1
|
Vulasala SS, Virarkar M, Gopireddy D, Waters R, Alkhasawneh A, Awad Z, Maxwell J, Ramani N, Kumar S, Onteddu N, Morani AC. Small Bowel Neuroendocrine Neoplasms-A Review. J Comput Assist Tomogr 2024; 48:563-576. [PMID: 38110305 DOI: 10.1097/rct.0000000000001541] [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: 12/20/2023]
Abstract
Neuroendocrine neoplasms (NENs) are rapidly evolving small bowel tumors, and the patients are asymptomatic at the initial stages. Metastases are commonly observed at the time of presentation and diagnosis. This review addresses the small bowel NEN (SB-NEN) and its molecular, histological, and imaging features, which aid diagnosis and therapy guidance. Somatic cell number alterations and epigenetic mutations are studied to be responsible for sporadic and familial SB-NEN. The review also describes the grading of SB-NEN in addition to rare histological findings such as mixed neuroendocrine-non-NENs. Anatomic and nuclear imaging with conventional computed tomography, magnetic resonance imaging, computed tomographic enterography, and positron emission tomography are adopted in clinical practice for diagnosing, staging, and follow-up of NEN. Along with the characteristic imaging features of SB-NEN, the therapeutic aspects of imaging, such as peptide receptor radionuclide therapy, are discussed in this review.
Collapse
Affiliation(s)
- Sai Swarupa Vulasala
- From the Department of Radiology, University of Florida College of Medicine, Jacksonville
| | - Mayur Virarkar
- Department of Radiology, University of Florida College of Medicine, Jacksonville, FL
| | - Dheeraj Gopireddy
- Department of Radiology, University of Florida College of Medicine, Jacksonville, FL
| | - Rebecca Waters
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Ziad Awad
- Surgery, University of Florida College of Medicine, Jacksonville, FL
| | - Jessica Maxwell
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center
| | - Nisha Ramani
- Department of Pathology, Michael E. DeBakey VA Medical Center, Houston, TX
| | - Sindhu Kumar
- Department of Radiology, University of Florida College of Medicine, Jacksonville, FL
| | - Nirmal Onteddu
- Department of Internal Medicine, University of Florida College of Medicine, Jacksonville, FL
| | - Ajaykumar C Morani
- Department of Diagnostic Radiology, University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
2
|
Delpassand ES, Yazdi SM, Ghantoji S, Nakasato A, Strickland C, Nunez R, Shafie A, Cork S, Byrne C, Tang J, Patel J. Effectiveness and Safety of Retreatment with 177Lu-DOTATATE in Patients with Progressive Neuroendocrine Tumors: A Retrospective Real-World Study in the United States. J Nucl Med 2024; 65:746-752. [PMID: 38514088 DOI: 10.2967/jnumed.123.265703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 02/13/2024] [Indexed: 03/23/2024] Open
Abstract
Advanced neuroendocrine tumors (NETs) are associated with a poor prognosis. A regimen of 4 cycles of 177Lu-DOTATATE has been shown to improve both progression-free survival (PFS) and overall survival (OS) in patients with advanced NETs. To the best of our knowledge, this is the first study in the United States to evaluate the effectiveness and safety of additional cycles of 177Lu-DOTATATE therapy in patients with progressive NETs. Methods: This was a retrospective chart review of adults with advanced NETs. The patients had undergone initial treatment with up to 4 cycles of 177Lu-DOTATATE and, after disease progression and a period of at least 6 mo since the end of the initial treatment, were retreated with at least 1 additional cycle at a single center (2010-2020). Patient characteristics, treatment patterns, and clinical outcomes were evaluated descriptively. Response was evaluated according to RECIST 1.1; toxicity was defined using criteria from Common Terminology Criteria for Adverse Events, version 5.0. Kaplan-Meier plots were used to evaluate PFS and OS. Results: Of the 31 patients who received 177Lu-DOTATATE retreatment, 61% were male and 94% were White. Overall, patients received a median of 6 cycles (4 initial cycles and 2 retreatment cycles), and the mean administered activity was 41.9 GBq. Two patients also went on to receive additional retreatment (1 and 2 cycles, individually) after a second period of at least 6 mo and progression after retreatment. Best responses of partial response and stable disease were observed in 35% and 65% of patients after the initial treatment and 23% and 45% of patients after retreatment, respectively. The median PFS after the initial treatment was 20.2 mo and after retreatment was 9.6 mo. The median OS after the initial treatment was 42.6 mo and after retreatment was 12.6 mo. Hematologic parameters decreased significantly during both the initial treatment and retreatment but recovered such that there was little difference between the values before the initial treatment and before the retreatment. Clinically significant hematotoxicity occurred in 1 and 3 patients after the initial treatment and retreatment, respectively. No grade 3 or 4 nephrotoxicity was observed. Conclusion: Retreatment with 177Lu-DOTATATE after progression appeared to be well tolerated and offered disease control in patients with progressive NETs after initial 177Lu-DOTATATE treatment.
Collapse
Affiliation(s)
| | - Soheil M Yazdi
- Excel Diagnostics and Nuclear Oncology Center, Houston, Texas
| | | | | | | | - Rodolfo Nunez
- Excel Diagnostics and Nuclear Oncology Center, Houston, Texas
| | - Afshin Shafie
- Excel Diagnostics and Nuclear Oncology Center, Houston, Texas
| | - Susan Cork
- Excel Diagnostics and Nuclear Oncology Center, Houston, Texas
| | | | | | - Jeetvan Patel
- Novartis Pharmaceuticals Corp., East Hanover, New Jersey; and
| |
Collapse
|
3
|
Prinzi N, Corti F, Torchio M, Niger M, Antista M, Pagani F, Beninato T, Pulice I, Rossi RE, Coppa J, Cascella T, Giacomelli L, Di Bartolomeo M, Milione M, de Braud F, Pusceddu S. Metastatic pheochromocytomas and paragangliomas: where are we? TUMORI JOURNAL 2022; 108:526-540. [PMID: 35593402 DOI: 10.1177/03008916221078621] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Pheochromocytomas and paragangliomas (PPGLs) can metastasize in approximately 15-20% of cases. This review discusses the available evidence on the biology and treatment of metastatic PPGLs. Chemotherapy is the first-line treatment option for this evolving and symptomatic disease. In patients with high MIBG uptake and positive PETGa-68, radiometabolic treatment may be considered. The efficacy of sunitinib has been shown in observational studies, and pembrolizumab has been evaluated in phase II clinical studies, while other agents investigated in this setting are anti-angiogenic drugs cabozantinib, dovitinib, axitinib and lenvatinib. As these agents' efficacy and safety data, alone or in combination, are scant and based on few treated patients, enrollment in clinical trials is mandatory. Future therapeutic options may be represented by DNA repair system inhibitors (such as olaparib), HIF2 inhibitors and immunotherapy.
Collapse
Affiliation(s)
- Natalie Prinzi
- Department of Medical Oncology, Fondazione IRCCS Istituto Tumori Milano, Milan, Italy
| | - Francesca Corti
- Department of Medical Oncology, Fondazione IRCCS Istituto Tumori Milano, Milan, Italy
| | - Martina Torchio
- Department of Medical Oncology, Fondazione IRCCS Istituto Tumori Milano, Milan, Italy
| | - Monica Niger
- Department of Medical Oncology, Fondazione IRCCS Istituto Tumori Milano, Milan, Italy
| | - Maria Antista
- Department of Medical Oncology, Fondazione IRCCS Istituto Tumori Milano, Milan, Italy
| | - Filippo Pagani
- Department of Medical Oncology, Fondazione IRCCS Istituto Tumori Milano, Milan, Italy
| | - Teresa Beninato
- Department of Medical Oncology, Fondazione IRCCS Istituto Tumori Milano, Milan, Italy
| | - Iolanda Pulice
- Clinical Trial Center, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Roberta Elisa Rossi
- Gastro-intestinal Surgery and Liver Transplantation Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.,Department of Pathophysiology and Organ Transplant, Università degli Studi di Milano, Milan, Italy
| | - Jorgelina Coppa
- Gastro-intestinal Surgery and Liver Transplantation Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Tommaso Cascella
- Radiology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | - Maria Di Bartolomeo
- Department of Medical Oncology, Fondazione IRCCS Istituto Tumori Milano, Milan, Italy
| | - Massimo Milione
- Diagnostic Pathology and Laboratory Medicine Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Filippo de Braud
- Department of Medical Oncology, Fondazione IRCCS Istituto Tumori Milano, Milan, Italy.,Oncology and Hemato-Oncology Department, Università degli Studi di Milano, Milan, Italy
| | - Sara Pusceddu
- Department of Medical Oncology, Fondazione IRCCS Istituto Tumori Milano, Milan, Italy
| |
Collapse
|
4
|
Pusceddu S, Prinzi N, Tafuto S, Ibrahim T, Filice A, Brizzi MP, Panzuto F, Baldari S, Grana CM, Campana D, Davì MV, Giuffrida D, Zatelli MC, Partelli S, Razzore P, Marconcini R, Massironi S, Gelsomino F, Faggiano A, Giannetta E, Bajetta E, Grimaldi F, Cives M, Cirillo F, Perfetti V, Corti F, Ricci C, Giacomelli L, Porcu L, Di Maio M, Seregni E, Maccauro M, Lastoria S, Bongiovanni A, Versari A, Persano I, Rinzivillo M, Pignata SA, Rocca PA, Lamberti G, Cingarlini S, Puliafito I, Ambrosio MR, Zanata I, Bracigliano A, Severi S, Spada F, Andreasi V, Modica R, Scalorbi F, Milione M, Sabella G, Coppa J, Casadei R, Di Bartolomeo M, Falconi M, de Braud F. Association of Upfront Peptide Receptor Radionuclide Therapy With Progression-Free Survival Among Patients With Enteropancreatic Neuroendocrine Tumors. JAMA Netw Open 2022; 5:e220290. [PMID: 35201309 PMCID: PMC8874344 DOI: 10.1001/jamanetworkopen.2022.0290] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 12/03/2021] [Indexed: 02/05/2023] Open
Abstract
IMPORTANCE Data about the optimal timing for the initiation of peptide receptor radionuclide therapy (PRRT) for advanced, well-differentiated enteropancreatic neuroendocrine tumors are lacking. OBJECTIVE To evaluate the association of upfront PRRT vs upfront chemotherapy or targeted therapy with progression-free survival (PFS) among patients with advanced enteropancreatic neuroendocrine tumors who experienced disease progression after treatment with somatostatin analogues (SSAs). DESIGN, SETTING, AND PARTICIPANTS This retrospective, multicenter cohort study analyzed the clinical records from 25 Italian oncology centers for patients aged 18 years or older who had unresectable, locally advanced or metastatic, well-differentiated, grades 1 to 3 enteropancreatic neuroendocrine tumors and received either PRRT or chemotherapy or targeted therapy after experiencing disease progression after treatment with SSAs between January 24, 2000, and July 1, 2020. Propensity score matching was done to minimize the selection bias. EXPOSURES Upfront PRRT or upfront chemotherapy or targeted therapy. MAIN OUTCOMES AND MEASURES The main outcome was the difference in PFS among patients who received upfront PRRT vs among those who received upfront chemotherapy or targeted therapy. A secondary outcome was the difference in overall survival between these groups. Hazard ratios (HRs) were fitted in a multivariable Cox proportional hazards regression model to adjust for relevant factors associated with PFS and were corrected for interaction with these factors. RESULTS Of 508 evaluated patients (mean ([SD] age, 55.7 [0.5] years; 278 [54.7%] were male), 329 (64.8%) received upfront PRRT and 179 (35.2%) received upfront chemotherapy or targeted therapy. The matched group included 222 patients (124 [55.9%] male; mean [SD] age, 56.1 [0.8] years), with 111 in each treatment group. Median PFS was longer in the PRRT group than in the chemotherapy or targeted therapy group in the unmatched (2.5 years [95% CI, 2.3-3.0 years] vs 0.7 years [95% CI, 0.5-1.0 years]; HR, 0.35 [95% CI, 0.28-0.44; P < .001]) and matched (2.2 years [95% CI, 1.8-2.8 years] vs 0.6 years [95% CI, 0.4-1.0 years]; HR, 0.37 [95% CI, 0.27-0.51; P < .001]) populations. No significant differences were shown in median overall survival between the PRRT and chemotherapy or targeted therapy groups in the unmatched (12.0 years [95% CI, 10.7-14.1 years] vs 11.6 years [95% CI, 9.1-13.4 years]; HR, 0.81 [95% CI, 0.62-1.06; P = .11]) and matched (12.2 years [95% CI, 9.1-14.2 years] vs 11.5 years [95% CI, 9.2-17.9 years]; HR, 0.83 [95% CI, 0.56-1.24; P = .36]) populations. The use of upfront PRRT was independently associated with improved PFS (HR, 0.37; 95% CI, 0.26-0.51; P < .001) in multivariable analysis. After adjustment of values for interaction, upfront PRRT was associated with longer PFS regardless of tumor functional status (functioning: adjusted HR [aHR], 0.39 [95% CI, 0.27-0.57]; nonfunctioning: aHR, 0.29 [95% CI, 0.16-0.56]), grade of 1 to 2 (grade 1: aHR, 0.21 [95% CI, 0.12-0.34]; grade 2: aHR, 0.52 [95% CI, 0.29-0.73]), and site of tumor origin (pancreatic: aHR, 0.41 [95% CI, 0.24-0.61]; intestinal: aHR, 0.19 [95% CI, 0.11-0.43]) (P < .001 for all). Conversely, the advantage was not retained in grade 3 tumors (aHR, 0.31; 95% CI, 0.12-1.37; P = .13) or in tumors with a Ki-67 proliferation index greater than 10% (aHR, 0.73; 95% CI, 0.29-1.43; P = .31). CONCLUSIONS AND RELEVANCE In this cohort study, treatment with upfront PRRT in patients with enteropancreatic neuroendocrine tumors who had experienced disease progression with SSA treatment was associated with significantly improved survival outcomes compared with upfront chemotherapy or targeted therapy. Further research is needed to investigate the correct strategy, timing, and optimal specific sequence of these therapeutic options.
