | - Stefano Panareo
- Nuclear Medicine Unit, University Hospital, 44124 Ferrara, Italy; (S.P.); (A.N.); (L.U.); (M.C.); (L.L.); (M.B.)
| | - Eugenia Tonini
- Medical Physics Unit, University Hospital, 44124 Ferrara, Italy;
| | - Alberto Nieri
- Nuclear Medicine Unit, University Hospital, 44124 Ferrara, Italy; (S.P.); (A.N.); (L.U.); (M.C.); (L.L.); (M.B.)
| | - Luca Urso
- Nuclear Medicine Unit, University Hospital, 44124 Ferrara, Italy; (S.P.); (A.N.); (L.U.); (M.C.); (L.L.); (M.B.)
| | - Matteo Caracciolo
- Nuclear Medicine Unit, University Hospital, 44124 Ferrara, Italy; (S.P.); (A.N.); (L.U.); (M.C.); (L.L.); (M.B.)
| | - Luca Lodi
- Nuclear Medicine Unit, University Hospital, 44124 Ferrara, Italy; (S.P.); (A.N.); (L.U.); (M.C.); (L.L.); (M.B.)
| | - Aldo Carnevale
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (L.U.); (A.C.); (M.G.)
- Radiology Unit, University Hospital, 44124 Ferrara, Italy
| | - Melchiore Giganti
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (L.U.); (A.C.); (M.G.)
- Radiology Unit, University Hospital, 44124 Ferrara, Italy
| | - Mirco Bartolomei
- Nuclear Medicine Unit, University Hospital, 44124 Ferrara, Italy; (S.P.); (A.N.); (L.U.); (M.C.); (L.L.); (M.B.)
| |
Collapse
|
27
|
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: 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] [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
|
28
|
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: 3.0] [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
|
29
|
Han S, Lee HS, Woo S, Kim TH, Yoo C, Ryoo BY, Ryu JS. Prognostic Value of 18F-FDG PET in Neuroendocrine Neoplasm: A Systematic Review and Meta-analysis. Clin Nucl Med 2021; 46:723-731. [PMID: 34028406 DOI: 10.1097/rlu.0000000000003682] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVES Accurate assessment of the prognosis is critical for the rational treatment of neuroendocrine neoplasms (NENs). We performed a systematic review and meta-analysis of the prognostic value of 18F-FDG PET for NENs. PATIENTS AND METHODS PubMed and Embase databases were searched up to September 2020 for studies that evaluated 18F-FDG PET as prognostic factors in patients with NENs with overall survival (OS) and event-free survival (EFS) as outcomes. Hazards ratios (HRs) comparing high and low FDG uptakes were pooled using the DerSimonian-Laird method. Publication bias was assessed and adjusted for using the trim-and-fill method. Metaregression and subgroup analyses were performed to explore the cause of heterogeneity. RESULTS Twenty-three studies (1799 patients) were included. The overall pooled HRs of high FDG uptake on EFS and OS were 2.84 (95% confidence interval [CI], 2.21-3.64) and 3.50 (95% CI, 2.42-4.12), respectively. Publication biases were present regarding both EFS and OS (P = 0.0342 and 0.0009, respectively). After adjustment, effect sizes remained significant for EFS and OS (adjusted HR, 2.26 [95% CI, 1.76-2.89]; 3.16 [95% CI, 2.42-4.12]). In metaregression analyses, the proportion of grade 3 tumors positively correlated with the HR of OS (adjusted P = 0.0422). CONCLUSIONS 18F-FDG PET is a significant prognostic factor in patients with NENs. 18F-FDG PET might be a useful prognostic biomarker in conjunction with the histologic grade and can help select the optimal treatment.
