101
|
Zamponi V, La Salvia A, Tarsitano MG, Mikovic N, Rinzivillo M, Panzuto F, Giannetta E, Faggiano A, Mazzilli R. Effect of Neuroendocrine Neoplasm Treatment on Human Reproductive Health and Sexual Function. J Clin Med 2022; 11:jcm11143983. [PMID: 35887747 PMCID: PMC9324753 DOI: 10.3390/jcm11143983] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 07/06/2022] [Accepted: 07/07/2022] [Indexed: 02/02/2023] Open
Abstract
Neuroendocrine neoplasms (NEN) are characterized by a wide clinical heterogeneity and biological variability, with slow progression and long survival in most cases. Although these tumors can affect young adults, there are few studies that focus on the sexual and reproductive system. The aim of this review was to evaluate the effect of NEN treatment, including somatostatin analogues (SSA), targeted therapy (Everolimus and Sunitinib), radiolabeled-SSA and chemotherapy, on male and female reproductive systems and sexual function. This narrative review was performed for all available prospective and retrospective studies, case reports and review articles published up to March 2022 in PubMed. To date, few data are available on the impact of SSA on human fertility and most of studies come from acromegalic patients. However, SSAs seem to cross the blood–placental barrier; therefore, pregnancy planning is strongly recommended. Furthermore, the effect of targeted therapy on reproductive function is still undefined. Conversely, chemotherapy has a well-known negative impact on male and female fertility. The effect of temozolomide on reproductive function is still undefined, even if changes in semen parameters after the treatment have been described. Finally, very few data are available on the sexual function of NEN treatment.
Collapse
Affiliation(s)
- Virginia Zamponi
- Department of Clinical and Molecular Medicine, ENETS Center of Excellence, Sapienza University of Rome, 00185 Rome, Italy; (V.Z.); (N.M.); (A.F.); (R.M.)
| | - Anna La Salvia
- Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
- Correspondence: ; Tel.: +39-0652665698
| | - Maria Grazia Tarsitano
- Department of Medical and Surgical Science, University Magna Graecia, 88100 Catanzaro, Italy;
| | - Nevena Mikovic
- Department of Clinical and Molecular Medicine, ENETS Center of Excellence, Sapienza University of Rome, 00185 Rome, Italy; (V.Z.); (N.M.); (A.F.); (R.M.)
| | - Maria Rinzivillo
- Digestive Disease Unit, ENETS Center of Excellence, Sant’Andrea University Hospital, 00189 Rome, Italy; (M.R.); (F.P.)
| | - Francesco Panzuto
- Digestive Disease Unit, ENETS Center of Excellence, Sant’Andrea University Hospital, 00189 Rome, Italy; (M.R.); (F.P.)
- Department of Medical-Surgical Sciences and Translational Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Elisa Giannetta
- Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy;
| | - Antongiulio Faggiano
- Department of Clinical and Molecular Medicine, ENETS Center of Excellence, Sapienza University of Rome, 00185 Rome, Italy; (V.Z.); (N.M.); (A.F.); (R.M.)
| | - Rossella Mazzilli
- Department of Clinical and Molecular Medicine, ENETS Center of Excellence, Sapienza University of Rome, 00185 Rome, Italy; (V.Z.); (N.M.); (A.F.); (R.M.)
| |
Collapse
|
102
|
Beykan S, Tran-Gia J, Borup Jensen S, Lassmann M. Is a single late SPECT/CT based kidney 177Lu-dosimetry superior to hybrid dosimetry with sequential multiple time-point whole-body planar scans in combination with an early SPECT/CT? Phys Med 2022; 100:39-50. [PMID: 35724608 DOI: 10.1016/j.ejmp.2022.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 05/25/2022] [Accepted: 06/11/2022] [Indexed: 11/18/2022] Open
Abstract
PURPOSE The aim is to assess the impact of different imaging-protocols on image-based kidney dosimetry in 177Lu labelled peptide receptor radiotherapies. METHODS Kidney data of five [177Lu]Lu-OPS201 injected pigs and a 3D printed phantom were used for comparing the absorbed doses and time-integrated activity coefficients calculated based on the following imaging-protocols: A-) multiple time-point SPECT/CTs, B-) multiple time-point planar scans in combination with one SPECT/CT, C-) single time-point SPECT/CT. In addition, the influence of late scan time-points on kidney dosimetry was investigated by sequentially eliminating scan data at > 100 h from the pig/phantom datasets for imaging-protocols A and B. RESULTS Compared to imaging-protocol A, absorbed doses based on imaging-protocols B and C (scans at > 24 h post-injection) were always lower (differences > 34%). The best agreement in absorbed dose was achieved by imaging-protocol C at ∼ 100 h post-injection (difference: 4%). Regarding the phantom/pig experiments, eliminating scan data at > 100 h post-injection increased the time-integrated activity coefficients calculated based on imaging-protocols A and B by up to 83%. CONCLUSION While imaging-protocol A is accurate if scans at >∼100 h are included, it is time-consuming. In addition to being time-consuming, imaging-protocol B shows high differences associated with organ-count overlay, a lack of accuracy concerning the geometric mean based 2D attenuation correction, and 2D background subtraction due to the inhomogeneous and time-varying background contributions. Our findings indicate that dosimetry based on imaging-protocol C, if appropriately performed, provides similar kidney absorbed doses compared to imaging-protocol A, while only a single scan time-point is necessary.
Collapse
Affiliation(s)
- Seval Beykan
- Department of Nuclear Medicine, University of Würzburg, Würzburg, Germany.
| | - Johannes Tran-Gia
- Department of Nuclear Medicine, University of Würzburg, Würzburg, Germany
| | - Svend Borup Jensen
- Department of Nuclear Medicine, Aalborg University Hospital, Aalborg, Denmark; Department of Chemistry and Bioscience, Aalborg University, Aalborg, Denmark
| | - Michael Lassmann
- Department of Nuclear Medicine, University of Würzburg, Würzburg, Germany
| |
Collapse
|
103
|
Aalbersberg EA, de Vries-Huizing DMV, Tesselaar MET, Stokkel MPM, Versleijen MWJ. Post-PRRT scans: which scans to make and what to look for. Cancer Imaging 2022; 22:29. [PMID: 35715867 PMCID: PMC9205039 DOI: 10.1186/s40644-022-00467-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 05/26/2022] [Indexed: 11/15/2022] Open
Abstract
Aim The aim of this study was to evaluate the clinical utility of SPECT/CT (imaging of uptake in tumor lesions and additional findings) and the additional value of planar imaging in order to simplify clinical imaging protocols and decrease patients burden. Materials and methods One hundred consecutive patients with metastatic neuroendocrine tumor (NET) treated with PRRT were included. Post-therapy imaging was performed 24 h after each PRRT cycle by both whole-body planar imaging and abdominal- and thoracic SPECT/CT. All images were evaluated for (1) the presence of new lesions, (2) discordant lesions between the two acquisitions (planar or SPECT), (3) location of lesions on SPECT (abdominal, thoracic, or both), and (4) additional findings on non-contrast enhanced CT imaging. Results In total 368 PRRT cycles including post-therapy imaging were performed in 100 patients. 45 patients had abdominal disease only, whilst in 55 patients the disease was observed on both abdominal and thoracic SPECT. 16 patients had known bone lesions that were visible only on planar imaging as these were out of range of the SPECT/CT. During PRRT, one patient developed multiple new bone metastases after the second cycle of PRRT, which were visible on both planar and SPECT/CT images. In 11 patients additional findings were found on CT images, the most common and relevant being bowel obstruction, pleural effusion, and ascites. Patients who developed ascites during PRRT appeared to do extremely poor; a post-hoc analysis showed that overall survival was 13.2 months in patients that showed ascites during PRRT at any moment and 37.9 months in patients without ascites (p < 0.001). Conclusion From a clinical point of view, thoracoabdominal SPECT/CT imaging is the preferred method for post-PRRT imaging; planar imaging had no added value over SPECT/CT in this cohort. In patients with abdominal disease only on baseline imaging, SPECT/CT of the abdomen only might be sufficient for imaging during the PRRT course. All accompanying CT images should be reviewed for additional findings, especially ascites, which is suggested to be a poor prognostic factor in patients receiving PRRT.
Collapse
Affiliation(s)
- Else A Aalbersberg
- Department of Nuclear Medicine, Plesmanlaan 121, 1066 CX, Amsterdam, the Netherlands.
| | | | - Margot E T Tesselaar
- Department of Medical Oncology, Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - Marcel P M Stokkel
- Department of Nuclear Medicine, Plesmanlaan 121, 1066 CX, Amsterdam, the Netherlands
| | | |
Collapse
|
104
|
Striking Size Reduction of Rapidly Growing Pancreatic Neuroendocrine Carcinoma Metastatic Nodal Conglomerate After Only 2 Cycles of 177Lu-DOTATATE. Clin Nucl Med 2022; 47:961-962. [PMID: 35695695 DOI: 10.1097/rlu.0000000000004262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
ABSTRACT Peptide receptor radionuclide therapy (PRRT) with 177Lu-DOTATATE has shown great treatment efficacy in patients with well-differentiated metastatic neuroendocrine tumors and a metastatic size reduction of ~20% for metastatic lesions <3 cm in size. We present a 66-year-old man with pancreatic neuroendocrine carcinoma, who had a rapidly growing metastatic nodal conglomerate, which measured close to 10 cm in size. After only 2 cycles of PRRT with 177Lu-DOTATATE, the nodal conglomerate had a striking size reduction greater than 75%. This case highlights the potential efficacy of PRRT with 177Lu-DOTATATE for treatment of aggressive neuroendocrine neoplasms.
Collapse
|
105
|
Cigrovski Berkovic M, Ulamec M, Marinovic S, Balen I, Mrzljak A. Malignant insulinoma: Can we predict the long-term outcomes? World J Clin Cases 2022; 10:5124-5132. [PMID: 35812675 PMCID: PMC9210919 DOI: 10.12998/wjcc.v10.i16.5124] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 01/17/2022] [Accepted: 04/21/2022] [Indexed: 02/06/2023] Open
Abstract
Insulinomas are the most frequent type of functional pancreatic neuroendocrine tumors with a variety of neuroglycopenic and autonomic symptoms and well-defined diagnostic criteria; however, prediction of their clinical behavior and early differentiation between benign and malignant lesions remain a challenge. The comparative studies between benign and malignant cases are limited, suggesting that short clinical history, early hypoglycemia during fasting, high proinsulin, insulin, and C-peptide concentrations raise suspicion of malignancy. Indeed, malignant tumors are larger with higher mitotic count and Ki-67 proliferative activity, but there are no accurate histological criteria to distinguish benign from malignant forms. Several signaling pathways have been suggested to affect the pathophysiology and behavior of insulinomas; however, our knowledge is limited, urging a further understanding of molecular genetics. Therefore, there is a need for the identification of reliable markers of metastatic disease that could also serve as therapeutic targets in patients with malignant insulinoma. This opinion review reflects on current gaps in diagnostic and clinical aspects related to the malignant behavior of insulinoma.