Collapse
Affiliation(s)
- Sara Pusceddu
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, European Neuroendocrine Tumor Society (ENETS) Center of Excellence, Milan, Italy
| | - Natalie Prinzi
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, European Neuroendocrine Tumor Society (ENETS) Center of Excellence, Milan, Italy
| | - Salvatore Tafuto
- Oncologia Clinica e Sperimentale Sarcomi e Tumori Rari, Istituto Nazionale Tumori IRCCS, Fondazione G. Pascale, Naples, Italy
| | - Toni Ibrahim
- Osteoncology and Rare Tumors Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori “Dino Amadori,” Meldola, Italy
| | - Angelina Filice
- Nuclear Medicine Unit, Azienda Unità Sanitaria Locale–IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Maria Pia Brizzi
- Azienda Ospedaliera Universitaria San Luigi Gonzaga, Orbassano, Italy
| | - Francesco Panzuto
- Digestive Disease Unit, Sant’Andrea University Hospital, ENETS Center of Excellence, Rome, Italy
| | - Sergio Baldari
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Nuclear Medicine Unit, University of Messina, Messina, Italy
| | - Chiara M. Grana
- Division of Nuclear Medicine, IRCCS Istituto Europeo di Oncologia, Milan, Italy
| | - Davide Campana
- Department of Experimental Diagnostic and Specialized Medicine, Alma Mater Studiorum, University of Bologna, Bologna, Italy
- Division of Medical Oncology, IRCCS Azienda Ospedaliera–Universitaria Bologna, Neuroendocrine Tumor Team Bologna, ENETS Center of Excellence Bologna, Bologna, Italy
| | - Maria Vittoria Davì
- Department of Medicine, Section of Endocrinology, University and Hospital Trust of Verona, ENETS Center of Excellence, Verona, Italy
| | - Dario Giuffrida
- Oncologia Medica, Istituto Oncologico del Mediterraneo, Viagrande (Catania), Italy
| | - Maria Chiara Zatelli
- Department of Medical Sciences, Section of Endocrinology, Geriatrics and Internal Medicine, University of Ferrara, Ferrara, Italy
| | - Stefano Partelli
- Pancreatic Surgery, Pancreas Translational and Clinical Research Center, San Raffaele Hospital IRCCS, Università Vita-Salute San Raffaele, ENETS Center of Excellence, Milano, Italy
| | - Paola Razzore
- Department of Internal Medicine, Division of Endocrinology, A.O. Ordine Mauriziano, Turin, Italy
| | - Riccardo Marconcini
- Department of Oncology, Santa Chiara Hospital, Azienda Ospedaliero–Universitaria Pisana, Pisa, Italy
| | - Sara Massironi
- Division of Gastroenterology, Ospedale San Gerardo, University of Milano–Bicocca, Monza, Italy
| | - Fabio Gelsomino
- Department of Oncology and Haematology, University Hospital of Modena, Modena, Italy
| | - Antongiulio Faggiano
- Department of Clinical and Molecular Medicine, Endocrinology Unit, Sant’Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Elisa Giannetta
- Department of Experimental Medicine, Sapienza Università Roma, Rome, Italy
| | - Emilio Bajetta
- Istituto di Oncologia, Policlinico di Monza, Monza, Italy
| | - Franco Grimaldi
- Endocrinology and Metabolism Unit, University Hospital S. Maria della Misericordia, Udine, Italy
| | - Mauro Cives
- Department of Biomedical Sciences and Human Oncology, University of Bari, Bari, Italy
- National Cancer Center, Tumori Institute Giovanni Paolo II, Bari, Italy
| | - Fernando Cirillo
- Department of Surgery, General Surgery Unit, Gruppo Tumori Rari, Azienda Socio-Sanitaria Territoriale–Cremona, Cremona, Italy
| | | | - Francesca Corti
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, European Neuroendocrine Tumor Society (ENETS) Center of Excellence, Milan, Italy
| | - Claudio Ricci
- Division of Pancreatic Surgery, IRCCS Azienda Ospedaliero–Universitaria Di Bologna, Bologna, Italy
- Department of Internal Medicine and Surgery, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | | | - Luca Porcu
- Methodology for Clinical Research Laboratory, Oncology Department, IRCCS Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Massimo Di Maio
- Department of Oncology, University of Turin, A.O. Ordine Mauriziano, Torino, Italy
| | - Ettore Seregni
- Department of Nuclear Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Milan, Italy
| | - Marco Maccauro
- Department of Nuclear Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Milan, Italy
| | - Secondo Lastoria
- Nuclear Medicine Unit, Istituto Nazionale Tumori IRCCS, Fondazione G. Pascale, Naples, Italy
| | - Alberto Bongiovanni
- Osteoncology and Rare Tumors Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori “Dino Amadori,” Meldola, Italy
| | - Annibale Versari
- Nuclear Medicine Unit, Azienda Unità Sanitaria Locale–IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Irene Persano
- Azienda Ospedaliera Universitaria San Luigi Gonzaga, Orbassano, Italy
| | - Maria Rinzivillo
- Digestive Disease Unit, Sant’Andrea University Hospital, ENETS Center of Excellence, Rome, Italy
| | - Salvatore Antonio Pignata
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Nuclear Medicine Unit, University of Messina, Messina, Italy
- Nuclear Medicine Unit, Azienda Ospedaliera Papardo, Messina, Italy
| | - Paola Anna Rocca
- Division of Nuclear Medicine, IRCCS Istituto Europeo di Oncologia, Milan, Italy
| | - Giuseppe Lamberti
- Department of Experimental Diagnostic and Specialized Medicine, Alma Mater Studiorum, University of Bologna, Bologna, Italy
- Division of Medical Oncology, IRCCS Azienda Ospedaliera–Universitaria Bologna, Neuroendocrine Tumor Team Bologna, ENETS Center of Excellence Bologna, Bologna, Italy
| | - Sara Cingarlini
- Department of Medicine, Oncology, University and Hospital Trust of Verona, ENETS Center of Excellence, Verona, Italy
| | - Ivana Puliafito
- Oncologia Medica, Istituto Oncologico del Mediterraneo, Viagrande (Catania), Italy
| | - Maria Rosaria Ambrosio
- Department of Medical Sciences, Section of Endocrinology, Geriatrics and Internal Medicine, University of Ferrara, Ferrara, Italy
| | - Isabella Zanata
- Department of Medical Sciences, Section of Endocrinology, Geriatrics and Internal Medicine, University of Ferrara, Ferrara, Italy
| | - Alessandra Bracigliano
- Oncologia Clinica e Sperimentale Sarcomi e Tumori Rari, Istituto Nazionale Tumori IRCCS, Fondazione G. Pascale, Naples, Italy
| | - Stefano Severi
- Nuclear Medicine Therapy Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori “Dino Amadori,” Meldola, Italy
| | - Francesca Spada
- Division of Medical Oncology, IRCCS Istituto Europeo di Oncologia, Milan, Italy
| | - Valentina Andreasi
- Pancreatic Surgery, Pancreas Translational and Clinical Research Center, San Raffaele Hospital IRCCS, Università Vita-Salute San Raffaele, ENETS Center of Excellence, Milano, Italy
| | - Roberta Modica
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Federica Scalorbi
- Department of Nuclear Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Milan, Italy
| | - Massimo Milione
- First Division of Pathology, Department of Pathology and Laboratory Medicine, IRCCS Foundation National Cancer Institute, Milan, Italy
| | - Giovanna Sabella
- First Division of Pathology, Department of Pathology and Laboratory Medicine, IRCCS Foundation National Cancer Institute, Milan, Italy
| | - Jorgelina Coppa
- Gastro-entero-pancreatic Surgical and Liver Transplantation Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, ENETS Center of Excellence, Milan, Italy
| | - Riccardo Casadei
- Division of Pancreatic Surgery, IRCCS Azienda Ospedaliero–Universitaria Di Bologna, Bologna, Italy
- Department of Internal Medicine and Surgery, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Maria Di Bartolomeo
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, European Neuroendocrine Tumor Society (ENETS) Center of Excellence, Milan, Italy
| | - Massimo Falconi
- Pancreatic Surgery, Pancreas Translational and Clinical Research Center, San Raffaele Hospital IRCCS, Università Vita-Salute San Raffaele, ENETS Center of Excellence, Milano, Italy
| | - Filippo de Braud
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, European Neuroendocrine Tumor Society (ENETS) Center of Excellence, Milan, Italy
- Department of Oncology and Hemato-Oncology, Università deli Studi di Milano, Milan, Italy
| |
Collapse
|
5
|
Zidan L, Iravani A, Oleinikov K, Ben-Haim S, Gross DJ, Meirovitz A, Maimon O, Akhurst T, Michael M, Hicks RJ, Grozinsky-Glasberg S, Kong G. Efficacy and safety of 177Lu-DOTATATE in lung neuroendocrine tumors: a bi-center study. J Nucl Med 2021; 63:218-225. [PMID: 34049983 DOI: 10.2967/jnumed.120.260760] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 04/21/2021] [Indexed: 11/16/2022] Open
Abstract
To assess the efficacy and safety of 177Lu-DOTATATE in patients with somatostatin receptor (SSR) positive lung neuroendocrine tumor (NET). Methods: This is a retrospective review of the outcome of patients with typical carcinoid (TC) and atypical carcinoid (AC), treated with 177Lu-DOTATATE at two ENETS Centres of Excellence. Morphological imaging (RECIST 1.1) and 68Ga-DOTATATE PET/CT responses were assessed at 3 months after completion of 177Lu-DOTATATE. Concordance between two response assessment methods was evaluated by Kappa statistics. Progression-free survival (PFS) and overall survival (OS) was estimated by Kaplan-Meier analysis and compared by Log-rank test. Treatment-related adverse events (AEs) were graded based on CTCAE version 5. Results: Of 48 patients (median age, 63 years, 13 female), 43 (90%) had AC and 5 (10%) TC. Almost all patients (47, 98%) were treated due to progression. Majority (40, 83%) received somatostatin analogs and 10 patients (20%) had prior everolimus, chemotherapy or both. All patients had high SSR expression (≥ modified Krenning score 3) on pre-treatment 68Ga-DOTATATE PET/CT. Patients received a median 4 (range 1-4) cycles of 177Lu-DOTATATE (33% with concurrent radiosensitizing chemotherapy) to a median cumulative activity of 27GBq (range 6-43GBq). At median follow-up of 42 months, the median PFS and OS were 23 months (95% CI 18-28 months) and 59 months (95% CI 50-not reached [NR]), respectively. Of 40 patients with RECIST-measurable disease and 39 patients with available 68Ga-DOTATATE PET/CT response categories were: partial response, 20% (95% CI 10-35%) and 44% (95% CI 30-59%); stable disease, 68% (95% CI 52-80%) and 44% (95% CI 30-59%) and progressive disease 12% (95% CI 5-27%) by both, respectively. There was a moderate concordance between response categories by RECIST and 68Ga-DOTATATE PET/CT, weighted Kappa of 0.51 (95% CI 0.21-0.68). Of patients with stable disease by RECIST, those with partial response on 68Ga-DOTATATE PET/CT had longer OS compared to those with no response, NR vs 52 months (95% CI 28-64), HR 0.2 (95% CI 0.1-0.6), p 0.001. Most grade 3/4 AEs were reversible and the most common was lymphopenia (14%) with no incidence of myelodysplasia/leukemia. Conclusion: In patients with advanced progressive lung NET and satisfactory SSR expression, 177Lu-DOTATATE is effective and safe with a high disease control rate and encouraging PFS and OS.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Grace Kong
- Peter MacCallum Cancer Centre, Australia
| |
Collapse
|
6
|
Prinzi N, Rossi RE, Leuzzi G, Pusceddu S. NETs of the Lung. NEUROENDOCRINE NEOPLASIA MANAGEMENT 2021:163-178. [DOI: 10.1007/978-3-030-72830-4_12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
7
|
Neuroendocrine Lung Cancer Mouse Models: An Overview. Cancers (Basel) 2020; 13:cancers13010014. [PMID: 33375066 PMCID: PMC7792789 DOI: 10.3390/cancers13010014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/18/2020] [Accepted: 12/19/2020] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Neuroendocrine lung tumors are a heterogeneous group of malignancies that share a common neuroendocrine nature. They range from low- and intermediate-grade typical and atypical carcinoma, to the highly malignant large cell neuroendocrine lung carcinoma and small cell carcinoma, with marked differences in incidences and prognosis. This review delineates the current knowledge of the genetic landscape of the human tumors, its influence in the development of genetically engineered mouse models (GEMMs) and the molecular imaging tools available to detect and monitor these diseases. While small cell lung carcinoma is one of the diseases best represented by GEMMs, there is a worrying lack of animal models for the other members of the group, these being understudied diseases. Regardless of the incidence and material available, they all are in urgent need of effective therapies. Abstract Neuroendocrine lung tumors comprise a range of malignancies that extend from benign tumorlets to the most prevalent and aggressive Small Cell Lung Carcinoma (SCLC). They also include low-grade Typical Carcinoids (TC), intermediate-grade Atypical Carcinoids (AC) and high-grade Large Cell Neuroendocrine Carcinoma (LCNEC). Optimal treatment options have not been adequately established: surgical resection when possible is the choice for AC and TC, and for SCLC chemotherapy and very recently, immune checkpoint inhibitors. Some mouse models have been generated based on the molecular alterations identified in genomic analyses of human tumors. With the exception of SCLC, there is a limited availability of (preclinical) models making their development an unmet need for the understanding of the molecular mechanisms underlying these diseases. For SCLC, these models are crucial for translational research and novel drug testing, given the paucity of human material from surgery. The lack of early detection systems for lung cancer point them out as suitable frameworks for the identification of biomarkers at the initial stages of tumor development and for testing molecular imaging methods based on somatostatin receptors. Here, we review the relevant models reported to date, their impact on the understanding of the biology of the tumor subtypes and their relationships, as well as the effect of the analyses of the genetic landscape of the human tumors and molecular imaging tools in their development.