Collapse
Affiliation(s)
- Sangwon Han
- From the Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul
| | - Hyo Sang Lee
- Department of Nuclear Medicine, GangNeung Asan Hospital, University of Ulsan College of Medicine, Gangneung, South Korea
| | - Sungmin Woo
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Changhoon Yoo
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Baek-Yeol Ryoo
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Jin-Sook Ryu
- From the Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul
| |
Collapse
|
30
|
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: 0] [Impact Index Per Article: 0] [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
|
31
|
Severi S, Bongiovanni A, Ferrara M, Nicolini S, Di Mauro F, Sansovini M, Lolli I, Tardelli E, Cittanti C, Di Iorio V, Mezzenga E, Scarpi E, Ibrahim T, Paganelli G, Zovato S. Peptide receptor radionuclide therapy in patients with metastatic progressive pheochromocytoma and paraganglioma: long-term toxicity, efficacy and prognostic biomarker data of phase II clinical trials. ESMO Open 2021; 6:100171. [PMID: 34139487 PMCID: PMC8219772 DOI: 10.1016/j.esmoop.2021.100171] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 04/22/2021] [Accepted: 05/08/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Pheochromocytoma and paraganglioma (PPGL) have currently only limited treatment options available for patients in the metastatic phase (mPPGL) in either post-surgery or inoperable settings. However, these rare tumors overexpress somatostatin receptors and can thus be treated with peptide receptor radionuclide therapy (PRRT). We present data about our 10-year experience treating 46 consecutive mPPGL patients with 90Y-DOTATOC or 177Lu-DOTATATE. PATIENTS AND METHODS All patients (20 men and 26 women, median age 52 years) showed positive scintigraphic imaging at 111In-octreotide or 68Ga-DOTATOC positron emission tomography/computed tomography (PET/CT). 90Y-DOTATOC was administered in 12 patients, with cumulative dosages ranging from 7.4 to 11 GBq, while 34 patients received 18.5 or 27.5GBq of 177Lu-DOTATATE. We used Southwest Oncology Group Response Evaluation Criteria in Solid Tumors criteria to evaluate treatment efficacy and Common Terminology Criteria for Adverse Events criteria to assess toxicity. The prognostic role of primary tumor site, hormone secretion, succinate dehydrogenase (SDHx) mutation, and metastatic involvement was also evaluated. RESULTS Both 90Y-DOTATOC and 177Lu-DOTATATE PRRT were well tolerated by patients without significant renal or bone marrow toxicity. The median follow-up was 73 months (range 5-146 months). The overall disease control rate (DCR) was 80% [95% confidence interval (CI) 68.9% to 91.9%] with a mean five cycles of therapy. However, 177Lu-DOTATATE patients showed a longer median overall survival (mOS) than those receiving 90Y-Dotatoc and a better DCR when higher dosages were administered, even if a direct comparison was not carried out. Syndromic patients had a poorer mOS. SDHx mutations did not interfere with treatment efficacy. CONCLUSIONS PRRT is safe and effective for the treatment of patients with progressive mPPGL, especially at higher dosages. The longer mOS of 177Lu-DOTATATE-treated patients in our protocols indicates the former radiopharmaceutical as the better candidate for further clinical application.
Collapse
Affiliation(s)
- S Severi
- Nuclear Medicine Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) 'Dino Amadori', Meldola, Italy
| | - A Bongiovanni
- Osteoncology and Rare Tumors Center (CDO-TR), IRCCS IRST 'Dino Amadori', Meldola, Italy
| | - M Ferrara
- Familial Cancer Clinic, Istituto Oncologico Veneto (IOV) IRCCS, Padua, Italy
| | - S Nicolini
- Nuclear Medicine Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) 'Dino Amadori', Meldola, Italy
| | - F Di Mauro
- Nuclear Medicine, Ospedale 'M. Bufalini', Cesena, Italy
| | - M Sansovini
- Nuclear Medicine Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) 'Dino Amadori', Meldola, Italy
| | - I Lolli
- Medical Oncology, Ospedale IRCCS 'S. De Bellis', Castellana Grotte, Italy
| | - E Tardelli
- Nuclear Medicine Unit, Department of Diagnostic Imaging, Ospedale 'San Luca', Lucca, Italy
| | - C Cittanti
- Nuclear Medicine Unit, University of Ferrara, Ferrara, Italy
| | - V Di Iorio
- Oncology Pharmacy, IRCCS IRST 'Dino Amadori', Meldola, Italy
| | - E Mezzenga
- Medical Physics Unit, IRCCS IRST 'Dino Amadori', Meldola, Italy
| | - E Scarpi
- Unit of Biostatistics and Clinical Trials, IRCCS IRST 'Dino Amadori', Meldola, Italy
| | - T Ibrahim
- Familial Cancer Clinic, Istituto Oncologico Veneto (IOV) IRCCS, Padua, Italy
| | - G Paganelli
- Nuclear Medicine Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) 'Dino Amadori', Meldola, Italy.
| | - S Zovato
- Familial Cancer Clinic, Istituto Oncologico Veneto (IOV) IRCCS, Padua, Italy
| |
Collapse
|
32
|
Gomes Marin JF, Nunes RF, Coutinho AM, Zaniboni EC, Costa LB, Barbosa FG, Queiroz MA, Cerri GG, Buchpiguel CA. Theranostics in Nuclear Medicine: Emerging and Re-emerging Integrated Imaging and Therapies in the Era of Precision Oncology. Radiographics 2021; 40:1715-1740. [PMID: 33001789 DOI: 10.1148/rg.2020200021] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Theranostics refers to the pairing of diagnostic biomarkers with therapeutic agents that share a specific target in diseased cells or tissues. Nuclear medicine, particularly with regard to applications in oncology, is currently one of the greatest components of the theranostic concept in clinical and research scenarios. Theranostics in nuclear medicine, or nuclear theranostics, refers to the use of radioactive compounds to image biologic phenomena by means of expression of specific disease targets such as cell surface receptors or membrane transporters, and then to use specifically designed agents to deliver ionizing radiation to the tissues that express these targets. The nuclear theranostic approach has sparked increasing interest and gained importance in parallel to the growth in molecular imaging and personalized medicine, helping to provide customized management for various diseases; improving patient selection, prediction of response and toxicity, and determination of prognosis; and avoiding futile and costly diagnostic examinations and treatment of many diseases. The authors provide an overview of theranostic approaches in nuclear medicine, starting with a review of the main concepts and unique features of nuclear theranostics and aided by a retrospective discussion of the progress of theranostic agents since early applications, with illustrative cases emphasizing the imaging features. Advanced concepts regarding the role of fluorine 18-fluorodeoxyglucose PET in theranostics, as well as developments in and future directions of theranostics, are discussed. ©RSNA, 2020 See discussion on this article by Greenspan and Jadvar.