Collapse
Affiliation(s)
- Maja Cigrovski Berkovic
- Department of Endocrinology, Diabetes, Metabolism and Clinical Pharmacology, Clinical Hospital Dubrava, Zagreb 10000, Croatia
- Department of Kinesiological Anthropology and Methodology, Faculty of Kinesiology, University of Zagreb, Zagreb 10000, Croatia
| | - Monika Ulamec
- Department of Pathology and Cytology “Ljudevit Jurak”, University Hospital Center “Sestre milosrdnice”, Zagreb 10000, Croatia
- Scientific Group for Research on Epigenetic Biomarkers and Department of Pathology, School of Medicine, University of Zagreb, Zagreb 10000, Croatia
| | - Sonja Marinovic
- Laboratory for Personalized Medicine, Division of Molecular Medicine, Rudjer Boskovic Institute, Zagreb 10000, Croatia
| | - Ivan Balen
- Department of Gastroenterology and Endocrinology, General Hospital “Dr. Josip Bencevic”, Slavonski Brod 35000, Croatia
| | - Anna Mrzljak
- Department of Gastroenterology and Hepatology, UHC Zagreb, Zagreb 10000, Croatia
- School of Medicine, University of Zagreb, Zagreb 10000, Croatia
| |
Collapse
|
106
|
Muros MA, Aroui T, Rivas-Navas D, Fernandez-Fernadez J. Integration of molecular imaging in the personalized approach to neuroendocrine tumors. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF... 2022; 66:116-129. [PMID: 35238519 DOI: 10.23736/s1824-4785.22.03431-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
NETs lesions can be difficult to characterize with conventional anatomic imaging (CT and MRI). Functional imaging techniques, and especially PET imaging, are very useful for detecting small neuroendocrine tumors that would not be seen with other techniques. The role of nuclear medicine in the localization, staging, restaging, treatment and monitoring of neuroendocrine tumors (NETs) has become progressively more relevant due to: the availability of tracers on new targets, tracers for positron emission tomography (PET); the development of cyclotrons and generators that allow this availability; as well as to hybrid systems (SPECT/CT, PET/CT and PET/MRI) that, by joining the functional and anatomical image, improve the quality of the images. Teragnosis, a new emerging therapy, in NET used receptor-mediated or nonreceptor- mediated mechanism to facilitate penetration and high-affinity binding between the radiopharmaceutical and the tumor cell. Teragnosis offers the possibility of personalized targeted radionuclide therapy.
Collapse
Affiliation(s)
- Maria A Muros
- Department of Nuclear Medicine, Virgen de las Nieves Hospital, Granada, Spain -
| | - Tarik Aroui
- Department of Nuclear Medicine, Virgen de las Nieves Hospital, Granada, Spain
| | - Daniel Rivas-Navas
- Department of Nuclear Medicine, Virgen de las Nieves Hospital, Granada, Spain
| | | |
Collapse
|
107
|
Herrmann K, Giovanella L, Santos A, Gear J, Kiratli PO, Kurth J, Denis-Bacelar AM, Hustinx R, Patt M, Wahl RL, Paez D, Giammarile F, Jadvar H, Pandit-Taskar N, Ghesani M, Kunikowska J. Joint EANM, SNMMI and IAEA enabling guide: how to set up a theranostics centre. Eur J Nucl Med Mol Imaging 2022; 49:2300-2309. [PMID: 35403861 PMCID: PMC9165261 DOI: 10.1007/s00259-022-05785-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 03/25/2022] [Indexed: 01/14/2023]
Abstract
The theranostics concept using the same target for both imaging and therapy dates back to the middle of the last century, when radioactive iodine was first used to treat thyroid diseases. Since then, radioiodine has become broadly established clinically for diagnostic imaging and therapy of benign and malignant thyroid disease, worldwide. However, only since the approval of SSTR2-targeting theranostics following the NETTER-1 trial in neuroendocrine tumours and the positive outcome of the VISION trial has theranostics gained substantial attention beyond nuclear medicine. The roll-out of radioligand therapy for treating a high-incidence tumour such as prostate cancer requires the expansion of existing and the establishment of new theranostics centres. Despite wide global variation in the regulatory, financial and medical landscapes, this guide attempts to provide valuable information to enable interested stakeholders to safely initiate and operate theranostics centres. This enabling guide does not intend to answer all possible questions, but rather to serve as an overarching framework for multiple, more detailed future initiatives. It recognizes that there are regional differences in the specifics of regulation of radiation safety, but common elements of best practice valid globally.
Collapse
Affiliation(s)
- Ken Herrmann
- Department of Nuclear Medicine, University of Duisburg-Essen, Duisburg, Germany.
- German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany.
| | - Luca Giovanella
- Clinic for Nuclear Medicine and Molecular Imaging, Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Andrea Santos
- Department of Nuclear Medicine, Hospital Cuf Descobertas, Lisbon, Portugal
| | - Jonathan Gear
- Joint Department of Physics, Royal Marsden NHS Foundation Trust, Sutton, UK
| | | | - Jens Kurth
- Department of Nuclear Medicine, Rostock University Medical Center, Rostock, Germany
| | | | - Roland Hustinx
- Division of Nuclear Medicine and Oncological Imaging, University Hospital of Liège, Liège, Belgium
- GIGA-CRC in vivo imaging, University of Liège, Liège, Belgium
| | - Marianne Patt
- Department for Nuclear Medicine, University Hospital Leipzig, Leipzig, Germany
| | - Richard L Wahl
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Diana Paez
- Nuclear Medicine and Diagnostic Imaging Section, Division of Human Health, Department of Nuclear Sciences and Application, International Atomic Energy Agency, Vienna, Austria
| | - Francesco Giammarile
- Nuclear Medicine and Diagnostic Imaging Section, Division of Human Health, Department of Nuclear Sciences and Application, International Atomic Energy Agency, Vienna, Austria
| | - Hossein Jadvar
- Division of Nuclear Medicine, Department of Radiology, University of Southern California, Los Angeles, CA, USA
| | - Neeta Pandit-Taskar
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Munir Ghesani
- Diagnostic, Molecular & Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jolanta Kunikowska
- Nuclear Medicine Department, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
108
|
Love C, Desai NB, Abraham T, Banks KP, Bodei L, Boike T, Brown RKJ, Bushnell DL, DeBlanche LE, Dominello MM, Francis T, Grady EC, Hobbs RF, Hope TA, Kempf JS, Pryma DA, Rule W, Savir-Baruch B, Sethi I, Subramaniam RM, Xiao Y, Schechter NR. ACR-ACNM-ASTRO-SNMMI Practice Parameter for Lutetium-177 (Lu-177) DOTATATE Therapy. Am J Clin Oncol 2022; 45:233-242. [PMID: 35507413 DOI: 10.1097/coc.0000000000000903] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES This practice parameter (PP) for Lutetium-177 (Lu-177) DOTATATE peptide receptor radionuclide therapy (PRRT) aims to guide authorized users in selection of appropriate adult candidates with gastroeneropancreatic neuroendocrine tumors (GEP-NETs) from foregut, midgut, and hindgut. The essential selection criteria include somatostatin receptor-positive GEP-NETs, which are usually inoperable and progressed despite standard therapy. Lu-177 DOTATATE is a radiopharmaceutical with high avidity for somatostatin receptors that are overexpressed by these tumors. This document ensures safe handling of Lu-177 DOTATATE by the authorized users and safe management of affected patients. METHODS The document was developed according to the systematic process developed by the American College of Radiology (ACR) and described on the ACR Web site (https://www.acr.org/Clinical-Resources/Practice-Parameters-and-Technical-Standards). The PP development was led by 2 ACR Committees on Practice Parameters (Nuclear Medicine and Molecular Imaging and Radiation Oncology) collaboratively with the American College of Nuclear Medicine, American Society of Radiation Oncology, and Society of Nuclear Medicine and Molecular Imaging. RESULTS The Lu-177 DOTATATE PP reviewed pharmacology, indications, adverse effects, personnel qualifications, and required clinical evaluation before starting the treatment, as well as the recommended posttherapy monitoring, quality assurance, documentation, and appropriate radiation safety instructions provided in written form and explained to the patients. CONCLUSIONS Lu-177 DOTATATE is available for therapy of inoperable and/or advanced GEP-NETs when conventional therapy had failed. It can reduce tumor size, improve symptoms, and increase the progression free survival. The PP document provides clinical guidance for authorized users to assure an appropriate, consistent, and safe practice of Lu-177 DOTATATE.