Collapse
|
8
|
Prinzi N, Rossi RE, Proto C, Leuzzi G, Raimondi A, Torchio M, Milione M, Corti F, Colombo E, Prisciandaro M, Cascella T, Spreafico C, Beninato T, Coppa J, Lo Russo G, Di Bartolomeo M, de Braud F, Pusceddu S. Recent Advances in the Management of Typical and Atypical Lung Carcinoids. Clin Lung Cancer 2020; 22:161-169. [PMID: 33618994 DOI: 10.1016/j.cllc.2020.12.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 11/30/2020] [Accepted: 12/10/2020] [Indexed: 11/17/2022]
Abstract
Neuroendocrine neoplasms of the lung represent about 20% to 30% of all neuroendocrine tumors. On the basis of clinical and pathologic characteristics, 2 different categories of tumors may be defined: poorly differentiated neuroendocrine neoplasms, characterized by a high rate of recurrences and poor prognosis, and well-differentiated neuroendocrine neoplasms (typical carcinoids and atypical carcinoids), which generally display an indolent course. Lung carcinoids represent only 1% to 5% of all lung malignancies, but their incidence has significantly increased over the past 30 years. Surgery is the reference standard of treatment for lung carcinoids with locoregional disease. For advanced or unresectable lung carcinoids, several therapeutic options are available, but the choice should be shared within a multidisciplinary team to ensure optimal therapeutic outcomes. We describe the current management of these rare neoplasms.
Collapse
Affiliation(s)
| | - Roberta Elisa Rossi
- Gastrointestinal Surgery and Liver Transplantation Unit; Department of Pathophysiology and Organ Transplant, Università degli Studi di Milano, Milan, Italy
| | | | | | | | | | - Massimo Milione
- Diagnostic Pathology and Laboratory Medicine Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | | | | | - Tommaso Cascella
- Radiology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Carlo Spreafico
- Radiology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | | | | | | | - Filippo de Braud
- Department of Medical Oncology; Oncology and Hemato-oncology Department, Università degli Studi di Milano, Milan, Italy
| | | |
Collapse
|
9
|
Briganti V, Cuccurullo V, Berti V, Di Stasio GD, Linguanti F, Mungai F, Mansi L. 99mTc-EDDA/HYNIC-TOC is a New Opportunity in Neuroendocrine Tumors of the Lung (and in other Malignant and Benign Pulmonary Diseases). Curr Radiopharm 2020; 13:166-176. [PMID: 31886756 PMCID: PMC8193811 DOI: 10.2174/1874471013666191230143610] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 03/11/2019] [Accepted: 11/11/2019] [Indexed: 12/25/2022]
Abstract
Neuroendocrine tumors (NETs) consist of a relatively rare spectrum of malignancies that can arise from neuroendocrine cells; lung NETs (L-NETs) represent about 25% of primary lung neoplasm and 10% of all carcinoid tumors. Diagnostic algorithm usually takes into consideration chest Xray, contrast-enhanced CT and MRI. Nuclear medicine plays a crucial role in the detection and correct assessment of neoplastic functional status as it provides in vivo metabolic data related to the overexpression of Somatostatin Receptors (SSTRs) and also predicting response to peptide receptor radionuclide therapy (PRRT). 111In-Pentreotide (Octreoscan®) is commercially available for imaging of neuroendocrine tumors, their metastases and the management of patients with NETs. More recently, 99mTc-EDDA/HYNIC-TOC(Tektrotyd®) was introduced into the market and its use has been approved for imaging of patients with L-NETs and other SSTR-positive tumors. 99mTc-EDDA/HYNIC-TOC could also represent a good alternative to 68Ga-DOTA-peptides (DOTA-TOC, DOTA-NOC, DOTATATE) in hospitals or centers where PET/CT or 68Ge/68Ga generators are not available. When compared to 111In-Pentetreotide, Tektrotyd® showed slightly higher sensitivity, in the presence of higher imaging quality and lower radiation exposure for patients. Interesting perspectives depending on the kinetic analysis allowed by Tektrotyd® may be obtained in differential diagnosis of non-small cells lung cancer (NSCLC) versus small cells lung cancer (SCLC) and NETs. An interesting perspective could be also associated with a surgery radio-guided by Tektrotyd® in operable lung tumors, including either NETs and NSCLC.
Collapse
Affiliation(s)
| | - Vincenzo Cuccurullo
- Address correspondence to this author at the Medicina Nucleare, Università della Campania “Luigi Vanvitelli”, P.zza Miraglia 2, 80138 Napoli, Italy; E-mail:
| | | | | | | | | | | |
Collapse
|
10
|
Taïeb D, Jha A, Treglia G, Pacak K. Molecular imaging and radionuclide therapy of pheochromocytoma and paraganglioma in the era of genomic characterization of disease subgroups. Endocr Relat Cancer 2019; 26:R627-R652. [PMID: 31561209 PMCID: PMC7002202 DOI: 10.1530/erc-19-0165] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 09/12/2019] [Indexed: 12/13/2022]
Abstract
In recent years, advancement in genetics has profoundly helped to gain a more comprehensive molecular, pathogenic, and prognostic picture of pheochromocytomas and paragangliomas (PPGLs). Newly discovered molecular targets, particularly those that target cell membranes or signaling pathways have helped move nuclear medicine in the forefront of PPGL precision medicine. This is mainly based on the introduction and increasing experience of various PET radiopharmaceuticals across PPGL genotypes quickly followed by implementation of novel radiotherapies and revised imaging algorithms. Particularly, 68Ga-labeled-SSAs have shown excellent results in the diagnosis and staging of PPGLs and in selecting patients for PRRT as a potential alternative to 123/131I-MIBG theranostics. PRRT using 90Y/177Lu-DOTA-SSAs has shown promise for treatment of PPGLs with improvement of clinical symptoms and/or disease control. However, more well-designed prospective studies are required to confirm these findings, in order to fully exploit PRRT's antitumoral properties to obtain the final FDA approval. Such an approval has recently been obtained for high-specific-activity 131I-MIBG for inoperable/metastatic PPGL. The increasing experience and encouraging preliminary results of these radiotherapeutic approaches in PPGLs now raises an important question of how to further integrate them into PPGL management (e.g. monotherapy or in combination with other systemic therapies), carefully taking into account the PPGLs locations, genotypes, and growth rate. Thus, targeted radionuclide therapy (TRT) should preferably be performed at specialized centers with an experienced interdisciplinary team. Future perspectives include the introduction of dosimetry and biomarkers for therapeutic responses for more individualized treatment plans, α-emitting isotopes, and the combination of TRT with other systemic therapies.
Collapse
Affiliation(s)
- David Taïeb
- Department of Nuclear Medicine, La Timone University Hospital, CERIMED, Aix-Marseille University, Marseille, France
| | - Abhishek Jha
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Giorgio Treglia
- Clinic of Nuclear Medicine and PET/CT Center, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Lausanne, Switzerland
- Health Technology Assessment Unit, General Directorate, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
11
|
Nölting S, Ullrich M, Pietzsch J, Ziegler CG, Eisenhofer G, Grossman A, Pacak K. Current Management of Pheochromocytoma/Paraganglioma: A Guide for the Practicing Clinician in the Era of Precision Medicine. Cancers (Basel) 2019; 11:cancers11101505. [PMID: 31597347 PMCID: PMC6827093 DOI: 10.3390/cancers11101505] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 09/18/2019] [Accepted: 09/24/2019] [Indexed: 12/13/2022] Open
Abstract
Pheochromocytomas and paragangliomas (PCC/PGLs) are rare, mostly catecholamine-producing neuroendocrine tumors of the adrenal gland (PCCs) or the extra-adrenal paraganglia (PGL). They can be separated into three different molecular clusters depending on their underlying gene mutations in any of the at least 20 known susceptibility genes: The pseudohypoxia-associated cluster 1, the kinase signaling-associated cluster 2, and the Wnt signaling-associated cluster 3. In addition to tumor size, location (adrenal vs. extra-adrenal), multiplicity, age of first diagnosis, and presence of metastatic disease (including tumor burden), other decisive factors for best clinical management of PCC/PGL include the underlying germline mutation. The above factors can impact the choice of different biomarkers and imaging modalities for PCC/PGL diagnosis, as well as screening for other neoplasms, staging, follow-up, and therapy options. This review provides a guide for practicing clinicians summarizing current management of PCC/PGL according to tumor size, location, age of first diagnosis, presence of metastases, and especially underlying mutations in the era of precision medicine.
Collapse
Affiliation(s)
- Svenja Nölting
- Department of Medicine IV, University Hospital, LMU Munich, Ziemssenstraße 1, 80336 München, Germany.
| | - Martin Ullrich
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstrasse 400, 01328 Dresden, Germany.
| | - Jens Pietzsch
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstrasse 400, 01328 Dresden, Germany.
- Department of Chemistry and Food Chemistry, School of Science, Technische Universität Dresden, Mommsenstrasse 9, 01062 Dresden, Germany.
| | - Christian G Ziegler
- Department of Medicine III, University Hospital Carl Gustav Carus Dresden, Fetscherstraße 74, 01307 Dresden, Germany.
| | - Graeme Eisenhofer
- Department of Medicine III, University Hospital Carl Gustav Carus Dresden, Fetscherstraße 74, 01307 Dresden, Germany.
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus at Technische Universität Dresden, 01307 Dresden, Germany.
| | - Ashley Grossman
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford Ox3 7LJ, UK.
- Department of Gastroenterology, Royal Free Hospital ENETS Centre of Excellence, London NW3 2QG, UK.
| | - Karel Pacak
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20814, USA.
| |
Collapse
|
12
|
Cuccurullo V, Di Stasio GD, Mansi L. Physiopathological Premises to Nuclear Medicine Imaging of Pancreatic Neuroendocrine Tumours. Curr Radiopharm 2019; 12:98-106. [DOI: 10.2174/1874471012666190206094555] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 02/19/2018] [Accepted: 10/29/2018] [Indexed: 02/07/2023]
Abstract
Background:
Pancreatic Neuroendocrine Tumors (P-NETs) are a challenge in terms of both
diagnosis and therapy; morphological studies need to be frequently implemented with nonstandard
techniques such as Endoscopic Ultrasounds, Dynamic CT, and functional Magnetic Resonance.