Collapse
Affiliation(s)
- José Flávio Gomes Marin
- From the Department of Radiology, Hospital Sírio-Libanês, Rua Dona Adma Jafet 115, CEP 01308-060, São Paulo, SP, Brazil (J.F.G.M., R.F.N., A.M.C., E.C.Z., L.B.C., F.G.B., M.A.Q., G.G.C., C.A.B.); and Department of Radiology and Oncology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil (J.F.G.M., A.M.C., M.A.Q., G.G.C., C.A.B.)
| | - Rafael F Nunes
- From the Department of Radiology, Hospital Sírio-Libanês, Rua Dona Adma Jafet 115, CEP 01308-060, São Paulo, SP, Brazil (J.F.G.M., R.F.N., A.M.C., E.C.Z., L.B.C., F.G.B., M.A.Q., G.G.C., C.A.B.); and Department of Radiology and Oncology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil (J.F.G.M., A.M.C., M.A.Q., G.G.C., C.A.B.)
| | - Artur M Coutinho
- From the Department of Radiology, Hospital Sírio-Libanês, Rua Dona Adma Jafet 115, CEP 01308-060, São Paulo, SP, Brazil (J.F.G.M., R.F.N., A.M.C., E.C.Z., L.B.C., F.G.B., M.A.Q., G.G.C., C.A.B.); and Department of Radiology and Oncology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil (J.F.G.M., A.M.C., M.A.Q., G.G.C., C.A.B.)
| | - Elaine C Zaniboni
- From the Department of Radiology, Hospital Sírio-Libanês, Rua Dona Adma Jafet 115, CEP 01308-060, São Paulo, SP, Brazil (J.F.G.M., R.F.N., A.M.C., E.C.Z., L.B.C., F.G.B., M.A.Q., G.G.C., C.A.B.); and Department of Radiology and Oncology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil (J.F.G.M., A.M.C., M.A.Q., G.G.C., C.A.B.)
| | - Larissa B Costa
- From the Department of Radiology, Hospital Sírio-Libanês, Rua Dona Adma Jafet 115, CEP 01308-060, São Paulo, SP, Brazil (J.F.G.M., R.F.N., A.M.C., E.C.Z., L.B.C., F.G.B., M.A.Q., G.G.C., C.A.B.); and Department of Radiology and Oncology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil (J.F.G.M., A.M.C., M.A.Q., G.G.C., C.A.B.)
| | - Felipe G Barbosa
- From the Department of Radiology, Hospital Sírio-Libanês, Rua Dona Adma Jafet 115, CEP 01308-060, São Paulo, SP, Brazil (J.F.G.M., R.F.N., A.M.C., E.C.Z., L.B.C., F.G.B., M.A.Q., G.G.C., C.A.B.); and Department of Radiology and Oncology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil (J.F.G.M., A.M.C., M.A.Q., G.G.C., C.A.B.)
| | - Marcelo A Queiroz
- From the Department of Radiology, Hospital Sírio-Libanês, Rua Dona Adma Jafet 115, CEP 01308-060, São Paulo, SP, Brazil (J.F.G.M., R.F.N., A.M.C., E.C.Z., L.B.C., F.G.B., M.A.Q., G.G.C., C.A.B.); and Department of Radiology and Oncology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil (J.F.G.M., A.M.C., M.A.Q., G.G.C., C.A.B.)
| | - Giovanni G Cerri
- From the Department of Radiology, Hospital Sírio-Libanês, Rua Dona Adma Jafet 115, CEP 01308-060, São Paulo, SP, Brazil (J.F.G.M., R.F.N., A.M.C., E.C.Z., L.B.C., F.G.B., M.A.Q., G.G.C., C.A.B.); and Department of Radiology and Oncology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil (J.F.G.M., A.M.C., M.A.Q., G.G.C., C.A.B.)
| | - Carlos A Buchpiguel
- From the Department of Radiology, Hospital Sírio-Libanês, Rua Dona Adma Jafet 115, CEP 01308-060, São Paulo, SP, Brazil (J.F.G.M., R.F.N., A.M.C., E.C.Z., L.B.C., F.G.B., M.A.Q., G.G.C., C.A.B.); and Department of Radiology and Oncology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil (J.F.G.M., A.M.C., M.A.Q., G.G.C., C.A.B.)