Collapse
Affiliation(s)
- Charito Love
- Albert Einstein College of Medicine, New York
- New York Cancer and Blood Specialists, Port Jefferson Station, NY
| | | | - Tony Abraham
- New York Cancer and Blood Specialists, Port Jefferson Station, NY
| | - Kevin P Banks
- Uniformed Services University, Bethesda, MD
- San Antonio Military Medical Center, San Antonio, TX
| | - Lisa Bodei
- Memorial Sloan Kettering Cancer Center, New York, NY
| | - Thomas Boike
- Genesis Care USA/MHP Radiation Oncology, Asheville, NC
| | - Richard K J Brown
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT
| | | | | | | | | | - Erin C Grady
- Emory University School of Medicine, Atlanta, GA
| | | | - Thomas A Hope
- University of California, San Francisco, San Francisco, CA
| | - Jeffrey S Kempf
- Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Daniel A Pryma
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | | | | | - Ila Sethi
- Emory University School of Medicine, Atlanta, GA
| | | | - Ying Xiao
- University of Pennsylvania, Philadelphia, PA
| | - Naomi R Schechter
- Keck Medical Center of USC, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA
| |
Collapse
|
109
|
Hartrampf P, Werner R, Buck A. Theranostics bei gut bis mäßig differenzierten GEP-NEN. Zentralbl Chir 2022; 147:249-255. [DOI: 10.1055/a-1826-3423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
ZusammenfassungNeuroendokrine Neoplasien (NEN) sind seltene, heterogene und typischerweise langsam wachsende Tumoren. Die häufigsten Lokalisationen finden sich im gastro-entero-pankreatischen System
(GEP-NEN). NENs werden nach proliferativer Aktivität (Ki-67-Index) eingeteilt (G1–3). Gut differenzierte Tumoren exprimieren dabei typischerweise Somatostatinrezeptoren (SSTR), die als
Zielstruktur in der nuklearmedizinischen Theranostik dienen. Bei diesem Prinzip kann nach einer diagnostischen molekularen Bildgebung, meist mittels
Positronenemissionstomografie/Computertomografie (PET/CT), eine individuell zugeschnittene Peptidradiorezeptortherapie (PRRT) mit einem β-Strahler-markierten Radiopharmakon erfolgen. In
Metaanalysen zeigte die Diagnostik mittels SSTR-gerichteter PET/CT eine Sensitivität von 93% und eine Spezifität von 96%. Die SSTR-gerichtete Diagnostik kann auch zur radioaktiven Markierung
von Tumoren verwendet werden, um eine zielgerichtete Chirurgie zu ermöglichen. Die Indikation zur Einleitung einer PRRT soll stets in einer interdisziplinären Tumorkonferenz getroffen
werden. Ein Tumorprogress unter der vorangegangenen Therapie sollte dokumentiert sein. Die Therapie wird intravenös und insgesamt 4-mal in 8-wöchigem Abstand in spezialisierten
nuklearmedizinischen Zentren verabreicht. Die Wirksamkeit der PRRT wurde in der NETTER-1-Studie prospektiv untersucht und konnte eine signifikante Verbesserung des progressionsfreien
Überlebens (primärer Endpunkt) zeigen. Ausgehend von diesen Studienergebnissen steht mit Lutathera (177Lu-DOTATATE) inzwischen ein in Deutschland zugelassenes Radiopharmazeutikum zu
Behandlung von nicht resektablen oder metastasierten bzw. progredienten, gut differenzierten (G1 und G2), SSTR-positiven GEP-NEN zur Verfügung.
Collapse
Affiliation(s)
- Philipp Hartrampf
- Klinik und Poliklinik für Nuklearmedizin, Universitätsklinikum Würzburg, Würzburg, Deutschland
| | - Rudolf Werner
- Klinik und Poliklinik für Nuklearmedizin, Universitätsklinikum Würzburg, Würzburg, Deutschland
| | - Andreas Buck
- Klinik und Poliklinik für Nuklearmedizin, Universitätsklinikum Würzburg, Würzburg, Deutschland
| |
Collapse
|
110
|
Love C, Desai NB, Abraham T, Banks KP, Bodei L, Boike T, Brown RKJ, Bushnell DL, DeBlanche LE, Dominello MM, Francis T, Grady EC, Hobbs RF, Hope TA, Kempf JS, Pryma DA, Rule W, Savir-Baruch B, Sethi I, Subramaniam RM, Xiao Y, Schechter NR. ACR-ACNM-ASTRO-SNMMI Practice Parameter for Lutetium-177 (Lu-177) DOTATATE Therapy. Clin Nucl Med 2022; 47:503-511. [PMID: 35507433 DOI: 10.1097/rlu.0000000000004182] [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: 11/26/2022]
Abstract
OBJECTIVES This practice parameter (PP) for Lutetium-177 (Lu-177) DOTATATE peptide receptor radionuclide therapy (PRRT) aims to guide authorized users in selection of appropriate adult candidates with gastroeneropancreatic neuroendocrine tumors (GEP-NETs) from foregut, midgut, and hindgut. The essential selection criteria include somatostatin receptor-positive GEP-NETs, which are usually inoperable and progressed despite standard therapy. Lu-177 DOTATATE is a radiopharmaceutical with high avidity for somatostatin receptors that are overexpressed by these tumors. This document ensures safe handling of Lu-177 DOTATATE by the authorized users and safe management of affected patients. METHODS The document was developed according to the systematic process developed by the American College of Radiology (ACR) and described on the ACR Web site (https://www.acr.org/Clinical-Resources/Practice-Parameters-and-Technical-Standards). The PP development was led by 2 ACR Committees on Practice Parameters (Nuclear Medicine and Molecular Imaging and Radiation Oncology) collaboratively with the American College of Nuclear Medicine, American Society of Radiation Oncology, and Society of Nuclear Medicine and Molecular Imaging. RESULTS The Lu-177 DOTATATE PP reviewed pharmacology, indications, adverse effects, personnel qualifications, and required clinical evaluation before starting the treatment, as well as the recommended posttherapy monitoring, quality assurance, documentation, and appropriate radiation safety instructions provided in written form and explained to the patients. CONCLUSIONS Lu-177 DOTATATE is available for therapy of inoperable and/or advanced GEP-NETs when conventional therapy had failed. It can reduce tumor size, improve symptoms, and increase the progression free survival. The PP document provides clinical guidance for authorized users to assure an appropriate, consistent, and safe practice of Lu-177 DOTATATE.
Collapse
Affiliation(s)
| | | | - Tony Abraham
- New York Cancer and Blood Specialists, Port Jefferson Station, NY
| | | | - Lisa Bodei
- Memorial Sloan Kettering Cancer Center, New York, NY
| | - Thomas Boike
- Genesis Care USA/MHP Radiation Oncology, Asheville, NC
| | - Richard K J Brown
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT
| | | | | | | | | | - Erin C Grady
- Emory University School of Medicine, Atlanta, GA
| | | | - Thomas A Hope
- University of California, San Francisco, San Francisco, CA
| | - Jeffrey S Kempf
- Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Daniel A Pryma
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | | | | | - Ila Sethi
- Emory University School of Medicine, Atlanta, GA
| | | | - Ying Xiao
- University of Pennsylvania, Philadelphia, PA
| | - Naomi R Schechter
- Keck Medical Center of USC, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA
| |
Collapse
|
111
|
Syguła A, Ledwon A, Hasse-Lazar K, Jurecka-Lubieniecka B, Michalik B, Paliczka-Cieślik E, Zeman M, Chmielik E, Sczasny J, Jarzab B, Handkiewicz-Junak D. In patients with well-differentiated neuroendocrine tumours, there is no apparent benefit of somatostatin analogues after disease control by peptide receptor radionuclide therapy. Eur J Nucl Med Mol Imaging 2022; 49:3841-3851. [PMID: 35503379 DOI: 10.1007/s00259-022-05792-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 04/03/2022] [Indexed: 12/13/2022]
Abstract
PURPOSE Peptide receptor radionuclide therapy (PRRT) and somatostatin analogues (SSAs) are commonly combined as primary treatment for neuroendocrine neoplasms (NEN), and SSAs given as maintenance. We sought to evaluate whether sequential therapy with PRRT followed by SSAs has progression or survival benefits in patients with NEN after disease control by PRRT. METHODS This prospective, randomised, single-centre study had as principal eligibility criteria: unresectable, locally advanced, or metastatic, histologically confirmed well-differentiated NEN; no symptoms/biochemical diagnosis of carcinoid syndrome; no SSAs or ≤ 3 months of SSAs before PRRT; and stable disease or partial or complete response after PRRT. Altogether, 115 patients were randomised 2:1 to an SSA group (n = 74) given octreotide acetate LAR every 4 weeks, or a control group (n = 41) receiving only best supportive care. Octreotide treatment was to stop upon intolerable toxicity or patient refusal, or, at physician/patient discretion, upon NEN progression. The primary endpoint was progression-free survival (PFS), the secondary endpoint, and overall survival (OS). RESULTS Median (25th-75th percentile) follow-up from the first PRRT activity to death or latest observation was 6.6 (3.18-10.22) years. During that time, 71/115 patients (62%) progressed, 52/74 (70%) in the SSA group, and 19/41 (46%) in the control group (p = 0.01). Eighty-eight/115 patients (76%) died, 58/74 (78%) in the SSA group, and 30/41 (73%) in the control group (p = 0.52). Median (95% CI) PFS was 4.7 (2.8-7.7) years in the SSA group, and 6.4 (4.1-not reached) years in controls. Overall, median OS was 6.6 years. Neither PFS nor OS differed between groups (p = 0.129, p = 0.985, respectively). CONCLUSIONS In patients with disease control after PRRT, subsequent SSA treatment appeared not to be associated with better PFS or OS. Whether to continue SSA administration upon progression after PRRT requires evaluation in a prospective, randomised, controlled multicentre study with a relatively homogeneous sample.
Collapse
Affiliation(s)
- Aleksandra Syguła
- Department of Nuclear Medicine and Endocrine Oncology, Gliwice Branch, Maria Sklodowska-Curie National Research Institute of Oncology, Wybrzeże Armii Krajowej 16, 44-101, Gliwice, Poland
| | - Aleksandra Ledwon
- Department of Nuclear Medicine and Endocrine Oncology, Gliwice Branch, Maria Sklodowska-Curie National Research Institute of Oncology, Wybrzeże Armii Krajowej 16, 44-101, Gliwice, Poland
| | - Kornelia Hasse-Lazar
- Department of Nuclear Medicine and Endocrine Oncology, Gliwice Branch, Maria Sklodowska-Curie National Research Institute of Oncology, Wybrzeże Armii Krajowej 16, 44-101, Gliwice, Poland
| | - Beata Jurecka-Lubieniecka
- Department of Nuclear Medicine and Endocrine Oncology, Gliwice Branch, Maria Sklodowska-Curie National Research Institute of Oncology, Wybrzeże Armii Krajowej 16, 44-101, Gliwice, Poland
| | - Barbara Michalik
- Department of Nuclear Medicine and Endocrine Oncology, Gliwice Branch, Maria Sklodowska-Curie National Research Institute of Oncology, Wybrzeże Armii Krajowej 16, 44-101, Gliwice, Poland
| | - Ewa Paliczka-Cieślik
- Department of Nuclear Medicine and Endocrine Oncology, Gliwice Branch, Maria Sklodowska-Curie National Research Institute of Oncology, Wybrzeże Armii Krajowej 16, 44-101, Gliwice, Poland
| | - Marcin Zeman
- The Oncologic and Reconstructive Surgery Clinic, Gliwice Branch, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, Poland
| | - Ewa Chmielik
- Tumor Pathology Department, Gliwice Branch, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, Poland
| | - Joanna Sczasny
- Radiology and Diagnostic Imaging Department, Gliwice Branch, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, Poland
| | - Barbara Jarzab
- Department of Nuclear Medicine and Endocrine Oncology, Gliwice Branch, Maria Sklodowska-Curie National Research Institute of Oncology, Wybrzeże Armii Krajowej 16, 44-101, Gliwice, Poland
| | - Daria Handkiewicz-Junak
- Department of Nuclear Medicine and Endocrine Oncology, Gliwice Branch, Maria Sklodowska-Curie National Research Institute of Oncology, Wybrzeże Armii Krajowej 16, 44-101, Gliwice, Poland.