Discussion:
The role of nuclear medicine, being scarcely sensitive F-18 Fluorodeoxyglucose, is mainly
based on the over-expression of Somatostatin Receptors (SSTR) on neuroendocrine tumor cells surface.
Therefore, SSTR can be used as a target for both diagnosis, using radiotracers labeled with gamma or
positron emitters, and therapy. SSTRs subtypes are capable of homo and heterodimerization in specific
combinations that alter both the response to ligand activation and receptor internalization.
Conclusion:
Although agonists usually provide efficient internalization, also somatostatin antagonists
(SS-ANTs) could be used for imaging and therapy. Peptide Receptor Radionuclide Therapy (PRRT)
represents the most successful option for targeted therapy. The theranostic model based on SSTR does
not work in insulinoma, in which different radiotracers such as F-18 FluoroDOPA or tracers for the
glucagon-like peptide-1 receptor have to be preferred.
Collapse
Affiliation(s)
- Vincenzo Cuccurullo
- Nuclear Medicine Unit, Department of Clinical and Experimental Medicine "F.Magrassi, A.Lanzara" – Universita della Campania "Luigi Vanvitelli", Napoli, Italy
| | - Giuseppe Danilo Di Stasio
- Nuclear Medicine Unit, Department of Clinical and Experimental Medicine "F.Magrassi, A.Lanzara" – Universita della Campania "Luigi Vanvitelli", Napoli, Italy
| | - Luigi Mansi
- Nuclear Medicine Unit, Department of Clinical and Experimental Medicine "F.Magrassi, A.Lanzara" – Universita della Campania "Luigi Vanvitelli", Napoli, Italy
| |
Collapse
|
13
|
Mahajan S, O'Donoghue J, Weber W, Bodei L. Integrating Early Rapid Post-Peptide Receptor Radionuclide Therapy Quality Assurance Scan into the Outpatient Setting. ACTA ACUST UNITED AC 2019; 10. [PMID: 31057988 DOI: 10.4172/2155-9619.1000395] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Objective To understand value of early rapid, quality-assurance (QA), post-therapy whole-body scan (Tx-WBS) in patients receiving peptide receptor radionuclide therapy (PRRT) in outpatient setting. Methods Sixteen patients with metastatic neuroendocrine tumors received PRRT and underwent Tx-WBS after each cycle. Early imaging (3 hour post-injection) was favored. Planar-images obtained on dual-headed gamma camera (speed 30 cm/min) were visually assessed and qualitatively compared with pre-therapy diagnostic scans. Retention% and lesion/spleen (L/S) ratios were calculated. Results Fifty three Tx-WBS were analyzed. No cutaneous contamination, extravasation or unexpected tracer distribution was observed. 46/53 (87%) Tx-WBS in 14/16 (88%) patients demonstrated uptake in metastatic lesions. No significant correlation was seen between L/S ratios and response on follow-up imaging. Qualitative assessment of follow-up images during four-cycles of PRRT provided preliminary estimate of disease course in 11/16 patients; with unexpected findings in 2. Conclusion In daily practice, especially in outpatient setting, an early QA post-PRRT scan proved effective for validating successful treatment and allowing preliminary disease monitoring, at no additional cost.
Collapse
Affiliation(s)
- Sonia Mahajan
- Department of Radiology, Molecular Imaging and Therapy Service, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Joseph O'Donoghue
- Department of Medical Physics and Radiology, Memorial Sloan Kettering Cancer Center, New York, USA
| | | | - Lisa Bodei
- Department of Radiology, Molecular Imaging and Therapy Service, Memorial Sloan Kettering Cancer Center, New York, USA
| |
Collapse
|
14
|
Zhang J, Wang H, Jacobson O, Cheng Y, Niu G, Li F, Bai C, Zhu Z, Chen X. Safety, Pharmacokinetics, and Dosimetry of a Long-Acting Radiolabeled Somatostatin Analog 177Lu-DOTA-EB-TATE in Patients with Advanced Metastatic Neuroendocrine Tumors. J Nucl Med 2018; 59:1699-1705. [PMID: 29653971 PMCID: PMC6225536 DOI: 10.2967/jnumed.118.209841] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 03/26/2018] [Indexed: 12/21/2022] Open
Abstract
Radiolabeled somatostatin analog therapy has become an established treatment method for patients with well to moderately differentiated unresectable or metastatic neuroendocrine tumors (NETs). The most frequently used somatostatin analogs in clinical practice are octreotide and octreotate. However, both peptides showed suboptimal retention within tumors. The aim of this first-in-humans study is to explore the safety and dosimetry of a long-acting radiolabeled somatostatin analog, 177Lu-1, 4, 7, 10-tetra-azacyclododecane-1, 4, 7, 10-tetraacetic acid-Evans blue-octreotate (177Lu-DOTA-EB-TATE). Methods: Eight patients (6 men and 2 women; age range, 27-61 y) with advanced metastatic NETs were recruited. Five patients received a single dose, 0.35-0.70 GBq (9.5-18.9 mCi), of 177Lu-DOTA-EB-TATE and underwent serial whole-body planar and SPECT/CT scans at 2, 24, 72, 120, and 168 h after injection. The other 3 patients received intravenous injection of 0.28-0.41 GBq (7.5-11.1 mCi) of 177Lu-DOTATATE for the same imaging acquisition procedures at 1, 3, 4, 24, and 72 h after injection. The dosimetry was calculated using the OLINDA/EXM 1.1 software. Results: Administration of 177Lu-DOTA-EB-TATE was well tolerated, with no adverse symptoms being noticed or reported in any of the patients. Compared with 177Lu-DOTATATE, 177Lu-DOTA-EB-TATE showed extended circulation in the blood and achieved a 7.9-fold increase of tumor dose delivery. The total-body effective doses were 0.205 ± 0.161 mSv/MBq for 177Lu-DOTA-EB-TATE and 0.174 ± 0.072 mSv/MBq for 177Lu-DOTATATE. Significant dose delivery increases to the kidneys and bone marrow were also observed in patients receiving 177Lu-DOTA-EB-TATE compared with those receiving 177Lu-DOTATATE (3.2 and 18.2-fold, respectively). Conclusion: By introducing an albumin-binding moiety, 177Lu-DOTA-EB-TATE showed remarkably higher uptake and retention in NETs as well as significantly increased accumulation in the kidneys and red marrow. It has great potential to be used in peptide receptor radionuclide therapy for NETs with lower dose and less frequency of administration.
Collapse
Affiliation(s)
- Jingjing Zhang
- Department of Nuclear Medicine, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, Maryland
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing, China; and
| | - Hao Wang
- Department of Nuclear Medicine, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing, China; and
| | - Orit Jacobson
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, Maryland
| | - Yuejuan Cheng
- Oncology Department of Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Gang Niu
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, Maryland
| | - Fang Li
- Department of Nuclear Medicine, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing, China; and
| | - Chunmei Bai
- Oncology Department of Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Zhaohui Zhu
- Department of Nuclear Medicine, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing, China; and
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, Maryland
| |
Collapse
|
15
|
Aristizabal Prada ET, Heinzle V, Knösel T, Nölting S, Spöttl G, Maurer J, Spitzweg C, Angele M, Schmidt N, Beuschlein F, Stalla GK, Blaser R, Kuhn KA, Auernhammer CJ. Tropomyosin receptor kinase: a novel target in screened neuroendocrine tumors. Endocr Relat Cancer 2018; 25:547-560. [PMID: 29563190 DOI: 10.1530/erc-17-0201] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 03/21/2018] [Indexed: 01/27/2023]
Abstract
Tropomyosin receptor kinase (Trk) inhibitors are investigated as a novel targeted therapy in various cancers. We investigated the in vitro effects of the pan-Trk inhibitor GNF-5837 in human neuroendocrine tumor (NET) cells. The human neuroendocrine pancreatic BON1, bronchopulmonary NCI-H727 and ileal GOT1 cell lines were treated with GNF-5837 alone and in combination with everolimus. Cell viability decreased in a time- and dose-dependent manner in GOT1 cells in response to GNF-5837 treatment, while treatment in BON1 and NCI-H727 cells showed no effect on cellular viability. Trk receptor expression determined GNF-5837 sensitivity. GNF-5837 caused downregulation of PI3K-Akt-mTOR signaling, Ras-Raf-MEK-ERK signaling, the cell cycle and increased apoptotic cell death. The combinational treatment of GNF-5837 with everolimus showed a significant enhancement in inhibition of cell viability vs single substance treatments, due to a cooperative PI3K-Akt-mTOR and Ras-Raf-MEK-ERK pathway downregulation, as well as an enhanced cell cycle component downregulation. Immunohistochemical staining for Trk receptors were performed using a tissue microarray containing 107 tumor samples of gastroenteropancreatic NETs. Immunohistochemical staining with TrkA receptor and pan-Trk receptor antibodies revealed a positive staining in pancreatic NETs in 24.2% (8/33) and 33.3% (11/33), respectively. We demonstrated that the pan-Trk inhibitor GNF-5837 has promising anti-tumoral properties in human NET cell lines expressing the TrkA receptor. Immunohistochemical or molecular screening for Trk expression particularly in pancreatic NETs might serve as predictive marker for molecular targeted therapy with Trk inhibitors.
Collapse
Affiliation(s)
- Elke Tatjana Aristizabal Prada
- Interdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM), Klinikum der Universität München, Ludwig-Maximilians-University of Munich, Campus Grosshadern, Munich, Germany
- Department of Internal Medicine 2, University-Hospital, Klinikum der Universität München, Ludwig-Maximilians-University of Munich, Munich, Germany
- Department of Internal Medicine 4, University-Hospital, Klinikum der Universität München, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Vera Heinzle
- Interdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM), Klinikum der Universität München, Ludwig-Maximilians-University of Munich, Campus Grosshadern, Munich, Germany
- Department of Internal Medicine 2, University-Hospital, Klinikum der Universität München, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Thomas Knösel
- Interdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM), Klinikum der Universität München, Ludwig-Maximilians-University of Munich, Campus Grosshadern, Munich, Germany
- Institute of Pathology, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Svenja Nölting
- Interdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM), Klinikum der Universität München, Ludwig-Maximilians-University of Munich, Campus Grosshadern, Munich, Germany
- Department of Internal Medicine 2, University-Hospital, Klinikum der Universität München, Ludwig-Maximilians-University of Munich, Munich, Germany
- Department of Internal Medicine 4, University-Hospital, Klinikum der Universität München, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Gerald Spöttl
- Interdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM), Klinikum der Universität München, Ludwig-Maximilians-University of Munich, Campus Grosshadern, Munich, Germany
- Department of Internal Medicine 2, University-Hospital, Klinikum der Universität München, Ludwig-Maximilians-University of Munich, Munich, Germany
- Department of Internal Medicine 4, University-Hospital, Klinikum der Universität München, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Julian Maurer
- Interdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM), Klinikum der Universität München, Ludwig-Maximilians-University of Munich, Campus Grosshadern, Munich, Germany
- Department of Internal Medicine 2, University-Hospital, Klinikum der Universität München, Ludwig-Maximilians-University of Munich, Munich, Germany
- Department of Internal Medicine 4, University-Hospital, Klinikum der Universität München, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Christine Spitzweg
- Interdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM), Klinikum der Universität München, Ludwig-Maximilians-University of Munich, Campus Grosshadern, Munich, Germany
- Department of Internal Medicine 2, University-Hospital, Klinikum der Universität München, Ludwig-Maximilians-University of Munich, Munich, Germany
- Department of Internal Medicine 4, University-Hospital, Klinikum der Universität München, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Martin Angele
- Interdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM), Klinikum der Universität München, Ludwig-Maximilians-University of Munich, Campus Grosshadern, Munich, Germany
- Department of General, Visceral, Transplantation, Vascular and Thoracic Surgery, University-Hospital, Klinikum der Universität München, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Nina Schmidt
- Interdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM), Klinikum der Universität München, Ludwig-Maximilians-University of Munich, Campus Grosshadern, Munich, Germany
- Department of Internal Medicine 2, University-Hospital, Klinikum der Universität München, Ludwig-Maximilians-University of Munich, Munich, Germany
- Department of Internal Medicine 4, University-Hospital, Klinikum der Universität München, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Felix Beuschlein
- Department of Internal Medicine 4, University-Hospital, Klinikum der Universität München, Ludwig-Maximilians-University of Munich, Munich, Germany
- Klinik für Endokrinologie, Diabetologie und Klinische Ernährung, Universitätsspital Zürich, Zurich, Switzerland
| | - Günter K Stalla
- Clinical Neuroendocrinology, Max Planck Institute of Psychiatry, Munich, Germany
| | - Rainer Blaser
- Institute of Medical Statistics and Epidemiology, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany
| | - Klaus A Kuhn
- Institute of Medical Statistics and Epidemiology, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany
| | - Christoph J Auernhammer
- Interdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM), Klinikum der Universität München, Ludwig-Maximilians-University of Munich, Campus Grosshadern, Munich, Germany
- Department of Internal Medicine 2, University-Hospital, Klinikum der Universität München, Ludwig-Maximilians-University of Munich, Munich, Germany
- Department of Internal Medicine 4, University-Hospital, Klinikum der Universität München, Ludwig-Maximilians-University of Munich, Munich, Germany
| |
Collapse
|
16
|
Peptide Receptor Radionuclide Therapy and the Treatment of Gastroentero-pancreatic Neuroendocrine Tumors: Current Findings and Future Perspectives. Nucl Med Mol Imaging 2018; 52:190-199. [PMID: 29942397 DOI: 10.1007/s13139-018-0517-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 03/10/2018] [Accepted: 03/15/2018] [Indexed: 02/07/2023] Open
Abstract
Purpose and Methods Patients with inoperable and metastasized neuroendocrine tumors (NETs), particularly those with grades 1 and 2, usually receive treatment with somatostatin analogues (SSAs). Peptide receptor radionuclide therapy (PRRT) has gained momentum over the past two decades in patients who progress on SSAs. 177Lu-DOTATATE is currently the most widely used radiopeptide for PRRT. We reviewed the recent evidence on PRRT and the treatment of gastroentero-pancreatic neuroendocrine tumors (GEP-NETs). Results 177Lu-DOTATATE can be used as neoadjuvant treatment in patients with inoperable GEP-NETs, who might be candidate for surgery after treatment and as adjuvant therapy after surgical intervention. Combination treatments of PRRT with chemotherapy or targeted agents as well as combinations of radionuclides in patients with NETs have been explored over the last few years. The majority of patients with NETs experience partial response or have disease stabilization, a small percentage has complete response, while some 30% of patients, however, will have disease progression. The safety and efficacy of retreatment with extra cycles of PRRT as salvage therapy have been evaluated in small retrospective series. Conclusion Overall, there is evidence that disease control and quality of life improve significantly after 117Lu PRRT therapy. Clinical trials on this therapy are scarce, and there is a need for further studies to establish proper management guidelines.