| |
Collapse
|
33
|
Assi HA, Hornbacker K, Shaheen S, Wittenberg T, Silberman R, Kunz PL. Rapid Progression After 177Lu-DOTATATE in Patients With Neuroendocrine Tumors. Pancreas 2021; 50:890-894. [PMID: 34398071 DOI: 10.1097/mpa.0000000000001841] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
ABSTRACT Peptide receptor radionuclide therapy (PRRT) is a treatment option for somatostatin receptor-positive, unresectable or metastatic neuroendocrine tumors (NETs). Despite high disease control rates seen with PRRT, a subset of the NET population seems to have a short progression-free interval. We hypothesize that patients with NETs with rapid progression post-PRRT may have mixed low- and high-grade cell populations, and PRRT treats the lower-grade component, allowing the more aggressive high-grade component to progress.We report 7 patients with biopsy-proven NET who received PRRT with 177Lu-DOTATATE at the Stanford Cancer Center who had evidence of progressive disease (PD) on or within 6 months of therapy.All patients had primary pancreatic, metastatic, well-differentiated NET on diagnosis and were heavily pretreated before receiving PRRT. Two patients had PD while on PRRT; 5 had PD within 6 months of completing PRRT. The median time from the last cycle to PD was 3.2 months (range, 1.1-4.6 months). The median progression-free survival was 7.7 months (95% confidence interval, 5.7-9.8 months). Three patients had a repeat biopsy post-PRRT, 2 of which demonstrated higher disease grade compared with their initial pathology. Further evaluation in larger patient cohorts is warranted to elucidate predictive factors of PRRT response/nonresponse to enable better patient selection.
Collapse
Affiliation(s)
- Hussein A Assi
- From the Section of Hematology/Oncology, Department of Medicine, Boston University School of Medicine, Boston, MA
| | - Kathleen Hornbacker
- Section of Medical Oncology, Department of Medicine, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | - Shagufta Shaheen
- Section of Medical Oncology, Department of Medicine, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | - Theresa Wittenberg
- Section of Medical Oncology, Department of Medicine, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | - Robyn Silberman
- Section of Medical Oncology, Department of Medicine, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | - Pamela L Kunz
- Section of Medical Oncology, Department of Medicine, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| |
Collapse
|
34
|
Pirasteh A, Lovrec P, Pedrosa I. Imaging and its Impact on Defining the Oligometastatic State. Semin Radiat Oncol 2021; 31:186-199. [PMID: 34090645 DOI: 10.1016/j.semradonc.2021.03.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Successful treatment of oligometastatic disease (OMD) is facilitated through timely detection and localization of disease, both at the time of initial diagnosis (synchronous OMD) and following the initial therapy (metachronous OMD). Hence, imaging plays an indispensable role in management of patients with OMD. However, the challenges and complexities of OMD management are also reflected in the imaging of this entity. While innovations and advances in imaging technology have made a tremendous impact in disease detection and management, there remain substantial and unaddressed challenges for earlier and more accurate establishment of OMD state. This review will provide an overview of the available imaging modalities and their inherent strengths and weaknesses, with a focus on their role and potential in detection and evaluation of OMD in different organ systems. Furthermore, we will review the role of imaging in evaluation of OMD for malignancies of various primary organs, such as the lung, prostate, colon/rectum, breast, kidney, as well as neuroendocrine tumors and gynecologic malignancies. We aim to provide a practical overview about the utilization of imaging for clinicians who play a role in the care of those with, or at risk for OMD.
Collapse
Affiliation(s)
- Ali Pirasteh
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI
| | - Petra Lovrec
- Department of Radiology, University of Wisconsin-Madison, Madison, WI
| | - Ivan Pedrosa
- Departments of Radiology, Urology, and Advanced Imaging Research Center. University of Texas Southwestern, Dallas, TX.
| |
Collapse
|
35
|
Satapathy S, Bhattacharya A, Sood A, Kapoor R, Gupta R, Sood A, Sharma P, Khosla D, Mittal BR. Hematological Markers as Predictors of Treatment Outcomes with Lu-177 DOTATATE in Patients with Advanced Neuroendocrine Tumors. Cancer Biother Radiopharm 2021; 37:23-29. [PMID: 34185573 DOI: 10.1089/cbr.2021.0053] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background: Chronic inflammation has been linked to the development and prognosis of neuroendocrine tumors (NETs). The current study intended to evaluate the role of peripheral hematological inflammatory markers, viz. the platelet-lymphocyte ratio (PLR), neutrophil-lymphocyte ratio, and monocyte-lymphocyte ratio, as predictors of treatment outcomes in patients with advanced NETs after Lu-177 DOTATATE therapy. Materials and Methods: Data of consecutive patients with advanced metastatic and/or inoperable NETs treated with Lu-177 DOTATATE from the year 2012 to 2019 at the authors' center were retrospectively analyzed. Results: Forty-two NET patients (median age: 49.5 years) received a median cumulative activity of 29.6 GBq of Lu-177 DOTATATE over 2-5 cycles at 8-12-week intervals. The median progression-free survival (PFS) of the study cohort was 30 months (95% confidence interval, CI: 18.2-41.9 months). A baseline PLR ≥173.1 was found to be a significant predictor of poor PFS with a univariate hazard ratio of 3.82 (95% CI: 1.21-12.03); however, the association was not significant on multivariate analysis. The median overall survival was not reached and none of the parameters were significantly associated with it. Conclusions: A higher baseline PLR was shown to be associated with a negative outcome on PFS after 177Lu-DOTATATE therapy and is a promising marker for future larger studies.