| |
Collapse
|
112
|
Evaluation of radiation safety of the newly established national New Zealand 177–Lutetium (177-Lu or Lutate) Peptide Receptor Radiation Therapy (PRRT) service, a palliative treatment for patients with metastatic neuroendocrine tumours. J Med Imaging Radiat Sci 2022; 53:S22-S30. [DOI: 10.1016/j.jmir.2022.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 02/21/2022] [Accepted: 03/28/2022] [Indexed: 11/20/2022]
|
113
|
Navalkissoor S, Toumpanakis C, Caplin M, Gnanasegaran G. Treatment of neuroendocrine tumours with 177 Lu-peptide receptor radionuclide therapy: Challenging clinical scenarios and their management. J Neuroendocrinol 2022; 34:e13088. [PMID: 35078265 DOI: 10.1111/jne.13088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 12/09/2021] [Accepted: 12/31/2021] [Indexed: 11/28/2022]
Abstract
Peptide receptor radionuclide therapy (PRRT) for the treatment of patients with neuroendocrine tumours is usually well tolerated, with selection based on existing guidelines. However, there are various scenarios where the risk-benefit analysis of PRRT needs to be evaluated to limit any complications associated with PRRT. This review looks at 11 key scenarios where the risk-benefit of PRRT needs to be closely evaluated.
Collapse
Affiliation(s)
- Shaunak Navalkissoor
- Department of Nuclear Medicine, Royal Free London NHS Foundation Trust, London, UK
| | | | - Martyn Caplin
- Neuroendocrine Unit, Royal Free London NHS Foundation Trust, London, UK
| | | |
Collapse
|
114
|
Medical physics services in radiology and nuclear medicine in Africa: challenges and opportunities identified through workforce and infrastructure surveys. HEALTH AND TECHNOLOGY 2022. [DOI: 10.1007/s12553-022-00663-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
AbstractThe International Atomic Energy Agency (IAEA) developed a staffing model to estimate the number of clinically qualified medical physicists (CQMP) that are required in an imaging facility, including diagnostic radiology and nuclear medicine. For the first time this staffing model was applied on a large scale across Africa. Within the framework of the IAEA African Regional Agreement (AFRA) Technical Cooperation (TC) project RAF6/053 entitled “Enhancing Capacity Building of Medical Physics to Improve Safety and Effectiveness of Medical Imaging (AFRA)”, a survey based on the IAEA staffing model was used to investigate the current CQMP workforce needs in imaging and radionuclide therapy in Africa in order to establish a baseline, identify gaps and suggest steps for improvement. The survey was open for five months, after which data verification was performed. 82 responses were received from 21 countries, including data from 97 diagnostic radiology and 40 nuclear medicine departments, as well as 75 interventional radiology departments and/or catheterization laboratories. Only 26·8% of centres employed an adequate number of CQMPs. The staffing model indicated that 134·3 CQMPs were required for these centres, but only 63 are currently employed in medical imaging and/or nuclear medicine at these centres. At least 11 countries do not have a single institution with an adequate number of CQMPs. Data analysis indicated that the number of radiology and nuclear medicine CQMPs is largely inadequate, at least by a factor of 20 in almost all countries in the region.
Collapse
|
115
|
Antonella M, Laura O, Stefania D, Joniada D, Matteo S. Should peptide receptors radionuclide therapy (PRRT) be considered as a treatment of choice in functioning metastatic insulinomas? A review of literature and our center experience. Clin Transl Imaging 2022. [DOI: 10.1007/s40336-022-00490-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
116
|
EANM dosimetry committee recommendations for dosimetry of 177Lu-labelled somatostatin-receptor- and PSMA-targeting ligands. Eur J Nucl Med Mol Imaging 2022; 49:1778-1809. [PMID: 35284969 PMCID: PMC9015994 DOI: 10.1007/s00259-022-05727-7] [Citation(s) in RCA: 98] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 02/13/2022] [Indexed: 12/25/2022]
Abstract
The purpose of the EANM Dosimetry Committee is to provide recommendations and guidance to scientists and clinicians on patient-specific dosimetry. Radiopharmaceuticals labelled with lutetium-177 (177Lu) are increasingly used for therapeutic applications, in particular for the treatment of metastatic neuroendocrine tumours using ligands for somatostatin receptors and prostate adenocarcinoma with small-molecule PSMA-targeting ligands. This paper provides an overview of reported dosimetry data for these therapies and summarises current knowledge about radiation-induced side effects on normal tissues and dose-effect relationships for tumours. Dosimetry methods and data are summarised for kidneys, bone marrow, salivary glands, lacrimal glands, pituitary glands, tumours, and the skin in case of radiopharmaceutical extravasation. Where applicable, taking into account the present status of the field and recent evidence in the literature, guidance is provided. The purpose of these recommendations is to encourage the practice of patient-specific dosimetry in therapy with 177Lu-labelled compounds. The proposed methods should be within the scope of centres offering therapy with 177Lu-labelled ligands for somatostatin receptors or small-molecule PSMA.
Collapse
|
117
|
Weich A, Higuchi T, Bundschuh RA, Lapa C, Serfling SE, Rowe SP, Pomper MG, Herrmann K, Buck AK, Derlin T, Werner RA. Training on Reporting and Data System (RADS) for Somatostatin-Receptor Targeted Molecular Imaging Can Reduce the Test Anxiety of Inexperienced Readers. Mol Imaging Biol 2022; 24:631-640. [PMID: 35233654 PMCID: PMC9296379 DOI: 10.1007/s11307-022-01712-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 02/09/2022] [Accepted: 02/16/2022] [Indexed: 11/13/2022]
Abstract
Purpose For somatostatin receptor (SSTR)-positron emission tomography/computed tomography (PET/CT), a standardized framework termed SSTR-reporting and data system (RADS) has been proposed. We aimed to elucidate the impact of a RADS-focused training on reader’s anxiety to report on SSTR-PET/CT, the motivational beliefs in learning such a system, whether it increases reader’s confidence, and its implementation in clinical routine. Procedures A 3-day training course focusing on SSTR-RADS was conducted. Self-report questionnaires were handed out prior to the course (Pre) and thereafter (Post). The impact of the training on the following categories was evaluated: (1) test anxiety to report on SSTR-PET/CT, (2) motivational beliefs, (3) increase in reader’s confidence, and (4) clinical implementation. To assess the effect size of the course, Cohen’s d was calculated (small, d = 0.20; large effect, d = 0.80). Results Of 22 participants, Pre and Post were returned by 21/22 (95.5%). In total, 14/21 (66.7%) were considered inexperienced (IR, < 1 year experience in reading SSTR-PET/CTs) and 7/21 (33.3%) as experienced readers (ER, > 1 year). Applying SSTR-RADS, a large decrease in anxiety to report on SSTR-PET/CT was noted for IR (d = − 0.74, P = 0.02), but not for ER (d = 0.11, P = 0.78). For the other three categories motivational beliefs, reader’s confidence, and clinical implementation, agreement rates were already high prior to the training and persisted throughout the course (P ≥ 0.21). Conclusions A framework-focused reader training can reduce anxiety to report on SSTR-PET/CTs, in particular for inexperienced readers. This may allow for a more widespread adoption of this system, e.g., in multicenter trials for better intra- and interindividual comparison of scan results. Supplementary Information The online version contains supplementary material available at 10.1007/s11307-022-01712-6.
Collapse
Affiliation(s)
- Alexander Weich
- Department of Internal Medicine II and ENETS Center of Excellence, Gastroenterology, University Hospital Würzburg, Würzburg, Germany
| | - Takahiro Higuchi
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany.,Dentistry and Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Ralph A Bundschuh
- Department of Nuclear Medicine, University Hospital Bonn, Bonn, Germany
| | - Constantin Lapa
- Nuclear Medicine, Medical Faculty, University of Augsburg, Augsburg, Germany
| | | | - Steven P Rowe
- The Russell H Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Martin G Pomper
- The Russell H Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Ken Herrmann
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
| | - Andreas K Buck
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Thorsten Derlin
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Rudolf A Werner
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany. .,The Russell H Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
118
|
Park EA, Graves SA, Menda Y. The Impact of Radiopharmaceutical Therapy on Renal Function. Semin Nucl Med 2022; 52:467-474. [DOI: 10.1053/j.semnuclmed.2022.02.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 02/13/2022] [Accepted: 02/20/2022] [Indexed: 11/11/2022]
|
119
|
Ambrosini V, Zanoni L, Filice A, Lamberti G, Argalia G, Fortunati E, Campana D, Versari A, Fanti S. Radiolabeled Somatostatin Analogues for Diagnosis and Treatment of Neuroendocrine Tumors. Cancers (Basel) 2022; 14:1055. [PMID: 35205805 PMCID: PMC8870358 DOI: 10.3390/cancers14041055] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/10/2022] [Accepted: 02/17/2022] [Indexed: 02/04/2023] Open
Abstract
Neuroendocrine neoplasms (NENs) are rare and heterogeneous tumors that require multidisciplinary discussion for optimal care. The theranostic approach (DOTA peptides labelled with 68Ga for diagnosis and with 90Y or 177Lu for therapy) plays a crucial role in the management of NENs to assess disease extension and as a criteria for peptide receptor radionuclide therapy (PRRT) eligibility based on somatostatin receptor (SSTR) expression. On the diagnostic side, [68Ga]Ga-DOTA peptides PET/CT (SSTR PET/CT) is the gold standard for imaging well-differentiated SSTR-expressing neuroendocrine tumors (NETs). [18F]FDG PET/CT is useful in higher grade NENs (NET G2 with Ki-67 > 10% and NET G3; NEC) for more accurate disease characterization and prognostication. Promising emerging radiopharmaceuticals include somatostatin analogues labelled with 18F (to overcome the limits imposed by 68Ga), and SSTR antagonists (for both diagnosis and therapy). On the therapeutic side, the evidence gathered over the past two decades indicates that PRRT is to be considered as an effective and safe treatment option for SSTR-expressing NETs, and is currently included in the therapeutic algorithms of the main scientific societies. The positioning of PRRT in the treatment sequence, as well as treatment personalization (e.g., tailored dosimetry, re-treatment, selection criteria, and combination with other alternative treatment options), is warranted in order to improve its efficacy while reducing toxicity. Although very preliminary (being mostly hampered by lack of methodological standardization, especially regarding feature selection/extraction) and often including small patient cohorts, radiomic studies in NETs are also presented. To date, the implementation of radiomics in clinical practice is still unclear. The purpose of this review is to offer an overview of radiolabeled SSTR analogues for theranostic use in NENs.