Collapse
|
17
|
Cherk MH, Kong G, Hicks RJ, Hofman MS. Changes in biodistribution on 68Ga-DOTA-Octreotate PET/CT after long acting somatostatin analogue therapy in neuroendocrine tumour patients may result in pseudoprogression. Cancer Imaging 2018; 18:3. [PMID: 29361984 PMCID: PMC5781297 DOI: 10.1186/s40644-018-0136-x] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 01/15/2018] [Indexed: 02/06/2023] Open
Abstract
Background To evaluate the effects of long-acting somatostatin analogue (SSA) therapy on 68Ga-DOTA-octreotate (GaTate) uptake at physiological and metastatic sites in neuroendocrine tumour (NET) patients. Methods Twenty-one patients who underwent GaTate PET/CT before and after commencement of SSA therapy were reviewed. Maximum standardized uptake values (SUVmax) were measured in normal organs. Changes in uptake of 49 metastatic lesions in 12 patients with stable disease were also compared. Serum chromogranin-A (CgA) levels were available for correlation between scans in 17/21 patients. Results Mean thyroid, spleen and liver SUVmax decreased significantly following SSA therapy from a baseline of 5.9 to 3.5, 30.3 to 23.1 and 10.3 to 8.0, respectively (p = < 0.0001 for all). Pituitary SUVmax increased from 10.2 to 11.0 (p = 0.004) whereas adrenal and salivary gland SUVmax did not change. Tumour SUVmax increased in 7 of 12 patients with stable disease; CgA was stable or decreasing in 5 of these patients. 30/49 (61%) metastatic lesions had an increase in SUVmax and lesion-to-liver uptake ratio increased in 40/49 (82%) following SSA therapy. Conclusion Long-acting SSA therapy decreases GaTate uptake in the thyroid, spleen and liver but in most cases increases intensity of uptake within metastases. This has significant implications for interpretation of GaTate PET/CT following commencement of therapy as increased intensity alone may not represent true progression. Our findings also suggest pre-dosing with SSA prior to PRRT may enable higher doses to be delivered to tumour whilst decreasing dose to normal tissues. Electronic supplementary material The online version of this article (10.1186/s40644-018-0136-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Martin H Cherk
- Centre for Cancer Imaging, Peter MacCallum Cancer Centre, 305 Grattan Street, Melbourne, VIC, 3000, Australia.,Department of Medicine, Monash University, Melbourne, Australia.,Department of Nuclear Medicine, The Alfred, 55 Commercial Rd, Prahan, VIC, 3181, Australia
| | - Grace Kong
- Centre for Cancer Imaging, Peter MacCallum Cancer Centre, 305 Grattan Street, Melbourne, VIC, 3000, Australia
| | - Rodney J Hicks
- Centre for Cancer Imaging, Peter MacCallum Cancer Centre, 305 Grattan Street, Melbourne, VIC, 3000, Australia.,Department of Medicine / Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Michael S Hofman
- Centre for Cancer Imaging, Peter MacCallum Cancer Centre, 305 Grattan Street, Melbourne, VIC, 3000, Australia. .,Department of Medicine / Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia.
| |
Collapse
|
18
|
Tian R, Jacobson O, Niu G, Kiesewetter DO, Wang Z, Zhu G, Ma Y, Liu G, Chen X. Evans Blue Attachment Enhances Somatostatin Receptor Subtype-2 Imaging and Radiotherapy. Theranostics 2018; 8:735-745. [PMID: 29344302 PMCID: PMC5771089 DOI: 10.7150/thno.23491] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 11/25/2017] [Indexed: 01/07/2023] Open
Abstract
Purpose: Radionuclide therapy directed against tumors that express somatostatin receptors (SSTRs) has proven effective for the treatment of advanced, low- to intermediate-grade neuroendocrine tumors in the clinic. In clinical usage, somatostatin peptide-based analogs, labeled with therapeutic radionuclides, provide an overall response rate of about 30%, despite the high cumulative activity injected per patient. We set out to improve the effectiveness of somatostatin radiotherapy by preparing a chemical analog that would clear more slowly through the urinary tract and, concomitantly, have increased blood circulation half-life and higher targeted accumulation in the tumors. Experimental Design: We conjugated a common, clinically-used SST peptide derivative, DOTA-octreotate, to an Evans blue analog (EB), which reversibly binds to circulating serum albumin. The resulting molecule was used to chelate 86Y and 90Y, a diagnostic and a therapeutic radionuclide, respectively. The imaging capabilities and the radiotherapeutic efficacy of the resulting radioligand was evaluated in HCT116/SSTR2, HCT116, and AR42J cell lines that express differing levels of SST2 receptors. Results: The synthesized radiopharmaceutical retained affinity and specificity to SSTR2. The new molecule also retained the high internalization rate of DOTA-octreotate, and therefore, showed significantly higher accumulation in SSTR2-positive tumors. Labeling of our novel EB-octreotate derivative with the therapeutic, pure beta emitter, 90Y, resulted in improved tumor response and survival rates of mice bearing SSTR2 xenografts and had long term efficacy when compared to DOTA-octreotate itself. Conclusions: The coupling of a targeted peptide, a therapeutic radionuclide, and the EB‑based albumin binding provides for effective treatment of SSTR2-containing tumors.
Collapse
|
19
|
Spetz J, Langen B, Rudqvist N, Parris TZ, Helou K, Nilsson O, Forssell-Aronsson E. Hedgehog inhibitor sonidegib potentiates 177Lu-octreotate therapy of GOT1 human small intestine neuroendocrine tumors in nude mice. BMC Cancer 2017; 17:528. [PMID: 28789624 PMCID: PMC5549301 DOI: 10.1186/s12885-017-3524-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 08/01/2017] [Indexed: 01/04/2023] Open
Abstract
Background 177Lu-octreotate can be used to treat somatostatin receptor expressing neuroendocrine tumors. It is highly effective in animal models, but clinical studies have so far only demonstrated low cure rates. Hedgehog inhibitors have shown therapeutic effect as monotherapy in neuroendocrine tumor model systems and might be one option to enhance the efficacy of 177Lu-octreotate therapy. The aim of this study was to determine the therapeutic effect of combination therapy using 177Lu-octreotate and the Hedgehog signaling pathway inhibitor sonidegib. Methods GOT1-bearing BALB/c nude mice were treated with either sonidegib (80 mg/kg twice a week via oral gavage), a single injection of 30 MBq 177Lu-octreotate i.v., or a combination of both. Untreated animals served as controls. Tumor size was measured twice-weekly using calipers. The animals were killed 41 d after injection followed by excision of the tumors. Total RNA was extracted from each tumor sample and then subjected to gene expression analysis. Gene expression patterns were compared with those of untreated controls using Nexus Expression 3.0, IPA and Gene Ontology terms. Western blot was carried out on total protein extracted from the tumor samples to analyze activation-states of the Hh and PI3K/AKT/mTOR pathways. Results Sonidegib monotherapy resulted in inhibition of tumor growth, while a significant reduction in mean tumor volume was observed after 177Lu-octreotate monotherapy and combination therapy. Time to progression was prolonged in the combination therapy group compared with 177Lu-octreotate monotherapy. Gene expression analysis revealed a more pronounced response following combination therapy compared with both monotherapies, regarding the number of regulated genes and biological processes. Several cancer-related signaling pathways (i.e. Wnt/β-catenin, PI3K/AKT/mTOR, G-protein coupled receptor, and Notch) were affected by the combination therapy, but not by either monotherapy. Protein expression analysis revealed an activation of the Hh- and PI3K/AKT/mTOR pathways in tumors exposed to 177Lu-octreotate monotherapy and combination therapy. Conclusions A comparative analysis of the different treatment groups showed that combination therapy using sonidegib and 177Lu-octreotate could be beneficial to patients with neuroendocrine tumors. Gene expression analysis revealed a functional interaction between sonidegib and 177Lu-octreotate, i.e. several cancer-related signaling pathways were modulated that were not affected by either monotherapy. Protein expression analysis indicated a possible PI3K/AKT/mTOR-dependent activation of the Hh pathway, independent of SMO. Electronic supplementary material The online version of this article (doi:10.1186/s12885-017-3524-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Johan Spetz
- Department of Radiation Physics, Institute of Clinical Sciences, Sahlgrenska Cancer Center, Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 413 45, Gothenburg, SE, Sweden.
| | - Britta Langen
- Department of Radiation Physics, Institute of Clinical Sciences, Sahlgrenska Cancer Center, Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 413 45, Gothenburg, SE, Sweden
| | - Nils Rudqvist
- Department of Radiation Physics, Institute of Clinical Sciences, Sahlgrenska Cancer Center, Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 413 45, Gothenburg, SE, Sweden
| | - Toshima Z Parris
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Cancer Center, Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 413 45, Gothenburg, SE, Sweden
| | - Khalil Helou
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Cancer Center, Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 413 45, Gothenburg, SE, Sweden
| | - Ola Nilsson
- Department of Pathology, Institute of Biomedicine, Sahlgrenska Cancer Center, Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 413 45, Gothenburg, SE, Sweden
| | - Eva Forssell-Aronsson
- Department of Radiation Physics, Institute of Clinical Sciences, Sahlgrenska Cancer Center, Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 413 45, Gothenburg, SE, Sweden
| |
Collapse
|
20
|
Tian L, Chen Q, Yi X, Chen J, Liang C, Chao Y, Yang K, Liu Z. Albumin-Templated Manganese Dioxide Nanoparticles for Enhanced Radioisotope Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2017; 13:1700640. [PMID: 28544324 DOI: 10.1002/smll.201700640] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Revised: 03/20/2017] [Indexed: 06/07/2023]
Abstract
Although nanoparticle-based drug delivery systems have been widely explored for tumor-targeted delivery of radioisotope therapy (RIT), the hypoxia zones of tumors on one hand can hardly be reached by nanoparticles with relatively large sizes due to their limited intratumoral diffusion ability, on the other hand often exhibit hypoxia-associated resistance to radiation-induced cell damage. To improve RIT treatment of solid tumors, herein, radionuclide 131 I labeled human serum albumin (HSA)-bound manganese dioxide nanoparticles (131 I-HSA-MnO2 ) are developed as a novel RIT nanomedicine platform that is responsive to the tumor microenvironment (TME). Such 131 I-HSA-MnO2 nanoparticles with suitable sizes during blood circulation show rather efficient tumor passive uptake owing to the enhanced permeability and retention effect, as well as little retention in other normal organs to minimize radiotoxicity. The acidic TME can trigger gradual degradation of MnO2 and thus decomposition of 131 I-HSA-MnO2 nanoparticles into individual 131 I-HSA with sub-10 nm sizes and greatly improves intratumoral diffusion. Furthermore, oxygen produced by MnO2 -triggered decomposition of tumor endogenous H2 O2 would be helpful to relieve hypoxia-associated RIT resistant for those tumors. As the results, the 131 I-HSA-MnO2 nanoparticles appear to be a highly effective RIT agent showing great efficacy in tumor treatment upon systemic administration.