Collapse
Affiliation(s)
- Swayamjeet Satapathy
- Department of Nuclear Medicine, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Anish Bhattacharya
- Department of Nuclear Medicine, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Ashwani Sood
- Department of Nuclear Medicine, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Rakesh Kapoor
- Department of Radiotherapy, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Rajesh Gupta
- Department of Surgical Gastroenterology, and Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Apurva Sood
- Department of Nuclear Medicine, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Prashant Sharma
- Department of Hematology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Divya Khosla
- Department of Radiotherapy, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Bhagwant Rai Mittal
- Department of Nuclear Medicine, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
36
|
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]
|
37
|
Nunes RF, Zuppani RMF, Coutinho AM, Barbosa FG, Sapienza MT, Marin JFG, Buchpiguel CA. General Concepts in Theranostics. PET Clin 2021; 16:313-326. [PMID: 34053576 DOI: 10.1016/j.cpet.2021.03.010] [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] [Indexed: 12/23/2022]
Abstract
Theranostics describes the pairing of diagnostic biomarkers and therapeutic agents with common specific targets. Nuclear medicine is the greatest theranostics protagonist, relying on radioactive tracers for imaging biologic phenomena and delivering ionizing radiation to the tissues that take up those tracers. The concept has gained importance with the growth of personalized medicine, allowing customized management for diseases, refining patient selection, better predicting responses, reducing toxicity, and estimating prognosis. This work provides an overview of the general concepts of the theranostics approach in nuclear medicine discussing its background, features, and future directions in imaging and therapy.
Collapse
Affiliation(s)
- Rafael F Nunes
- Department of Radiology, Hospital Sirio-Libanes, Sao Paulo, Brazil.
| | - Roberta M F Zuppani
- Department of Radiology and Oncology, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Artur M Coutinho
- Department of Radiology, Hospital Sirio-Libanes, Sao Paulo, Brazil; Department of Radiology and Oncology, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Felipe G Barbosa
- Department of Radiology, Hospital Sirio-Libanes, Sao Paulo, Brazil
| | - Marcelo T Sapienza
- Department of Radiology and Oncology, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Jose Flavio G Marin
- Department of Radiology, Hospital Sirio-Libanes, Sao Paulo, Brazil; Department of Radiology and Oncology, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Carlos A Buchpiguel
- Department of Radiology, Hospital Sirio-Libanes, Sao Paulo, Brazil; Department of Radiology and Oncology, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
38
|
Rodriguez-Freixinos V, Thawer A, Capdevila J, Ferone D, Singh S. Advanced Pancreatic Neuroendocrine Neoplasms: Which Systemic Treatment Should I Start With? Curr Oncol Rep 2021; 23:80. [PMID: 33937962 DOI: 10.1007/s11912-021-01071-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/31/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE OF REVIEW Pancreatic neuroendocrine neoplasms (panNENs) often present as advanced disease and there is little data to guide treatment sequencing in the advance disease setting. Therefore, we aim to provide a comprehensive summary of the current evidence supporting the use of systemic treatment for patients with diagnosis of advanced and metastatic panNENs, as well as to provide strategies for treatment selection and address challenges for treatment selection and sequencing of therapy. RECENT FINDINGS Substantial advances have been made and many clinical trials have been performed over the past two decades expanding therapeutic options available for patients with advanced panNETs. Available systemic treatments for patients with well-differentiated pancreatic neuroendocrine tumors include somatostatin receptors ligands (SRLs), traditional cytotoxic chemotherapy regimens, peptide receptor radiotherapy (PRRT), and biologically targeted therapies, whereas patients with poorly differentiated neurodocrine carcinomas have more limited treatment options. Despite these advances, no clear guidelines exist to support the best sequence of treatments, not only the first-line, but also subsequent lines of therapy in patients with panNENs. Advances in molecular research and discovery of biomarkers for response allowing a more personalized approach to the multimodality therapy of panNENs are still limited. Understanding the impact of previous therapies on subsequent treatment efficacy and toxicity is also an ongoing research question. In the absence of definite predictive markers and paucity of comparative randomized trials, along with the heterogeneity of this patient population, systemic therapy selection in advanced non-resectable disease should be patient centered and often require evaluation within a multidisciplinary setting. The specific clinical context of the patient, with assessment of individual patient clinical and pathological features, somatostatin receptors imaging, and goals of treatment must all be considered when deciding on systemic therapy in the patient. Additional research is needed to address the gap in knowledge regarding optimal sequencing and timing of therapies and to provide individual care.