Collapse
Affiliation(s)
- Valentina Ambrosini
- Department of Experimental Diagnostic and Specialized Medicine, University of Bologna, 40138 Bologna, Italy; (V.A.); (G.L.); (G.A.); (E.F.); (D.C.); (S.F.)
- Nuclear Medicine Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Lucia Zanoni
- Nuclear Medicine Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Angelina Filice
- Nuclear Medicine Unit, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy; (A.F.); (A.V.)
| | - Giuseppe Lamberti
- Department of Experimental Diagnostic and Specialized Medicine, University of Bologna, 40138 Bologna, Italy; (V.A.); (G.L.); (G.A.); (E.F.); (D.C.); (S.F.)
- Division of Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Giulia Argalia
- Department of Experimental Diagnostic and Specialized Medicine, University of Bologna, 40138 Bologna, Italy; (V.A.); (G.L.); (G.A.); (E.F.); (D.C.); (S.F.)
| | - Emilia Fortunati
- Department of Experimental Diagnostic and Specialized Medicine, University of Bologna, 40138 Bologna, Italy; (V.A.); (G.L.); (G.A.); (E.F.); (D.C.); (S.F.)
| | - Davide Campana
- Department of Experimental Diagnostic and Specialized Medicine, University of Bologna, 40138 Bologna, Italy; (V.A.); (G.L.); (G.A.); (E.F.); (D.C.); (S.F.)
- Division of Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Annibale Versari
- Nuclear Medicine Unit, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy; (A.F.); (A.V.)
| | - Stefano Fanti
- Department of Experimental Diagnostic and Specialized Medicine, University of Bologna, 40138 Bologna, Italy; (V.A.); (G.L.); (G.A.); (E.F.); (D.C.); (S.F.)
- Nuclear Medicine Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| |
Collapse
|
120
|
Laudicella R, Comelli A, Liberini V, Vento A, Stefano A, Spataro A, Crocè L, Baldari S, Bambaci M, Deandreis D, Arico’ D, Ippolito M, Gaeta M, Alongi P, Minutoli F, Burger IA, Baldari S. [68Ga]DOTATOC PET/CT Radiomics to Predict the Response in GEP-NETs Undergoing [177Lu]DOTATOC PRRT: The “Theragnomics” Concept. Cancers (Basel) 2022; 14:cancers14040984. [PMID: 35205733 PMCID: PMC8870649 DOI: 10.3390/cancers14040984] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/12/2022] [Accepted: 02/14/2022] [Indexed: 02/01/2023] Open
Abstract
Despite impressive results, almost 30% of NET do not respond to PRRT and no well-established criteria are suitable to predict response. Therefore, we assessed the predictive value of radiomics [68Ga]DOTATOC PET/CT images pre-PRRT in metastatic GEP NET. We retrospectively analyzed the predictive value of radiomics in 324 SSTR-2-positive lesions from 38 metastatic GEP-NET patients (nine G1, 27 G2, and two G3) who underwent restaging [68Ga]DOTATOC PET/CT before complete PRRT with [177Lu]DOTATOC. Clinical, laboratory, and radiological follow-up data were collected for at least six months after the last cycle. Through LifeX, we extracted 65 PET features for each lesion. Grading, PRRT number of cycles, and cumulative activity, pre- and post-PRRT CgA values were also considered as additional clinical features. [68Ga]DOTATOC PET/CT follow-up with the same scanner for each patient determined the disease status (progression vs. response in terms of stability/reduction/disappearance) for each lesion. All features (PET and clinical) were also correlated with follow-up data in a per-site analysis (liver, lymph nodes, and bone), and for features significantly associated with response, the Δradiomics for each lesion was assessed on follow-up [68Ga]DOTATOC PET/CT performed until nine months post-PRRT. A statistical system based on the point-biserial correlation and logistic regression analysis was used for the reduction and selection of the features. Discriminant analysis was used, instead, to obtain the predictive model using the k-fold strategy to split data into training and validation sets. From the reduction and selection process, HISTO_Skewness and HISTO_Kurtosis were able to predict response with an area under the receiver operating characteristics curve (AUC ROC), sensitivity, and specificity of 0.745, 80.6%, 67.2% and 0.722, 61.2%, 75.9%, respectively. Moreover, a combination of three features (HISTO_Skewness; HISTO_Kurtosis, and Grading) did not improve the AUC significantly with 0.744. SUVmax. However, it could not predict response to PRRT (p = 0.49, AUC 0.523). The presented preliminary “theragnomics” model proved to be superior to conventional quantitative parameters to predict the response of GEP-NET lesions in patients treated with complete [177Lu]DOTATOC PRRT, regardless of the lesion site.
Collapse
Affiliation(s)
- Riccardo Laudicella
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98125 Messina, Italy; (A.V.); (A.S.); (L.C.); (F.M.); (S.B.)
- Ri.MED Foundation, 90134 Palermo, Italy;
- Department of Nuclear Medicine, University Hospital Zürich, University of Zürich, 8091 Zürich, Switzerland;
- Nuclear Medicine Unit, Fondazione Istituto G.Giglio, 90015 Cefalù, Italy;
- Correspondence: ; Tel.: +39-320-032-0150
| | | | - Virginia Liberini
- Nuclear Medicine Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (V.L.); (D.D.)
- Nuclear Medicine Department, S. Croce e Carle Hospital, 12100 Cuneo, Italy
| | - Antonio Vento
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98125 Messina, Italy; (A.V.); (A.S.); (L.C.); (F.M.); (S.B.)
| | - Alessandro Stefano
- Institute of Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), 90015 Cefalù, Italy;
| | - Alessandro Spataro
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98125 Messina, Italy; (A.V.); (A.S.); (L.C.); (F.M.); (S.B.)
| | - Ludovica Crocè
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98125 Messina, Italy; (A.V.); (A.S.); (L.C.); (F.M.); (S.B.)
| | - Sara Baldari
- Nuclear Medicine Department, Cannizzaro Hospital, 95126 Catania, Italy; (S.B.); (M.I.)
| | - Michelangelo Bambaci
- Department of Nuclear Medicine, Humanitas Oncological Centre of Catania, 95125 Catania, Italy; (M.B.); (D.A.)
| | - Desiree Deandreis
- Nuclear Medicine Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (V.L.); (D.D.)
| | - Demetrio Arico’
- Department of Nuclear Medicine, Humanitas Oncological Centre of Catania, 95125 Catania, Italy; (M.B.); (D.A.)
| | - Massimo Ippolito
- Nuclear Medicine Department, Cannizzaro Hospital, 95126 Catania, Italy; (S.B.); (M.I.)
| | - Michele Gaeta
- Section of Radiological Sciences, Department of Biomedical Sciences and Morphological and Functional Imaging, University of Messina, 98125 Messina, Italy;
| | - Pierpaolo Alongi
- Nuclear Medicine Unit, Fondazione Istituto G.Giglio, 90015 Cefalù, Italy;
| | - Fabio Minutoli
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98125 Messina, Italy; (A.V.); (A.S.); (L.C.); (F.M.); (S.B.)
| | - Irene A. Burger
- Department of Nuclear Medicine, University Hospital Zürich, University of Zürich, 8091 Zürich, Switzerland;
- Department of Nuclear Medicine, Kantonsspital Baden, 5404 Baden, Switzerland
| | - Sergio Baldari
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98125 Messina, Italy; (A.V.); (A.S.); (L.C.); (F.M.); (S.B.)
| |
Collapse
|
121
|
Early Complications of Radioisotope Therapy with Lutetium-177 and Yttrium-90 in Patients with Neuroendocrine Neoplasms-A Preliminary Study. J Clin Med 2022; 11:jcm11040919. [PMID: 35207193 PMCID: PMC8874379 DOI: 10.3390/jcm11040919] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/06/2022] [Accepted: 02/07/2022] [Indexed: 12/12/2022] Open
Abstract
Neuroendocrine neoplasms (NENs) constitute a heterogenous group of tumors originating from neuroendocrine cells scattered throughout the body. Peptide Receptor Radionuclide Therapy (PRRT) is a treatment of choice of unresectable metastasized progressive and well-differentiated NENs. The aim of the study was to assess early bone marrow and kidney injury after administration of Lutetium-177 or Lutetium-177 combined with Yttrium-90. Thirty-one patients received treatment with [177Lu]Lu-DOTATATE with the activity of 7.4 GBq. Eleven patients received tandem treatment with [90Y]Y-DOTATATE with the activity of 1.85 GBq + [177Lu]Lu-DOTATATE with the activity of 1.85 GBq. After PRRT a significant decrease in leukocyte, neutrophil, and lymphocyte counts was noted. Tandem treatment demonstrated a more marked decrease in white blood cell count compared to Lutetium-177 therapy only. Conversely, no significant influence on glomerular filtration was found in this assessment. However, PRRT triggered acute renal tubule dysfunction, regardless of the treatment type. Regarding the acute complications, PRRT appeared to be a safe modality in the treatment of patients with NEN.
Collapse
|
122
|
Naik M, Al-Nahhas A, Khan SR. Treatment of Neuroendocrine Neoplasms with Radiolabeled Peptides-Where Are We Now. Cancers (Basel) 2022; 14:761. [PMID: 35159027 PMCID: PMC8833798 DOI: 10.3390/cancers14030761] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 01/20/2022] [Indexed: 02/04/2023] Open
Abstract
Peptide receptor radionuclide therapy (PRRT) has been one of the most successful and exciting examples of theranostics in nuclear medicine in recent decades and is now firmly embedded in many treatment algorithms for unresectable or metastatic neuroendocrine neoplasms (NENs) worldwide. It is widely considered to be an effective treatment for well- or moderately differentiated neoplasms, which express high levels of somatostatin receptors that can be selectively targeted. This review article outlines the scientific basis of PRRT in treatment of NENs and describes its discovery dating back to the early 1990s. Early treatments utilizing Indium-111, a γ-emitter, showed promise in reduction in tumor size and improvement in biochemistry, but were also met with high radiation doses and myelotoxic and nephrotoxic effects. Subsequently, stable conjugation of DOTA-peptides with β-emitting radionuclides, such as Yttrium-90 and Lutetium-177, served as a breakthrough for PRRT and studies highlighted their potential in eliciting progression-free survival and quality of life benefits. This article will also elaborate on the key trials which paved the way for its approval and will discuss therapeutic considerations, such as patient selection and administration technique, to optimize its use.