Collapse
Affiliation(s)
- Longlong Tian
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Qian Chen
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Xuan Yi
- School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Jiawen Chen
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Chao Liang
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Yu Chao
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Kai Yang
- School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, Jiangsu, 215123, China
| |
Collapse
|
21
|
Pivonello C, Rousaki P, Negri M, Sarnataro M, Napolitano M, Marino FZ, Patalano R, De Martino MC, Sciammarella C, Faggiano A, Rocco G, Franco R, Kaltsas GA, Colao A, Pivonello R. Effects of the single and combined treatment with dopamine agonist, somatostatin analog and mTOR inhibitors in a human lung carcinoid cell line: an in vitro study. Endocrine 2017; 56:603-620. [PMID: 27688013 DOI: 10.1007/s12020-016-1079-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 08/03/2016] [Indexed: 12/14/2022]
Abstract
Somatostatin analogues and mTOR inhibitors have been used as medical therapy in lung carcinoids with variable results. No data are available on dopamine agonists as treatment for lung carcinoids. The main aim of the current study was to evaluate the effect of the combined treatment of somatostatin analogue octreotide and the dopamine agonist cabergoline with mTOR inhibitors in an in vitro model of typical lung carcinoids: the NCI-H727 cell line. In NCI-H727 cell line, reverse transcriptase-quantitative polymerase chain reaction and immunofluorescence were assessed to characterize the expression of the somatostatin receptor 2 and 5, dopamine receptor 2 and mTOR pathway components. Fifteen typical lung carcinoids tissue samples have been used for somatostatin receptor 2, dopamine receptor 2, and the main mTOR pathway component p70S6K expression and localization by immunohistochemistry. Cell viability, fluorescence-activated cell sorting analysis and western blot have been assessed to test the pharmacological effects of octreotide, cabergoline and mTOR inhibitors, and to evaluate the activation of specific cell signaling pathways in NCI-H727 cell line. NCI-H727 cell line expressed somatostatin receptor 2, somatostatin receptor 5 and dopamine receptor 2 and all mTOR pathway components at messenger and protein levels. Somatostatin receptor 2, dopamine receptor 2, and p70S6K (non phosphorylated and phosphorylated) proteins were expressed in most typical lung carcinoids tissue samples. Octreotide and cabergoline did not reduce cell viability as single agents but, when combined with mTOR inhibitors, they potentiate mTOR inhibitors effect after long-term exposure, reducing Akt and ERK phosphorylation, mTOR escape mechanisms, and increasing the expression DNA-damage-inducible transcript 4, an mTOR suppressor. In conclusion, the single use of octreotide and cabergoline is not sufficient to block cell viability but the combined approach of these agents with mTOR inhibitors might reduce the mTOR inhibitors-induced escape mechanisms and/or activate the endogenous mTOR suppressor, potentiating the effect of the mTOR inhibitors in an in vitro model of typical lung carcinoids.
Collapse
Affiliation(s)
- Claudia Pivonello
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, Via Sergio Pansini, 5, Naples, 80131, Italy
| | - Panagoula Rousaki
- Endocrine Unit, Department of Pathophysiology, University of Athens, Medical School, Laiko Hospital, Athens, Greece
| | - Mariarosaria Negri
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, Via Sergio Pansini, 5, Naples, 80131, Italy
| | - Maddalena Sarnataro
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, Via Sergio Pansini, 5, Naples, 80131, Italy
| | - Maria Napolitano
- Immunology Oncology Unit, National Cancer Institute 'G. Pascale' (IRCCS), Naples, Italy
| | | | - Roberta Patalano
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, Via Sergio Pansini, 5, Naples, 80131, Italy
| | - Maria Cristina De Martino
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, Via Sergio Pansini, 5, Naples, 80131, Italy
| | - Concetta Sciammarella
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, Via Sergio Pansini, 5, Naples, 80131, Italy
| | - Antongiulio Faggiano
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, Via Sergio Pansini, 5, Naples, 80131, Italy
| | - Gaetano Rocco
- Division of Thoracic Surgery, Department of Thoracic Surgical and Medical Oncology, National Cancer Institute 'G. Pascale' (IRCCS), Naples, Italy
| | - Renato Franco
- Pathology Unit, Second University of Naples - SUN, Naples, Italy
| | - Gregory A Kaltsas
- Endocrine Unit, Department of Pathophysiology, University of Athens, Medical School, Laiko Hospital, Athens, Greece
| | - Annamaria Colao
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, Via Sergio Pansini, 5, Naples, 80131, Italy
| | - Rosario Pivonello
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, Via Sergio Pansini, 5, Naples, 80131, Italy.
| |
Collapse
|
22
|
Clinical Response Profile of Metastatic/Advanced Pulmonary Neuroendocrine Tumors to Peptide Receptor Radionuclide Therapy with 177Lu-DOTATATE. Clin Nucl Med 2017; 42:428-435. [PMID: 28319500 DOI: 10.1097/rlu.0000000000001639] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
23
|
Pediatric Neuroendocrine Carcinoid Tumors: Review of Diagnostic Imaging Findings and Recent Advances. AJR Am J Roentgenol 2017; 208:868-877. [PMID: 28199138 DOI: 10.2214/ajr.16.17287] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Although rare, pediatric neuroendocrine tumors including carcinoid tumor have increased in incidence over the past few decades. Despite increased recognition, survival rates have not significantly improved. Earlier detection of these tumors is necessary to improve clinical outcomes. This article discusses imaging approaches for detecting pediatric carcinoid tumors including anatomic imaging methods such as CT and MRI as well as functional imaging methods targeting the somatostatin receptor including 111In-labeled octreotide and newer 68Ga-based radiotracers that may hold promise in hybrid PET/CT or PET/MRI. CONCLUSION Improvements in functional imaging with novel somatostatin receptor-specific radiotracers along with fused functional and anatomic imaging have substantially improved the clinical detection of carcinoid tumors. Although rare, these tumors are encountered in children, and an awareness of the appropriate use of various imaging methods is essential for pediatric specialists. Further research is needed to ascertain the diagnostic value of newer imaging methods and radiotracer-based treatment approaches, especially in the pediatric population.
Collapse
|
24
|
Peptide Receptor Radionuclide Therapy Outcomes in a North American Cohort With Metastatic Well-Differentiated Neuroendocrine Tumors. Pancreas 2017; 46:151-156. [PMID: 27759712 PMCID: PMC5595066 DOI: 10.1097/mpa.0000000000000734] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVES The objective of this study was to describe the outcomes of patients in the University of Iowa Neuroendocrine Tumor (NET) Database treated with peptide receptor radionuclide therapy (PRRT). METHODS One hundred thirty-five patients from the University of Iowa NET Database who received PRRT were analyzed, their characteristics were described, and survival was calculated. RESULTS The median age at diagnosis was 51 years, and 64% were men. The primary tumor was located in the small bowel (SBNET) in 37.8%, in the pancreas (PNET) in 26.0%, in the lung in 13.3%, in unknown primary in 9.6%, and in other sites in 13.3%. A radiographic response of any magnitude was observed in 65.8%, 11.1% had a mixed response, and 15.4% showed progression. The overall survival (OS) from the first PRRT was 40 months, and the median time to progression was 23.9 months. Higher pretreatment chromogranin A and pancreastatin levels predicted inferior OS. CONCLUSIONS Peptide receptor radionuclide therapy resulted in a relatively long OS and time to progression in heavily pretreated North American patients with advanced NETs. Elevated pretreatment chromogranin A and pancreastatin predicted shorter OS after therapy. Peptide receptor radionuclide therapy is a valuable treatment option in patients with advanced NETs, especially SBNETS.
Collapse
|
25
|
Hendifar AE, Marchevsky AM, Tuli R. Neuroendocrine Tumors of the Lung: Current Challenges and Advances in the Diagnosis and Management of Well-Differentiated Disease. J Thorac Oncol 2016; 12:425-436. [PMID: 27890494 DOI: 10.1016/j.jtho.2016.11.2222] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 11/08/2016] [Accepted: 11/10/2016] [Indexed: 12/31/2022]
Abstract
Neuroendocrine tumors (NETs) comprise a heterogeneous group of malignancies that arise from neuroendocrine cells throughout the body, most commonly originating from the lungs and gastrointestinal tract. Lung NETs can be classified as well differentiated (low-grade typical carcinoids [TCs] and intermediate-grade atypical carcinoids [ACs]) and poorly differentiated (high-grade large cell neuroendocrine carcinoma or SCLC). The incidence of these tumors is increasing, but disease awareness remains low among thoracic specialists, who are often involved in the diagnosis and early treatment for these patients. An accurate and timely diagnosis can ensure the implementation of appropriate treatment and have a substantial impact on prognosis. However, lung NET classification and diagnosis, particularly for TCs/ACs, are complicated by several factors, including a variable natural history and nonspecific symptoms. Surgery remains the only curative option for TCs/ACs, but there is a lack of consensus between lung NET management guidelines regarding optimal treatment approaches in the unresectable/metastatic setting on account of the limited availability of high-level clinical evidence. As a result, a multidisciplinary approach to management of lung NETs is required to ensure a consistent and optimal level of care. RADIANT-4 is the first phase III trial involving a large subpopulation of patients with advanced well-differentiated lung NETs to report reductions in the risk for disease progression and death with everolimus over placebo. This led to the recent U.S. approval of everolimus-the first agent approved for advanced lung TCs/ACs. To further improve evidence-based care, additional randomized controlled trials in patients with lung carcinoids are needed.
Collapse
Affiliation(s)
- Andrew E Hendifar
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California.
| | - Alberto M Marchevsky
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Richard Tuli
- Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, California
| |
Collapse
|
26
|
Chan HS, Konijnenberg MW, Daniels T, Nysus M, Makvandi M, de Blois E, Breeman WA, Atcher RW, de Jong M, Norenberg JP. Improved safety and efficacy of 213Bi-DOTATATE-targeted alpha therapy of somatostatin receptor-expressing neuroendocrine tumors in mice pre-treated with L-lysine. EJNMMI Res 2016; 6:83. [PMID: 27873240 PMCID: PMC5118228 DOI: 10.1186/s13550-016-0240-5] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2016] [Accepted: 11/15/2016] [Indexed: 02/07/2023] Open
Abstract
Background Targeted alpha therapy (TAT) offers advantages over current β-emitting conjugates for peptide receptor radionuclide therapy (PRRT) of neuroendocrine tumors. PRRT with 177Lu-DOTATATE or 90Y-DOTATOC has shown dose-limiting nephrotoxicity due to radiopeptide retention in the proximal tubules. Pharmacological protection can reduce renal uptake of radiopeptides, e.g., positively charged amino acids, to saturate in the proximal tubules, thereby enabling higher radioactivity to be safely administered. The aim of this preclinical study was to evaluate the therapeutic effect of 213Bi-DOTATATE with and without renal protection using L-lysine in mice. Tumor uptake and kinetics as a function of injected mass of peptide (range 0.03–3 nmol) were investigated using 111In-DOTATATE. These results allowed estimation of the mean radiation absorbed tumor dose for 213Bi-DOTATATE. Pharmacokinetics and dosimetry of 213Bi-DOTATATE was determined in mice, in combination with renal protection. A dose escalation study with 213Bi-DOTATATE was performed to determine the maximum tolerated dose (MTD) with and without pre-administration of l-lysine as for renal protection. Neutrophil gelatinase-associated lipocalin (NGAL) served as renal biomarker to determine kidney injury. Results The maximum mean radiation absorbed tumor dose occurred at 0.03 nmol and the minimum at 3 nmol. Similar mean radiation absorbed tumor doses were determined for 0.1 and 0.3 nmol with a mean radiation absorbed dose of approximately 0.5 Gy/MBq 213Bi-DOTATATE. The optimal mass of injected peptide was found to be 0.3 nmol. Tumor uptake was similar for 111In-DOTATATE and 213Bi-DOTATATE at 0.3 nmol peptide. Lysine reduced the renal uptake of 213Bi-DOTATATE by 50% with no effect on the tumor uptake. The MTD was <13.0 ± 1.6 MBq in absence of l-lysine and 21.7 ± 1.9 MBq with l-lysine renal protection, both imparting an LD50 mean renal radiation absorbed dose of 20 Gy. A correlation was found between the amount of injected radioactivity and NGAL levels. Conclusions The therapeutic potential of 213Bi-DOTATATE was illustrated by significantly decreased tumor burden and improved overall survival. Renal protection with l-lysine immediately prior to TAT with 213Bi-DOTATATE prolonged survival providing substantial evidence for pharmacological nephron blockade to mitigate nephrotoxicity. Electronic supplementary material The online version of this article (doi:10.1186/s13550-016-0240-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ho Sze Chan
- Department of Radiology and Nuclear Medicine, Erasmus MC, 's Gravendijkwal 230, 3015 CE, Rotterdam, The Netherlands.