Collapse
Affiliation(s)
- Victor Rodriguez-Freixinos
- Department of Medical Oncology and Hematology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, Toronto, ON, M4N 3M5, Canada
| | - Alia Thawer
- Department of Pharmacy, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, Canada
| | - Jaume Capdevila
- Department of Medical Oncology, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology (VIHO), Barcelona, Spain
| | - Diego Ferone
- Endocrinology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy.,Endocrinology, Department of Internal Medicine and Medical Specialties (DIMI) and Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| | - Simron Singh
- Department of Medical Oncology and Hematology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, Toronto, ON, M4N 3M5, Canada.
| |
Collapse
|
39
|
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: 7] [Impact Index Per Article: 2.3] [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
|
40
|
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: 35] [Impact Index Per Article: 11.7] [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
|
41
|
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
|
42
|
Fernandez CJ, Agarwal M, Pottakkat B, Haroon NN, George AS, Pappachan JM. Gastroenteropancreatic neuroendocrine neoplasms: A clinical snapshot. World J Gastrointest Surg 2021; 13:231-255. [PMID: 33796213 PMCID: PMC7993001 DOI: 10.4240/wjgs.v13.i3.231] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/17/2021] [Accepted: 03/02/2021] [Indexed: 02/06/2023] Open
Abstract
Our understanding about the epidemiological aspects, pathogenesis, molecular diagnosis, and targeted therapies of neuroendocrine neoplasms (NENs) have drastically advanced in the past decade. Gastroenteropancreatic (GEP) NENs originate from the enteroendocrine cells of the embryonic gut which share common endocrine and neural differentiation factors. Most NENs are well-differentiated, and slow growing. Specific neuroendocrine biomarkers that are used in the diagnosis of functional NENs include insulin, glucagon, vasoactive intestinal polypeptide, gastrin, somatostatin, adrenocorticotropin, growth hormone releasing hormone, parathyroid hormone-related peptide, serotonin, histamine, and 5-hydroxy indole acetic acid (5-HIAA). Biomarkers such as pancreatic polypeptide, human chorionic gonadotrophin subunits, neurotensin, ghrelin, and calcitonin are used in the diagnosis of non-functional NENs. 5-HIAA levels correlate with tumour burden, prognosis and development of carcinoid heart disease and mesenteric fibrosis, however several diseases, medications and edible products can falsely elevate the 5-HIAA levels. Organ-specific transcription factors are useful in the differential diagnosis of metastasis from an unknown primary of well-differentiated NENs. Emerging novel biomarkers include circulating tumour cells, circulating tumour DNA, circulating micro-RNAs, and neuroendocrine neoplasms test (NETest) (simultaneous measurement of 51 neuroendocrine-specific marker genes in the peripheral blood). NETest has high sensitivity (85%-98%) and specificity (93%-97%) for the detection of gastrointestinal NENs, and is useful for monitoring treatment response, recurrence, and prognosis. In terms of management, surgery, radiofrequency ablation, symptom control with medications, chemotherapy and molecular targeted therapies are all considered as options. Surgery is the mainstay of treatment, but depends on factors including age of the individual, location, stage, grade, functional status, and the heredity of the tumour (sporadic vs inherited). Medical management is helpful to alleviate the symptoms, manage inoperable lesions, suppress postoperative tumour growth, and manage recurrences. Several molecular-targeted therapies are considered second line to somatostatin analogues. This review is a clinical update on the pathophysiological aspects, diagnostic algorithm, and management of GEP NENs.
Collapse
Affiliation(s)
- Cornelius J Fernandez
- Department of Endocrinology and Metabolism, Pilgrim Hospital, United Lincolnshire Hospitals NHS Trust, Boston PE21 9QS, United Kingdom
| | - Mayuri Agarwal
- Department of Endocrinology and Metabolism, Pilgrim Hospital, United Lincolnshire Hospitals NHS Trust, Boston PE21 9QS, United Kingdom
| | - Biju Pottakkat
- Department of Surgical Gastroenterology, Jawaharlal Institute of Post Graduate Medical Education and Research (JIPMER), Puducherry 605006, India
| | - Nisha Nigil Haroon
- Department of Endocrinology and Internal Medicine, Northern Ontario School of Medicine, Sudbury P3E 2C6, Ontario, Canada
| | - Annu Susan George
- Department of Medical Oncology, VPS Lakeshore Hospital, Cochin 682040, Kerala, India
| | - Joseph M Pappachan
- Department of Endocrinology and Metabolism, Lancashire Teaching Hospitals NHS Trust, PR2 9HT, Preston, The University of Manchester, Oxford Road M13 9PL, Manchester Metropolitan University, All Saints Building M15 6BH, Manchester, United Kingdom
| |
Collapse
|
43
|
Nephrotoxicity/renal failure after therapy with 90Yttrium- and 177Lutetium-radiolabeled somatostatin analogs in different types of neuroendocrine tumors: a systematic review. Nucl Med Commun 2021; 41:601-617. [PMID: 32404645 DOI: 10.1097/mnm.0000000000001198] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND/OBJECTIVE Data regarding the nephrotoxicity of the peptide receptor radionuclide therapy (PRRT) with Yttrium- and Lutetium-radiolabeled somatostatin analogs (RSA) are inconclusive. We aimed to evaluate the short- and long-term nephrotoxicity following PRRT usage in patients with all types of neuroendocrine tumors (NETs). METHODS A systematic review of observational studies reporting data about nephrotoxicity after treatment with Yttrium and Lutetium RSA was performed. Data on serum creatinine, creatinine clearance, glomerular filtration rate (GFR) and need for renal replacement therapy were compiled. We included patients with progressive, inoperable symptomatic G1, G2 and G3 different types of NETs. After searching in three electronic databases PubMed, Scopus and the Cochrane Library, from 1 January 1978 to November 2018, data were extracted and summarized using a random-effects model. RESULTS The final analysis included 34 studies, comprising 5386 participants, enrolling patients with G1, G2, G3 NETs and a follow-up from 12 up to 191 months. Compared with renal function before treatment, measured/estimated glomerular filtration rate (m/eGFR) values changed after PRRT, with a mean annual decrease following PRRT between 2 and 4 mL/min/1.73 m suggesting different grades of nephrotoxicity after PRRT. When compared, Y-RSA and the Y-RSA-Lu-RSA combination are associated with a higher m/eGFR decline compared to Lu-RSA alone. CONCLUSIONS PRRT can be followed by potentially serious long-term nephrotoxicity, despite kidney protection. The use of the quantified renal function combined with a long follow-up period and personalized dosimetry-based PRRT can reduce nephrotoxicity, in order to use the whole PRRT potential in the management of NETs.