Collapse
Affiliation(s)
- Mitesh Naik
- Department of Imaging, Charing Cross Hospital, Fulham Palace Road, London W6 8RF, UK;
| | | | - Sairah R. Khan
- Department of Imaging, Charing Cross Hospital, Fulham Palace Road, London W6 8RF, UK;
| |
Collapse
|
123
|
Kavanal AJ, Satapathy S, Sood A, Khosla D, Mittal BR. Subclinical Hypothyroidism After 225Ac-DOTATATE Therapy in a Case of Metastatic Neuroendocrine Tumor: Unknown Adverse Effect of PRRT. Clin Nucl Med 2022; 47:e184-e186. [PMID: 34507333 DOI: 10.1097/rlu.0000000000003893] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
ABSTRACT Radiation-induced thyroid dysfunction is very rarely associated with peptide receptor radionuclide therapy (PRRT) for neuroendocrine tumors using β-emitting radionuclides. With the emergence of α radionuclide such as 225Ac, PRRT using these radionuclides has shown better results; however, data on the toxicity profile are limited. This is a case report describing the thyroid dysfunction developed in a patient with inoperable metastatic neuroendocrine tumor with unknown primary after PRRT using 225Ac-DOTATATE, which has never been reported previously.
Collapse
Affiliation(s)
| | | | | | - Divya Khosla
- Radiation Oncology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | | |
Collapse
|
124
|
Durmo R, Filice A, Fioroni F, Cervati V, Finocchiaro D, Coruzzi C, Besutti G, Fanello S, Frasoldati A, Versari A. Predictive and Prognostic Role of Pre-Therapy and Interim 68Ga-DOTATOC PET/CT Parameters in Metastatic Advanced Neuroendocrine Tumor Patients Treated with PRRT. Cancers (Basel) 2022; 14:cancers14030592. [PMID: 35158862 PMCID: PMC8833820 DOI: 10.3390/cancers14030592] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 01/18/2022] [Accepted: 01/22/2022] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Although a significant improvement has been achieved in the management of metastatic neuroendocrine tumor (NET), disease progression is observed in 20–30% of patients treated with peptide receptor radionuclide therapy (PRRT). Therefore, the early identification of patients who are at high risk of treatment failure is important to avoid futile therapy toxicities. The aim of this study was to identify biomarkers derived from baseline and interim 68Ga-DOTATOC PET/CT in patients undergoing PRRT. In 46 metastatic NET patients with available baseline and interim PET, only baseline total tumor volume (bTV) was able to discriminate responders to PRRT (partial response or stable disease) vs. non-responders. Patients with high bTV had also the worst overall survival. bTV, an imaging biomarker, integrated in the initial workup of NET patients could improve risk stratification and contribute to a tailored therapy approach. Abstract Peptide receptor radionuclide therapy (PRRT) is an effective therapeutic option in patients with metastatic neuroendocrine tumor (NET). However, PRRT fails in about 15–30% of cases. Identification of biomarkers predicting the response to PRRT is essential for treatment tailoring. We aimed to evaluate the predictive and prognostic role of semiquantitative and volumetric parameters obtained from the 68Ga-DOTATOC PET/CT before therapy (bPET) and after two cycles of PRRT (iPET). A total of 46 patients were included in this retrospective analysis. The primary tumor was 78% gastroenteropancreatic (GEP), 13% broncho-pulmonary and 9% of unknown origin. 35 patients (76.1%) with stable disease or partial response after PRRT were classified as responders and 11 (23.9%) as non-responders. Logistic regression analysis identified that baseline total volume (bTV) was associated with therapy outcome (OR 1.17; 95%CI 1.02–1.32; p = 0.02). No significant association with PRRT response was observed for other variables. High bTV was confirmed as the only variable independently associated with OS (HR 12.76, 95%CI 1.53–107, p = 0.01). In conclusion, high bTV is a negative predictor for PRRT response and is associated with worse OS rates. Early iPET during PRRT apparently does not provide information useful to change the management of NET patients.
Collapse
Affiliation(s)
- Rexhep Durmo
- Nuclear Medicine Unit, Azienda USL-IRCCS of Reggio Emilia, 42123 Reggio Emilia, Italy; (A.F.); (C.C.); (A.V.)
- PhD Program in Clinical and Experimental Medicine (CEM), University of Modena and Reggio Emilia, 41125 Modena, Italy
- Correspondence: ; Tel.: +39-0522296284
| | - Angelina Filice
- Nuclear Medicine Unit, Azienda USL-IRCCS of Reggio Emilia, 42123 Reggio Emilia, Italy; (A.F.); (C.C.); (A.V.)
| | - Federica Fioroni
- Medical Physics Unit, Azienda USL-IRCCS of Reggio Emilia, 42123 Reggio Emilia, Italy; (F.F.); (D.F.)
| | - Veronica Cervati
- Nuclear Medicine Unit, Azienda Ospedaliero-Universitaria di Parma, 43126 Parma, Italy;
| | - Domenico Finocchiaro
- Medical Physics Unit, Azienda USL-IRCCS of Reggio Emilia, 42123 Reggio Emilia, Italy; (F.F.); (D.F.)
| | - Chiara Coruzzi
- Nuclear Medicine Unit, Azienda USL-IRCCS of Reggio Emilia, 42123 Reggio Emilia, Italy; (A.F.); (C.C.); (A.V.)
| | - Giulia Besutti
- Radiology Unit, Azienda USL-IRCCS of Reggio Emilia, 42123 Reggio Emilia, Italy;
| | - Silvia Fanello
- Medical Oncology Unit, Azienda USL-IRCCS of Reggio Emilia, 42123 Reggio Emilia, Italy;
| | - Andrea Frasoldati
- Department of Endocrinology and Metabolism, Azienda USL-IRCCS of Reggio Emilia, 42123 Reggio Emilia, Italy;
| | - Annibale Versari
- Nuclear Medicine Unit, Azienda USL-IRCCS of Reggio Emilia, 42123 Reggio Emilia, Italy; (A.F.); (C.C.); (A.V.)
| |
Collapse
|
125
|
Peptide Receptor Radionuclide Therapy with [ 177Lu]Lu-DOTA-TATE in Patients with Advanced GEP NENS: Present and Future Directions. Cancers (Basel) 2022; 14:cancers14030584. [PMID: 35158852 PMCID: PMC8833790 DOI: 10.3390/cancers14030584] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/17/2022] [Accepted: 01/20/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Neuroendocrine neoplasms have been usually described as infrequent tumors, but their incidence has been rising over time. [177Lu]Lu-DOTA-TATE (PRRT-Lu) was approved by the European Medicines Agency and by the Food and Drug Administration as the first radiopharmaceutical for peptide receptor radionuclide therapy in progressive gastroenteropancreatic NET. PRRT-Lu is considered a therapeutic option in progressive SSTR-positive NETs with homogenous SSTR expression. The NETTER-1 study demonstrated that PRRT-Lu yielded a statistically and clinically significant improvement in PFS as a primary endpoint (HR: 0.18, p < 0.0001), as well as a clinical trend towards improvement in OS. These results made scientific societies incorporate PRRT-Lu into their clinical guidelines; however, some questions still remain unanswered. Abstract This review article summarizes findings published in the last years on peptide receptor radionuclide therapy in GEP NENs, as well as potential future developments and directions. Unanswered questions remain, such as the following: Which is the correct dose and individual dosimetry? Which is the place for salvage PRRT-Lu? Whicht is the role of PRRT-Lu in the pediatric population? Which is the optimal sequencing of PRRT-Lu in advanced GEP NETs? Which is the place of PRRT-Lu in G3 NENs? These, and future developments such as inclusion new radiopharmaceuticals and combination therapy with different agents, such as radiosensitizers, will be discussed.
Collapse
|
126
|
Earley DF, Esteban Flores J, Guillou A, Holland JP. Photoactivatable bis(thiosemicarbazone) derivatives for copper-64 radiotracer synthesis. Dalton Trans 2022; 51:5041-5052. [PMID: 35285835 PMCID: PMC8962981 DOI: 10.1039/d2dt00209d] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In recent years, copper-64 and copper-67 have been considered as a useful theranostic pair in nuclear medicine, due to their favourable and complementary decay properties. As 67Cu and 64Cu are chemically identical, development of both existing and new bifunctional chelators for 64Cu imaging agents can be readily adapted for the 67Cu-radionuclide. In this study, we explored the use of photoactivatable copper chelators based on the asymmetric bis(thiosemicarbazone) scaffold, H2ATSM/en, for the photoradiolabelling of protein. Photoactivatable 64CuATSM-derivatives were prepared by both direct synthesis and transmetallation from the corresponding natZn complex. Then, irradiation with UV light in the presence of a protein of interest in a pH buffered aqueous solution afforded the 64Cu-labelled protein conjugates in decay-corrected radiochemical yield of 86.9 ± 1.0% via the transmetallation method and 35.3 ± 1.7% from the direct radiolabelling method. This study successfully demonstrates the viability of photochemically induced conjugation methods for the development of copper-based radiotracers for potential applications in diagnostic positron emission tomography (PET) imaging and targeted radionuclide therapy. In recent years, copper-64 and copper-67 have been considered as a useful theranostic pair in nuclear medicine. Here, we report a photochemically-mediated approach for radiolabelling biologically relevant protein with copper radionuclides.![]()
Collapse
Affiliation(s)
- Daniel F Earley
- Department of Chemistry, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland.
| | - Jose Esteban Flores
- Department of Chemistry, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland.
| | - Amaury Guillou
- Department of Chemistry, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland.
| | - Jason P Holland
- Department of Chemistry, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland.