| | - Mark W Konijnenberg
- Department of Radiology and Nuclear Medicine, Erasmus MC, 's Gravendijkwal 230, 3015 CE, Rotterdam, The Netherlands
| | - Tamara Daniels
- Radiopharmaceutical Sciences Program, College of Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Monique Nysus
- Radiopharmaceutical Sciences Program, College of Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Mehran Makvandi
- Radiopharmaceutical Sciences Program, College of Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Erik de Blois
- Department of Radiology and Nuclear Medicine, Erasmus MC, 's Gravendijkwal 230, 3015 CE, Rotterdam, The Netherlands
| | - Wouter A Breeman
- Department of Radiology and Nuclear Medicine, Erasmus MC, 's Gravendijkwal 230, 3015 CE, Rotterdam, The Netherlands
| | - Robert W Atcher
- Radiopharmaceutical Sciences Program, College of Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, NM, USA.,Los Alamos National Laboratory, Los Alamos, NM, USA
| | - Marion de Jong
- Department of Radiology and Nuclear Medicine, Erasmus MC, 's Gravendijkwal 230, 3015 CE, Rotterdam, The Netherlands
| | - Jeffrey P Norenberg
- Radiopharmaceutical Sciences Program, College of Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| |
Collapse
|
27
|
Koraitim M, Spedding AV, Bradley K, Brennan PA. Widespread metachronous carcinoid tumour metastases to the head and neck: a unique presentation. Br J Oral Maxillofac Surg 2016; 54:1022-1024. [DOI: 10.1016/j.bjoms.2016.01.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 01/02/2016] [Indexed: 11/29/2022]
|
28
|
Lo Russo G, Pusceddu S, Prinzi N, Imbimbo M, Proto C, Signorelli D, Vitali M, Ganzinelli M, Maccauro M, Buzzoni R, Seregni E, de Braud F, Garassino MC. Peptide receptor radionuclide therapy: focus on bronchial neuroendocrine tumors. Tumour Biol 2016; 37:12991-13003. [PMID: 27460087 DOI: 10.1007/s13277-016-5258-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 07/15/2016] [Indexed: 12/29/2022] Open
Abstract
Well-differentiated bronchial neuroendocrine tumors (B-NETs) are rare. They represent 1-5 % of all lung cancers. The incidence of these neoplasms has risen over the past 30 years and, especially for advanced or metastatic disease, management is complex and requires a multidisciplinary approach. Treatment with somatostatin analogs (SSAs) is the most important first-line therapy, in particular in well-differentiated NETs with high somatostatin type receptor (SSTR) expression. In these tumors, the role of mammalian target of rapamycin (m-TOR) inhibitors and the potential utility of other target therapies remain unclear while chemotherapy represents the gold standard treatment only for aggressive forms with low SSTR expression. Peptide receptor radionuclide therapy (PRRT) is an emerging treatment modality for advanced NETs. There are many cumulative evidences about the effectiveness and tolerability of this therapeutic approach, especially in gastro-entero-pancreatic (GEP)-NETs. For B-NETs, scientific research is moving more slowly. Here, we performed a review in order to evaluate the efficacy and toxicity of PRRT with a focus on patients with inoperable or metastatic well-differentiated B-NETs.
Collapse
Affiliation(s)
- Giuseppe Lo Russo
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian 1, Milan, Italy.
| | - Sara Pusceddu
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian 1, Milan, Italy
| | - Natalie Prinzi
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Martina Imbimbo
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian 1, Milan, Italy
| | - Claudia Proto
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian 1, Milan, Italy
| | - Diego Signorelli
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian 1, Milan, Italy
| | - Milena Vitali
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian 1, Milan, Italy
| | - Monica Ganzinelli
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian 1, Milan, Italy
| | - Marco Maccauro
- Department of Nuclear Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Roberto Buzzoni
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian 1, Milan, Italy
| | - Ettore Seregni
- Department of Nuclear Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Filippo de Braud
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, University of Milan, Milan, Italy
| | - Marina Chiara Garassino
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian 1, Milan, Italy
| |
Collapse
|
29
|
Spinelli AE, Schiariti MP, Grana CM, Ferrari M, Cremonesi M, Boschi F. Cerenkov and radioluminescence imaging of brain tumor specimens during neurosurgery. JOURNAL OF BIOMEDICAL OPTICS 2016; 21:50502. [PMID: 27156713 DOI: 10.1117/1.jbo.21.5.050502] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 04/18/2016] [Indexed: 06/05/2023]
Abstract
We presented the first example of Cerenkov luminescence imaging (CLI) and radioluminescence imaging (RLI) of human tumor specimens. A patient with a brain meningioma localized in the left parietal region was injected with 166 MBq of 90Y-DOTATOC the day before neurosurgery. The specimens of the tumor removed during surgery were imaged using both CLI and RLI using an optical imager prototype developed in our laboratory. The system is based on a cooled electron multiplied charge coupled device coupled with an f ∕0.95 17-mm C-mount lens. We showed for the first time the possibility of obtaining CLI and RLI images of fresh human brain tumor specimens removed during neurosurgery.
Collapse
Affiliation(s)
- Antonello Enrico Spinelli
- San Raffaele Scientific Institute, Experimental Imaging Centre, Via Olgettina N. 60, Milan 20182 Italy
| | - Marco P Schiariti
- Neurological Institute C. Besta, Neurosurgery unit 2, Via Celoria 11, Milano 20133, Italy
| | - Chiara M Grana
- European Institute of Oncology, Nuclear Medicine Department, Via Ripamonti 435, Milan 20141, Italy
| | - Mahila Ferrari
- European Institute of Oncology, Medical Physics Unit, Via Ripamonti 435, Milan 20141, Italy
| | - Marta Cremonesi
- European Institute of Oncology, Medical Physics Unit, Via Ripamonti 435, Milan 20141, Italy
| | - Federico Boschi
- University of Verona, Department of Computer Science, Strada Le Grazie 15, Verona 37134, Italy
| |
Collapse
|
30
|
Kumar C, Shetake N, Desai S, Kumar A, Samuel G, Pandey BN. Relevance of radiobiological concepts in radionuclide therapy of cancer. Int J Radiat Biol 2016; 92:173-86. [PMID: 26917443 DOI: 10.3109/09553002.2016.1144944] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE Radionuclide therapy (RNT) is a rapidly growing area of clinical nuclear medicine, wherein radionuclides are employed to deliver cytotoxic dose of radiation to the diseased cells/tissues. During RNT, radionuclides are either directly administered or delivered through biomolecules targeting the diseased site. RNT has been clinically used for diverse range of diseases including cancer, which is the focus of the review. CONCLUSIONS The major emphasis in RNT has so far been given towards developing peptides/antibodies and other molecules to conjugate a variety of therapeutic radioisotopes for improved targeting/delivery of radiation dose to the tumor cells. Despite that, many of the RNT approaches have not achieved their desired therapeutic success probably due to poor knowledge about complex and dynamic (i) fate of radiolabeled molecules; (ii) radiation dose delivered; (iii) cellular heterogeneity in tumor mass; and (iv) cellular radiobiological response. Based on understanding gathered during recent years, it may be stated that besides the absorbed dose, the net radiobiological response of tumor/normal cells also determines the clinical response of radiotherapeutic modalities including RNT. The radiosensitivity of tumor/normal cells is governed by radiobiological phenomenon such as radiation-induced bystander effect, genomic instability, adaptive response and low dose hyper-radiosensitivity. These concepts have been well investigated in the context of external beam radiotherapy, but their clinical implications during RNT have received meagre attention. In this direction, a few studies performed using in vitro and in vivo models envisage the possibilities of exploiting the radiobiological knowledge for improved therapeutic outcome of RNT.
Collapse
Affiliation(s)
- Chandan Kumar
- a Radiopharmaceutical Chemistry Section , Bhabha Atomic Research Centre , Mumbai
| | - Neena Shetake
- b Radiation Biology and Health Sciences Division , Bhabha Atomic Research Centre , Mumbai
| | - Sejal Desai
- b Radiation Biology and Health Sciences Division , Bhabha Atomic Research Centre , Mumbai ;,d Homi Bhabha National Institute , Mumbai , India
| | - Amit Kumar
- b Radiation Biology and Health Sciences Division , Bhabha Atomic Research Centre , Mumbai ;,d Homi Bhabha National Institute , Mumbai , India
| | - Grace Samuel
- c Isotope Production and Applications Division , Bhabha Atomic Research Centre , Mumbai
| | - Badri N Pandey
- b Radiation Biology and Health Sciences Division , Bhabha Atomic Research Centre , Mumbai ;,d Homi Bhabha National Institute , Mumbai , India
| |
Collapse
|
31
|
Pusceddu S, Lo Russo G, Macerelli M, Proto C, Vitali M, Signorelli D, Ganzinelli M, Scanagatta P, Duranti L, Trama A, Buzzoni R, Pelosi G, Pastorino U, de Braud F, Garassino MC. Diagnosis and management of typical and atypical lung carcinoids. Crit Rev Oncol Hematol 2016; 100:167-76. [PMID: 26917456 DOI: 10.1016/j.critrevonc.2016.02.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 02/15/2016] [Indexed: 01/08/2023] Open
Abstract
An estimated 20% to 30% of all neuroendocrine tumours originate in the bronchial tree and lungs. According to the 2015 World Health Organization categorization, these tumours are separated into four subtypes characterized by increasing biological aggressiveness: typical carcinoid, atypical carcinoid, large-cell neuroendocrine carcinoma and small-cell carcinoma. Although typical and atypical lung carcinoids account for less than 1-5% of all pulmonary malignancies, the incidence of these neoplasms has risen significantly in recent decades. Surgery is the treatment of choice for loco-regional disease but for advanced lung carcinoids there is no recognized standard of care and successful management requires a multidisciplinary approach. The aim of this review is to provide a useful guide for the clinical management of lung carcinoids.
Collapse
Affiliation(s)
- Sara Pusceddu
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale di Tumori, Via G. Venezian 1, 20133 Milan, Italy
| | - Giuseppe Lo Russo
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale di Tumori, Via G. Venezian 1, 20133 Milan, Italy
| | - Marianna Macerelli
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale di Tumori, Via G. Venezian 1, 20133 Milan, Italy
| | - Claudia Proto
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale di Tumori, Via G. Venezian 1, 20133 Milan, Italy
| | - Milena Vitali
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale di Tumori, Via G. Venezian 1, 20133 Milan, Italy
| | - Diego Signorelli
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale di Tumori, Via G. Venezian 1, 20133 Milan, Italy
| | - Monica Ganzinelli
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale di Tumori, Via G. Venezian 1, 20133 Milan, Italy
| | - Paolo Scanagatta
- Division of Thoracic Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Via G. Venezian 1, 20133 Milan, Italy
| | - Leonardo Duranti
- Division of Thoracic Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Via G. Venezian 1, 20133 Milan, Italy
| | - Annalisa Trama
- Department of Preventive and Predictive Medicine, Fondazione IRCSS Istituto Nazionale dei Tumori, Via G. Venezian 1, 20133 Milan, Italy
| | - Roberto Buzzoni
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale di Tumori, Via G. Venezian 1, 20133 Milan, Italy
| | - Giuseppe Pelosi
- Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via G. Venezian 1, 20133 Milan, Italy
| | - Ugo Pastorino
- Division of Thoracic Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Via G. Venezian 1, 20133 Milan, Italy
| | - Filippo de Braud
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale di Tumori, Via G. Venezian 1, 20133 Milan, Italy
| | - Marina Chiara Garassino
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale di Tumori, Via G. Venezian 1, 20133 Milan, Italy.
| |
Collapse
|
32
|
Gudkov SV, Shilyagina NY, Vodeneev VA, Zvyagin AV. Targeted Radionuclide Therapy of Human Tumors. Int J Mol Sci 2015; 17:E33. [PMID: 26729091 PMCID: PMC4730279 DOI: 10.3390/ijms17010033] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2015] [Revised: 12/07/2015] [Accepted: 12/22/2015] [Indexed: 12/12/2022] Open
Abstract
Targeted radionuclide therapy is one of the most intensively developing directions of nuclear medicine. Unlike conventional external beam therapy, the targeted radionuclide therapy causes less collateral damage to normal tissues and allows targeted drug delivery to a clinically diagnosed neoplastic malformations, as well as metastasized cells and cellular clusters, thus providing systemic therapy of cancer. The methods of targeted radionuclide therapy are based on the use of molecular carriers of radionuclides with high affinity to antigens on the surface of tumor cells. The potential of targeted radionuclide therapy has markedly grown nowadays due to the expanded knowledge base in cancer biology, bioengineering, and radiochemistry. In this review, progress in the radionuclide therapy of hematological malignancies and approaches for treatment of solid tumors is addressed.