Collapse
|
44
|
Indications of Peptide Receptor Radionuclide Therapy (PRRT) in Gastroenteropancreatic and Pulmonary Neuroendocrine Tumors: An Updated Review. J Clin Med 2021; 10:jcm10061267. [PMID: 33803817 PMCID: PMC8003169 DOI: 10.3390/jcm10061267] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/10/2021] [Accepted: 03/12/2021] [Indexed: 12/14/2022] Open
Abstract
Radionuclide therapy for neuroendocrine tumors is a form of systemic radiotherapy that allows the administration of targeted radionuclides into tumor cells that express a large quantity of somatostatin receptors. The two most commonly used radio-peptides for radionuclide therapy in neuroendocrine tumors are 90Y-DOTATOC and 177Lu-DOTATATE. Radio-peptides have been used for several years in the treatment of advanced neuroendocrine tumors. Recently, the randomized Phase III study NETTER-1 compared177Lu-DOTATATE versus high-dose (double-dose) octreotide LAR in patients with metastatic midgut neuroendocrine tumors, and demonstrated its efficacy in this setting. Strong signals in favor of efficiency seem to exist for other tumors, in particular for pancreatic and pulmonary neuroendocrine tumors. This focus on radionuclide therapy in gastroenteropancreatic and pulmonary neuroendocrine tumors addresses the treatment modalities, the validated and potential indications, and the safety of the therapy.
Collapse
|
45
|
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: 26] [Impact Index Per Article: 8.7] [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
|
46
|
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: 132] [Impact Index Per Article: 44.0] [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
|
47
|
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
|
48
|
Hayes AR, Mak IYF, Evans N, Naik R, Crawford A, Khoo B, Grossman AB, Navalkissoor S, Watkins J, Luong TV, Mandair D, Toumpanakis C, Thirlwell C, Caplin ME, Meyer T. Understanding the Treatment Algorithm of Patients with Metastatic Pancreatic Neuroendocrine Neoplasms: A Single-Institution Retrospective Analysis Comparing Outcomes of Chemotherapy, Molecular Targeted Therapy, and Peptide Receptor Radionuclide Therapy in 255 Patients. Neuroendocrinology 2021; 111:863-875. [PMID: 32950978 DOI: 10.1159/000511662] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 08/31/2020] [Indexed: 11/19/2022]
Abstract
BACKGROUND The number of therapeutic options for patients with pancreatic neuroendocrine neoplasms (PNEN) has increased, but the optimal therapeutic algorithm has not been defined due to lack of randomised trials comparing different modalities. METHODS We performed a retrospective study in patients with metastatic PNEN treated with ≥1 line of systemic therapy. The relationship between baseline characteristics, treatment type, and time to treatment failure (TTF), time to progression (TTP), and overall survival (OS) was analysed using the Kaplan-Meier method. Univariate and multivariate analyses were performed using the Cox proportional hazards model. RESULTS Two hundred and fifty-five patients with metastatic PNEN had 491 evaluable lines of therapy. Independent predictors of TTF included treatment type, Ki-67, tumour grade, and chromogranin A. To reduce selection bias, a subgroup of 114 patients with grade 2 (G2) metastatic pancreatic neuroendocrine tumours (PNET) was analysed separately. These patients had received 234 lines of treatment (105 chemotherapy, 82 molecular targeted therapy, and 47 peptide receptor radionuclide therapy [PRRT]). In the G2 cohort, TTF and TTP were superior for PRRT compared with both chemotherapy and molecular targeted therapy. OS in the G2 cohort was also superior for those that had received PRRT compared with those that had not (median 84 vs. 56 months; HR 0.55, 95% CI: 0.31-0.98, p = 0.04). CONCLUSIONS This study suggests that PRRT is associated with superior clinical outcomes relative to other systemic therapies for G2 metastatic PNET. Prospective studies are required to confirm these observations.