| |
Collapse
|
127
|
Terapia con péptidos radiomarcados con [177Lu]Lu-DOTA-TATE. Rev Esp Med Nucl Imagen Mol 2022. [DOI: 10.1016/j.remn.2021.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
128
|
Prado-Wohlwend S, del Olmo-García MI, Bello-Arques P, Merino-Torres JF. [ 177Lu]Lu-DOTA-TATE and [ 131I]MIBG Phenotypic Imaging-Based Therapy in Metastatic/Inoperable Pheochromocytomas and Paragangliomas: Comparative Results in a Single Center. Front Endocrinol (Lausanne) 2022; 13:778322. [PMID: 35197929 PMCID: PMC8859101 DOI: 10.3389/fendo.2022.778322] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 01/10/2022] [Indexed: 11/15/2022] Open
Abstract
PURPOSE The aim of the study is to assess phenotypic imaging patterns and the response to treatment with [177Lu]Lu-DOTA-TATE and/or [131I]MIBG in paragangliomas (PGLs) and pheochromocytomas (PHEOs), globally and according to the primary location. METHODS This is a 17-patient retrospective observational study, with 9 cases treated with [177Lu]Lu-DOTA-TATE and 8 with [131I]MIBG (37 total treatments). Functional imaging scans and treatment responses were studied in order to choose the best therapeutic option and to define the progression-free survival (PFS) and disease control rate (DCR) according to treatment modality and primary location. RESULTS All patients were studied with phenotypic nuclear medicine images. Twelve of 17 patients were tested with both [123I]MIBG and somatostatin receptor images, and 6/12 showed appropriate expression of both targets to treatment in the phenotypic images. The rest of the patients were tested with one of the image modalities or only showed suitable uptake of a single radiotracer and were treated with the corresponding therapeutic option. [177Lu]Lu-DOTA-TATE PFS was 29 months with a DCR of 88.8%. [131I]MIBG PFS was 18.5 months with a 62.5% DCR. According to the primary location, the best PFS was in PHEOs treated with [177Lu]Lu-DOTA-TATE. Although the series are small due to the low disease prevalence and do not allow to yield statistically significant differences, this first study comparing [177Lu]Lu-DOTA-TATE and [131I]MIBG displays a trend to an overall longer PFS with [177Lu]Lu-DOTA-TATE, especially in the adrenal primary location. When both radionuclide targets are expressed, the patients' comorbidity and treatment effectiveness should be valued together with the intensity uptake in the phenotypic image in order to choose the best therapeutic option. These preliminary retrospective results reinforce the need for a prospective, multicentric trial to be confirmed.
Collapse
Affiliation(s)
- Stefan Prado-Wohlwend
- Nuclear Medicine Department, University and Polytechnic Hospital La Fe, Valencia, Spain
- *Correspondence: Stefan Prado-Wohlwend,
| | | | - Pilar Bello-Arques
- Nuclear Medicine Department, University and Polytechnic Hospital La Fe, Valencia, Spain
| | - Juan Francisco Merino-Torres
- Endocrinology and Nutrition Department, University and Polytechnic Hospital La Fe, Valencia, Spain
- Medicine Department, Universitat de València, Valencia, Spain
| |
Collapse
|
129
|
Forrer F. Side effects of radiopeptide therapy for neuroendocrine tumors. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00157-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
130
|
Kabasakal L, Demirci E, Selçuk NA. Radionuclide Therapy in Neuroendocrine Tumors. RADIONUCLIDE THERAPY 2022:173-186. [DOI: 10.1007/978-3-030-97220-2_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
131
|
First experiences with dynamic renal [ 68Ga]Ga-DOTA PET/CT: a comparison to renal scintigraphy and compartmental modelling to non-invasively estimate the glomerular filtration rate. Eur J Nucl Med Mol Imaging 2022; 49:3373-3386. [PMID: 35412053 PMCID: PMC9002049 DOI: 10.1007/s00259-022-05781-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 03/26/2022] [Indexed: 01/11/2023]
Abstract
PURPOSE The determination of the glomerular filtration rate (GFR) is decisive for a variety of clinical issues, for example, to monitor the renal function in radionuclide therapy patients. Renal scintigraphy using glomerularly filtered tracers allows combined acquisition of renograms and GFR estimation but requires repeated blood sampling for several hours. In contrast, dynamic PET imaging using the glomerularly filtered tracer [68Ga]Ga-DOTA bears the potential to non-invasively estimate the GFR by compartmental kinetic modelling. Here, we report the, to our knowledge, first comparison of human renal dynamic [68Ga]Ga-DOTA PET imaging in comparison to renal scintigraphy and compare PET-derived to serum creatinine-derived GFR measurements. METHODS Dynamic [68Ga]Ga-DOTA PET data were acquired for 30 min immediately after tracer injection in 12 patients. PET and renal scintigraphy images were visually interpreted in a consensus read by three nuclear medicine physicians. The functional renal cortex was segmented to obtain time-activity curves. The arterial input function was estimated from the PET signal in the abdominal aorta. Single-compartmental tracer kinetic modelling was performed to calculate the GFR using complete 30-min (GFRPET-30) and reduced 15-min PET data sets (GFRPET-15) to evaluate whether a shorter acquisition time is sufficient for an accurate GFR estimation. A modified approach excluding minutes 2 to 10 was applied to reduce urinary spill-over effects. Serum creatinine-derived GFRCKD (CKD-EPI-formula) was used as reference standard. RESULTS PET image interpretation revealed the same findings as conventional scintigraphy (2/12 patients with both- and 1/12 patients with right-sided urinary obstruction). Model fit functions were substantially improved for the modified approach to exclude spill-over. Depending on the modelling approach, GFRCKD and both GFRPET-30 and GFRPET-15 were well correlated with interclass correlation coefficients (ICCs) from 0.74 to 0.80 and Pearson's correlation coefficients (PCCs) from 0.74 to 0.81. For a subgroup of patients with undisturbed urinary efflux (n = 9), correlations were good to excellent (ICCs from 0.82 to 0.95 and PCCs from 0.83 to 0.95). Overall, GFRPET-30 and GFRPET-15 were excellently correlated (ICCs from 0.96 to 0.99 and PCCs from 0.96 to 0.99). CONCLUSION Renal [68Ga]Ga-DOTA PET can be a suitable alternative to conventional scintigraphy. Visual assessment of PET images and conventional renograms revealed comparable results. GFR values derived by non-invasive single-compartmental-modelling of PET data show a good correlation to serum creatinine-derived GFR values. In patients with undisturbed urinary efflux, the correlation was excellent. Dynamic PET data acquisition for 15 min is sufficient for visual evaluation and GFR derivation.
Collapse
|
132
|
Medullary Thyroid Cancer. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00106-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
133
|
Werner RA, Pomper MG, Buck AK, Rowe SP, Higuchi T. SPECT and PET Radiotracers in Renal Imaging. Semin Nucl Med 2022; 52:406-418. [DOI: 10.1053/j.semnuclmed.2021.12.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 12/06/2021] [Indexed: 12/24/2022]
|
134
|
Fanti S, Ambrosini V. PET Imaging in Neuro-Endocrine Neoplasms (NEN). Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00034-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
135
|
Elgazzar AH, Sarikaya I. Basis of Therapeutic Nuclear Medicine. THE PATHOPHYSIOLOGIC BASIS OF NUCLEAR MEDICINE 2022:569-594. [DOI: 10.1007/978-3-030-96252-4_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
136
|
Tabacchi E, Nanni C, Bossert I, Maffione AM, Fanti S. Diagnostic Applications of Nuclear Medicine: Pancreatic Cancer. NUCLEAR ONCOLOGY 2022:891-917. [DOI: 10.1007/978-3-031-05494-5_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
137
|
Covantev S, Volkov SI, Samsonova KI. Alternative Management of Cushing's Syndrome During Covid-19 Pandemic. CORONAVIRUSES 2022; 3. [DOI: 10.2174/2666796702666210913095645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 06/28/2021] [Accepted: 07/06/2021] [Indexed: 07/26/2024]
Abstract
:
Cushing's syndrome results from prolonged exposure to glucocorticoids. Surgery is often
the first-line treatment for this condition, regardless of etiology. However, the COVID-19 pandemic
caused a decrease in surgical procedures due to the risk of infection transmission. There are
still emergency cases of Cushing’s syndrome that are admitted to the hospital and require urgent
management. The current treatment should be focused on medical management and endovascular
embolization in selective cases. Embolization can be performed in facilities where there aretrained
personnel with experience in adrenal embolization. Surgery, which traditionally is a first-line therapy,
can increase the risk of infection, therefore, it should be avoided. The current review provides a
brief description of the possible options for the management of adrenal Cushing’s syndrome during
the COVID-19 pandemic.
Collapse
Affiliation(s)
- Serghei Covantev
- Department of Human Anatomy, State University of Medicine and Pharmacy N. Testemitanu, Moldova
| | - Stanislav I. Volkov
- Department
of Endocrinology, Russian Medical Academy of Continuous Professional Education, Moscow, Russia
| | - Kristina I. Samsonova
- Department of
Clinical Pharmacology, Multidisciplinary Medical Clinic of JSC “Medicine,” Moscow, Russia
| |
Collapse
|
138
|
Safety and Efficacy of Peptide-Receptor Radionuclide Therapy in Elderly Neuroendocrine Tumor Patients. Cancers (Basel) 2021; 13:cancers13246290. [PMID: 34944910 PMCID: PMC8699207 DOI: 10.3390/cancers13246290] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/07/2021] [Accepted: 12/13/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary We compared the safety and efficacy of targeted radionuclide therapy between elderly (79 years old and older) and disease-matched younger patients (between 60 and 70 years of age) with metastatic neuroendocrine tumour (NET). To our knowledge, this is the first paper addressing this important clinical question of the outcome of radionuclide therapy in this particularly vulnerable population. We found that targeted radionuclide therapy did not cause increased side effects in the elderly NET population, while toxicity remains modest and comparable in both groups. We also find that survival (after adjusting for differences in life expectancy) is not inferior for the elderly compared to younger NET patients. Abstract Peptide receptor radionuclide therapy (PRRT) is a well-established treatment in somatostatin receptor-expressing neuroendocrine tumours (NETs). The safety and efficacy of PRRT in >79 years old patients (EP) have not been systematically investigated. All patients with inoperable/metastatic/progressive G1/G2 NET, >79 years (EP), treated with PRRT at the University Hospital of Basel between 2006 and 2018, were enrolled in this retrospective matched cohort study. Each patient was manually matched with ≥1 younger patient (YP = 60–70 years). The primary endpoint was toxicity. Toxicity (subacute, long-term) was graded according to the criteria for adverse events (CTCAE) v5.0. All toxicity grades ≥ 3, or whose delta (Δ) to baseline were ≥2, were considered significant. The odds ratio (OR) for developing toxicity was tested for non-inferiority of EP vs. YP. Clinical response to PRRT and overall survival (OS) were assessed as secondary outcome measures. Forty-eight EP and 68 YP were enrolled. Both cohorts were balanced regarding median time since diagnosis, tumour location, grading, treatment scheme, and baseline biochemical parameters, except for eGFR (EP: 61 ± 16 vs. YP: 78 ± 19; mL/min/1.73 m2). Twenty-two grade ≥ 3 or Δ ≥ 2 subacute hematotoxicities occurred in 10 EP (10.3% of cycles) and 37 in 19 YP (11.6% of cycles; p = NS). Long-term grade ≥ 3 renal toxicity occurred in 7 EP and 2 YP (p = NS). The median OS was 3.4 years (EP) vs. 6.0 years (YP), HR: 1.50 [0.75, 2.98], p = NS. PRRT is a valid therapeutic option in elderly NET patients with similar toxicity and non-inferior survival compared to matched younger patients.