Collapse
Affiliation(s)
- Sergey V Gudkov
- Laboratory of Optical Theranostics, Lobachevsky Nizhny Novgorod State University, Gagarin Ave. 23, Nizhny Novgorod 603950, Russia.
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya St, 3, Pushchino, Moscow 142290, Russia.
- Prokhorov Institute of General Physics, Russian Academy of Sciences, Vavilova St, 38, Moscow 119991, Russia.
| | - Natalya Yu Shilyagina
- Laboratory of Optical Theranostics, Lobachevsky Nizhny Novgorod State University, Gagarin Ave. 23, Nizhny Novgorod 603950, Russia.
| | - Vladimir A Vodeneev
- Laboratory of Optical Theranostics, Lobachevsky Nizhny Novgorod State University, Gagarin Ave. 23, Nizhny Novgorod 603950, Russia.
| | - Andrei V Zvyagin
- Laboratory of Optical Theranostics, Lobachevsky Nizhny Novgorod State University, Gagarin Ave. 23, Nizhny Novgorod 603950, Russia.
- ARC Centre of Excellence for Nanoscale BioPhotonics (CNBP), Macquarie University, Sydney 2109, Australia.
| |
Collapse
|
33
|
Ianniello A, Sansovini M, Severi S, Nicolini S, Grana CM, Massri K, Bongiovanni A, Antonuzzo L, Di Iorio V, Sarnelli A, Caroli P, Monti M, Scarpi E, Paganelli G. Peptide receptor radionuclide therapy with (177)Lu-DOTATATE in advanced bronchial carcinoids: prognostic role of thyroid transcription factor 1 and (18)F-FDG PET. Eur J Nucl Med Mol Imaging 2015; 43:1040-6. [PMID: 26611427 DOI: 10.1007/s00259-015-3262-8] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 11/10/2015] [Indexed: 01/17/2023]
Abstract
PURPOSE Typical and atypical carcinoids (TC and AC) represent 20 - 25 % of all neuroendocrine tumours. No standard therapeutic approach is available for patients with advanced disease. The aim of this phase II study was to investigate the efficacy and safety of peptide receptor radionuclide therapy with (177)Lu-DOTATATE (Lu-PRRT) and the role of thyroid transcription factor 1 (TTF-1) and (18)F-FDG PET as prognostic factors in patients with advanced TC or AC. METHODS A total of 34 consecutive patients with radiologically documented progressive disease were treated with Lu-PRRT at a therapeutic cumulative activity of 18.5 or 27.8 GBq in four or five cycles according to the patient's kidney function and bone marrow reserve. Information on TTF-1 was available in all patients. FDG PET studies prior to Lu-PRRT were available in 29 patients. RESULTS The median follow-up was 29 months (range 7 - 69 months). The disease control rate (DCR) in patients with TC was 80 %: 6 % complete response, 27 % partial response and 47 % stable disease. The median progression-free survival (mPFS) was 20.1 months (95 % CI 11.8 - 26.8 months). Stable disease was achieved in 47 % of patients with AC with a mPFS of 15.7 months (95 % CI 10.6 - 25.9 months). No major acute or delayed toxicity occurred in either group or with either cumulative activity. mPFS in patients with TTF-1-negative TC was 26.3 months (95 % CI 12.9 - 45.2 months), but in patients with TTF-1-positive TC mPFS was 7.2 months (4.2 - 14.0 months; p = 0.0009). FDG PET was negative in 13 patients (10 TC and 3 AC) and positive in 16 patients (4 TC and 12 AC). The mPFS in the FDG PET-negative group was 26.4 months (95 % CI 14.2 - 48.9 months) and 15.3 months (11.7 - 31.1 months) in the FDG PET-positive group. CONCLUSION Lu-PRRT showed antitumour activity in terms of DCR and PFS and proved safe, even in patients with a higher risk of side effects. TTF-1 would appear to be a prognostic factor. FDG PET positivity in bronchial carcinoids is a hallmark of aggressive tumour and is more frequent in patients with AC than in those with TC.
Collapse
Affiliation(s)
- Annarita Ianniello
- Nuclear Medicine and Radiometabolic Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via Maroncelli 40, 47014, Meldola, Italy
| | - Maddalena Sansovini
- Nuclear Medicine and Radiometabolic Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via Maroncelli 40, 47014, Meldola, Italy
| | - Stefano Severi
- Nuclear Medicine and Radiometabolic Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via Maroncelli 40, 47014, Meldola, Italy
| | - Silvia Nicolini
- Nuclear Medicine and Radiometabolic Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via Maroncelli 40, 47014, Meldola, Italy
| | - Chiara Maria Grana
- Division of Nuclear Medicine, European Institute of Oncology Milan (IEO), Milan, Italy
| | - Katrin Massri
- Nuclear Medicine, Department of Radiology, Ospedale San Luca, Lucca, Italy
| | - Alberto Bongiovanni
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | | | - Valentina Di Iorio
- Oncology Pharmacy Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Anna Sarnelli
- Medical Physics Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Paola Caroli
- Nuclear Medicine and Radiometabolic Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via Maroncelli 40, 47014, Meldola, Italy
| | - Manuela Monti
- Unit of Biostatistics and Clinical Trials, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Emanuela Scarpi
- Unit of Biostatistics and Clinical Trials, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Giovanni Paganelli
- Nuclear Medicine and Radiometabolic Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via Maroncelli 40, 47014, Meldola, Italy.
| |
Collapse
|
34
|
Mariniello A, Bodei L, Tinelli C, Baio SM, Gilardi L, Colandrea M, Papi S, Valmadre G, Fazio N, Galetta D, Paganelli G, Grana CM. Long-term results of PRRT in advanced bronchopulmonary carcinoid. Eur J Nucl Med Mol Imaging 2015; 43:441-52. [PMID: 26392198 DOI: 10.1007/s00259-015-3190-7] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 09/04/2015] [Indexed: 11/30/2022]
Abstract
PURPOSE Peptide receptor radionuclide therapy (PRRT) for the treatment of neuroendocrine tumours (NET) has been explored for almost two decades, but there are still few trials that have exclusively investigated well-differentiated and moderately differentiated NET arising from the respiratory tree. Thus, the aim of this study was to explore the outcome in patients affected by bronchopulmonary carcinoid (BPC) following PRRT. METHODS We retrospectively analysed 114 patients with advanced stage BPC consecutively treated with PRRT at the European Institute of Oncology, Milan, from 1997 to 2012 and followed until October 2014. The objective responses, overall survival (OS) and progression-free survival (PFS) were rated, and three different PRRT protocols ((90)Y-DOTATOC vs. (177)Lu-DOTATATE vs. (90)Y-DOTATOC + (177)Lu-DOTATATE) were compared with regard to their efficacy and tolerability. RESULTS The median OS (evaluated in 94 of the 114 patients) was 58.8 months. The median PFS was 28.0 months. The (177)Lu-DOTATATE protocol resulted in the highest 5-year OS (61.4%). Morphological responses (partial responses + minor responses) were obtained in 26.5% of the cohort and were associated with longer OS and PFS. The (90)Y-DOTATOC + (177)Lu-DOTATATE protocol provided the highest response rate (38.1%). Adverse events were mild in the majority of patients. However, haematological toxicity negatively affected survival. No severe (grade 3/4) serum creatinine increase was observed. Patients treated with (90)Y-DOTATOC alone more frequently showed a mild/moderate decrease in renal function. In patients treated with chemotherapy before PRRT had a shorter OS and PFS, and a higher risk of developing nephrotoxicity. CONCLUSION In a large cohort of patients with advanced BPC treated in a "real-world" scenario and followed up for a median of 45.1 months (range 2-191 months), PRRT proved to be promising in prolonging survival and delaying disease progression. Despite the potential selection biases, considering the risk-benefit ratio, (177)Lu-DOTATATE monotherapy seems the best option for PRRT. Our results indicate that the use of PRRT in earlier stages of the disease could provide a more favorable outcome.
Collapse
Affiliation(s)
- Annapaola Mariniello
- Division of Nuclear Medicine, European Institute of Oncology, via Ripamonti 435, 20141, Milan, Italy.
| | - Lisa Bodei
- Division of Nuclear Medicine, European Institute of Oncology, via Ripamonti 435, 20141, Milan, Italy
| | - Carmine Tinelli
- Epidemiology and Biometric Unit, IRCCS Foundation Policlinico San Matteo, Pavia, Italy
| | - Silvia Melania Baio
- Division of Nuclear Medicine, European Institute of Oncology, via Ripamonti 435, 20141, Milan, Italy
| | - Laura Gilardi
- Division of Nuclear Medicine, European Institute of Oncology, via Ripamonti 435, 20141, Milan, Italy
| | - Marzia Colandrea
- Division of Nuclear Medicine, European Institute of Oncology, via Ripamonti 435, 20141, Milan, Italy
| | - Stefano Papi
- Division of Nuclear Medicine, European Institute of Oncology, via Ripamonti 435, 20141, Milan, Italy
| | | | - Nicola Fazio
- Unit of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology, Milan, Italy
| | - Domenico Galetta
- Thoracic Surgery Division, European Institute of Oncology, Milan, Italy
| | - Giovanni Paganelli
- Nuclear Medicine and Radiometabolic Units, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Chiara Maria Grana
- Division of Nuclear Medicine, European Institute of Oncology, via Ripamonti 435, 20141, Milan, Italy
| |
Collapse
|
35
|
Taïeb D, Garrigue P, Bardiès M, Abdullah AE, Pacak K. Application and Dosimetric Requirements for Gallium-68-labeled Somatostatin Analogues in Targeted Radionuclide Therapy for Gastroenteropancreatic Neuroendocrine Tumors. PET Clin 2015; 10:477-86. [PMID: 26384594 DOI: 10.1016/j.cpet.2015.06.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Neuroendocrine tumors (NETs) are associated with variable prognosis, with grade 1 and 2 NETs having more favorable outcomes than grade 3. Patients with gastroenteropancreatic (GEP)-NET need individualized interdisciplinary evaluations and treatment. New treatment options have become available with significant improvements in progression-free survival. Peptide receptor radionuclide therapy (PRRT) using (90)Y or (177)Lu-labeled somatostatin analogues (SSTa) has also shown promise in the treatment of advanced progressive NETs. (68)Ga-1,4,7,10-tetraazacyclodecane-1,4,7,10-tetraacetic acid (DOTA)-SSTa can be used as companion imaging agents to assist in radionuclide therapy selection. (68)Ga-DOTA-SSTa PET/computed tomography might also provide information for prognosis, tumor response assessment to PRRT, and internal dosimetry.
Collapse
Affiliation(s)
- David Taïeb
- Department of Nuclear Medicine, La Timone University Hospital, Aix-Marseille University, 264, rue Saint-Pierre, Marseille 13385, France; European Center for Research in Medical Imaging, Aix-Marseille University, Marseille, France; Marseille Cancerology Research Center, Inserm UMR1068, Institut Paoli-Calmettes, Marseille, France.
| | - Philippe Garrigue
- Department of Radiopharmacy, La Timone University Hospital, Aix-Marseille University, Marseille, France
| | - Manuel Bardiès
- UMR 1037 Inserm/UPS, Cancer Research Center of Toulouse, Toulouse, France
| | - Ahmad Esmaeel Abdullah
- Department of Nuclear Medicine, La Timone University Hospital, Aix-Marseille University, 264, rue Saint-Pierre, Marseille 13385, France
| | - Karel Pacak
- Program in Reproductive and Adult Endocrinology, Eunice Kennedy Shriver National Institute of Child Health & Human Development (NICHD), National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
36
|
Díez JJ, Grande E, Alonso T, Iglesias P. Abordaje multidisciplinar en el diagnóstico y tratamiento de pacientes con tumores endocrinos. Med Clin (Barc) 2015; 145:36-41. [DOI: 10.1016/j.medcli.2014.12.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 11/27/2014] [Accepted: 12/11/2014] [Indexed: 12/30/2022]
|
37
|
Santhanam P, Chandramahanti S, Kroiss A, Yu R, Ruszniewski P, Kumar R, Taïeb D. Nuclear imaging of neuroendocrine tumors with unknown primary: why, when and how? Eur J Nucl Med Mol Imaging 2015; 42:1144-55. [DOI: 10.1007/s00259-015-3027-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 02/19/2015] [Indexed: 01/22/2023]
|