Collapse
Affiliation(s)
- Aimee R Hayes
- Neuroendocrine Tumour Unit, ENETS Centre of Excellence, Royal Free Hospital, London, United Kingdom,
- Department of Oncology, Royal Free Hospital, London, United Kingdom,
| | - Ingrid Y F Mak
- Neuroendocrine Tumour Unit, ENETS Centre of Excellence, Royal Free Hospital, London, United Kingdom
| | - Nicholas Evans
- Medical School, University College London, London, United Kingdom
| | - Rishi Naik
- Medical School, University College London, London, United Kingdom
| | | | - Bernard Khoo
- Neuroendocrine Tumour Unit, ENETS Centre of Excellence, Royal Free Hospital, London, United Kingdom
| | - Ashley B Grossman
- Neuroendocrine Tumour Unit, ENETS Centre of Excellence, Royal Free Hospital, London, United Kingdom
| | - Shaunak Navalkissoor
- Neuroendocrine Tumour Unit, ENETS Centre of Excellence, Royal Free Hospital, London, United Kingdom
- Department of Nuclear Medicine, Royal Free Hospital, London, United Kingdom
| | - Jennifer Watkins
- Neuroendocrine Tumour Unit, ENETS Centre of Excellence, Royal Free Hospital, London, United Kingdom
- Department of Cellular Pathology, Royal Free Hospital, London, United Kingdom
| | - Tu Vinh Luong
- Neuroendocrine Tumour Unit, ENETS Centre of Excellence, Royal Free Hospital, London, United Kingdom
- Department of Cellular Pathology, Royal Free Hospital, London, United Kingdom
| | - Dalvinder Mandair
- Neuroendocrine Tumour Unit, ENETS Centre of Excellence, Royal Free Hospital, London, United Kingdom
| | - Christos Toumpanakis
- Neuroendocrine Tumour Unit, ENETS Centre of Excellence, Royal Free Hospital, London, United Kingdom
| | - Christina Thirlwell
- College of Medicine and Health, University of Exeter, Exeter, United Kingdom
- UCL Cancer Institute, University College London, London, United Kingdom
| | - Martyn E Caplin
- Neuroendocrine Tumour Unit, ENETS Centre of Excellence, Royal Free Hospital, London, United Kingdom
| | - Tim Meyer
- Department of Oncology, Royal Free Hospital, London, United Kingdom
- UCL Cancer Institute, University College London, London, United Kingdom
| |
Collapse
|
49
|
Fröss-Baron K, Garske-Roman U, Welin S, Granberg D, Eriksson B, Khan T, Sandström M, Sundin A. 177Lu-DOTATATE Therapy of Advanced Pancreatic Neuroendocrine Tumors Heavily Pretreated with Chemotherapy: Analysis of Outcome, Safety, and Their Determinants. Neuroendocrinology 2021; 111:330-343. [PMID: 32097917 DOI: 10.1159/000506746] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 02/21/2020] [Indexed: 01/27/2023]
Abstract
OBJECTIVE To retrospectively analyze toxicity, progression-free survival (PFS), overall survival (OS), and their determinants in patients with advanced pancreatic neuroendocrine tumors (PanNETs), previously pretreated with chemothe-r-apy, undergoing peptide receptor radionuclide therapy (PRRT) with 177Lu-DOTATATE. METHODS A total of 102 patients with advanced PanNETs, previously pretreated with one (67%) or several (33%) lines of chemotherapy, were included, of whom 90% had progressive disease and the majority (74.5%) had grade 2 tumors. 177Lu-DOTATATE, 7.4 GBq per cycle, was administered with 6- to 8-week intervals in 88% of patients utilizing a dosimetry-guided protocol until an absorbed dose of 23 Gy to the kidneys was reached. RESULTS A mean dose of 32 ± 10.9 GBq per patient was administered in 1-10 cycles starting a median of 36 months after PanNET diagnosis. The median follow-up was 34 months, the median PFS was 24 months, and the median OS was 42 months from start of PRRT. Independent risk factors for both progression and death were liver tumor burden >50%, more than one line of previous chemotherapy, and elevated alkaline phosphatase. Resection of the primary tumor was linked to longer survival. Bone marrow toxicity grade 3-4 occurred in 10.8%. One patient (1.0%) developed acute myeloid leukemia. Bone marrow toxicity was unrelated to type and length of previous chemotherapy, amount of administered activity, and absorbed dose to the bone marrow. CONCLUSION 177Lu-DOTATATE therapy was feasible, highly effective, and safe in patients with advanced PanNETs heavily pretreated with chemotherapy. More than one line of chemotherapy was a therapy-related independent risk factor for shorter PFS and OS.
Collapse
Affiliation(s)
| | - Ulrike Garske-Roman
- Department of Nuclear Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Staffan Welin
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Dan Granberg
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Barbro Eriksson
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Tanweera Khan
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Mattias Sandström
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Anders Sundin
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
50
|
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: 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: 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
|