Collapse
|
139
|
Prado-Wohlwend S, Bernal-Vergara JC, Utrera-Costero A, Cañón-Sánchez JR, Agudelo-Cifuentes M, Bello-Arques P. Peptide receptor radionuclide therapy with [ 177Lu]Lu-DOTA-TATE. Rev Esp Med Nucl Imagen Mol 2021; 41:55-65. [PMID: 34920969 DOI: 10.1016/j.remnie.2021.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 11/10/2021] [Indexed: 11/30/2022]
Abstract
This continuing education aims to present in a clear and easy-to-understand way, the biology of neuroendocrine tumors (NETs), the characteristics of somatostatin receptors, the selection of patients for radiolabelled peptide therapy (PRRT), the inclusion criteria to benefit from treatment with the minimum possible adverse effects, the administration protocol, follow-up and response evaluation. The functional imaging studies necessary to explore the biology of the tumor and to individualize the treatment are also carried out, and constitute the cornerstone for the development of teragnosis. Clinical trials are being developed to better define the position of PRRT within the broad therapeutic options, and among the future perspectives, there are several lines of research to improve the objective response rate and survival with PRRT, focused on the development of new agonists and somatostatin receptor antagonists, new radionuclides and radiosensitizing combination therapies. In conclusion, PRRT is a great therapeutic, well-tolerated and safe tool with generally mild and self-limited acute side effects, that must be sequenced at the best moment of the evolution of the disease of patients with NET. Candidate patients for PRRT should always be evaluated by a multidisciplinary clinical committee.
Collapse
Affiliation(s)
- S Prado-Wohlwend
- Servicio de Medicina Nuclear, Hospital Universitario y Politécnico La Fe, Valencia, Spain.
| | - J C Bernal-Vergara
- Servicio de Medicina Nuclear, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - A Utrera-Costero
- Servicio de Medicina Nuclear, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - J R Cañón-Sánchez
- Servicio de Medicina Nuclear, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - M Agudelo-Cifuentes
- Servicio de Medicina Nuclear, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - P Bello-Arques
- Servicio de Medicina Nuclear, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | -
- Servicio de Medicina Nuclear, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| |
Collapse
|
140
|
Kennedy J, Chicheportiche A, Keidar Z. Quantitative SPECT/CT for dosimetry of peptide receptor radionuclide therapy. Semin Nucl Med 2021; 52:229-242. [PMID: 34911637 DOI: 10.1053/j.semnuclmed.2021.11.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Neuroendocrine tumors (NETs) are uncommon malignancies of increasing incidence and prevalence. As these slow growing tumors usually overexpress somatostatin receptors (SSTRs), the use of 68Ga-DOTA-peptides (gallium-68 chelated with dodecane tetra-acetic acid to somatostatin), which bind to the SSTRs, allows for PET based imaging and selection of patients for peptide receptor radionuclide therapy (PRRT). PRRT with radiolabeled somatostatin analogues such as 177Lu-DOTATATE (lutetium-177-[DOTA,Tyr3]-octreotate), is mainly used for the treatment of metastatic or inoperable NETs. However, PRRT is generally administered at a fixed injected activity in order not to exceed dose limits in critical organs, which is suboptimal given the variability in radiopharmaceutical uptake among patients. Advances in SPECT (single photon emission computed tomography) imaging enable the absolute quantitative measure of the true radiopharmaceutical distribution providing for PRRT dosimetry in each patient. Personalized PRRT based on patient-specific dosimetry could improve therapeutic efficacy by optimizing effective tumor absorbed dose while limiting treatment related radiotoxicity.
Collapse
Affiliation(s)
- John Kennedy
- Department of Nuclear Medicine, Rambam Health Care Campus, Haifa, Israel; B. Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel.
| | - Alexandre Chicheportiche
- Department of Nuclear Medicine and Biophysics, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Zohar Keidar
- Department of Nuclear Medicine, Rambam Health Care Campus, Haifa, Israel; B. Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
141
|
Mahmoudi E, Pirayesh E, Deevband MR, Amoui M, Ghodsi Rad M, Ghorbani Rad M, Ghorbani M. Radiation Dose to Medical Staff in 177Lu-PSMA-DKFZ-617 therapy And Estimation of Annual Dose. J Nucl Med Technol 2021; 50:269-273. [PMID: 34872918 DOI: 10.2967/jnmt.121.263135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 10/22/2021] [Indexed: 11/16/2022] Open
Abstract
Radioligand therapy applications for metastatic castration-resistant prostate cancer have been continuously rising in most nuclear medicine departments in Iran, but to our knowledge, no one has studied the doses of staff who perform treatment procedures. The current study aimed to determine the external radiation dose received by the staff of patients treated with 177Lu- prostate-specific membrane antigen therapy with and without a lead shield. This study used a dose ionization chamber to measure dose rates to the staff at various distances from patients and determined the average time spent by staff at these distances using an ionization chamber. Deep-dose equivalent to staff was obtained. The measured deep-dose equivalent to staff per patient was whitening the range of 1.8 to 5.2 mSv using a lead shield and 3.3 to 8.1 mSv without a lead shield. This study showed that a 2-mm lead shield markedly reduced the external dose to staff.It was indicated that the skill, accuracy, and speed of action of staff can directly affect their received dose.
Collapse
Affiliation(s)
- Elahe Mahmoudi
- Shahid Beheshti University of Medical Sciences, Iran, Islamic Republic of
| | - Elahe Pirayesh
- Shahid Beheshti University of Medical Sciences, Iran, Islamic Republic of
| | | | - Mahasti Amoui
- Shahid Beheshti University of Medical Sciences, Iran, Islamic Republic of
| | | | - Mehrdad Ghorbani Rad
- Nuclear Medicine Department, Shohada_e Tajrish Hospital, Iran, Islamic Republic of
| | - Mahdi Ghorbani
- Shahid Beheshti University of Medical Sciences, Iran, Islamic Republic of
| |
Collapse
|
142
|
Somatostatin and Somatostatin Receptors: From Signaling to Clinical Applications in Neuroendocrine Neoplasms. Biomedicines 2021; 9:biomedicines9121810. [PMID: 34944626 PMCID: PMC8699000 DOI: 10.3390/biomedicines9121810] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/23/2021] [Accepted: 11/25/2021] [Indexed: 12/26/2022] Open
Abstract
Neuroendocrine neoplasms (NENs) are heterogeneous neoplasms which arise from neuroendocrine cells that are distributed widely throughout the body. Although heterogenous, many of them share their ability to overexpress somatostatin receptors (SSTR) on their cell surface. Due to this, SSTR and somatostatin have been a large subject of interest in the discovery of potential biomarkers and treatment options for the disease. The aim of this review is to describe the molecular characteristics of somatostatin and somatostatin receptors and its application in diagnosis and therapy on patients with NENs as well as the use in the near future of somatostatin antagonists.
Collapse
|
143
|
Guarnotta V, Ferrigno R, Martino M, Barbot M, Isidori AM, Scaroni C, Ferrante A, Arnaldi G, Pivonello R, Giordano C. Glucocorticoid excess and COVID-19 disease. Rev Endocr Metab Disord 2021; 22:703-714. [PMID: 33025384 PMCID: PMC7538187 DOI: 10.1007/s11154-020-09598-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/28/2020] [Indexed: 12/15/2022]
Abstract
The pandemic of coronavirus disease (COVID-19), a disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is causing high and rapid morbidity and mortality. Immune system response plays a crucial role in controlling and resolving the viral infection. Exogenous or endogenous glucocorticoid excess is characterized by increased susceptibility to infections, due to impairment of the innate and adaptive immune system. In addition, diabetes, hypertension, obesity and thromboembolism are conditions overrepresented in patients with hypercortisolism. Thus patients with chronic glucocorticoid (GC) excess may be at high risk of developing COVID-19 infection with a severe clinical course. Care and control of all comorbidities should be one of the primary goals in patients with hypercortisolism requiring immediate and aggressive treatment. The European Society of Endocrinology (ESE), has recently commissioned an urgent clinical guidance document on management of Cushing's syndrome in a COVID-19 period. In this review, we aim to discuss and expand some clinical points related to GC excess that may have an impact on COVID-19 infection, in terms of both contagion risk and clinical outcome. This document is addressed to all specialists who approach patients with endogenous or exogenous GC excess and COVID-19 infection.
Collapse
Affiliation(s)
- Valentina Guarnotta
- Dipartimento di Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza "G. D'Alessandro", UOC di Malattie endocrine, del Ricambio e della Nutrizione, Università degli studi di Palermo, Piazza delle Cliniche 2, 90127, Palermo, Italy
| | - Rosario Ferrigno
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, Via Sergio Pansini 5, 80131, Naples, Italy
| | - Marianna Martino
- Clinica di Endocrinologia e Malattie del Metabolismo, Dipartimento di Scienze Cliniche e Molecolari (DISCLIMO), Università Politecnica delle Marche, Ospedali Riuniti di Ancona, Via Conca 71, 60126, Ancona, Italy
| | - Mattia Barbot
- Endocrinology Unit, Department of Medicine, DIME University-Hospital of Padova, Padua, Italy
| | - Andrea M Isidori
- Department of Experimental Medicine, Policlinico Umberto I, COVID Hospital, Sapienza University of Rome, 00161, Rome, Italy
| | - Carla Scaroni
- Endocrinology Unit, Department of Medicine, DIME University-Hospital of Padova, Padua, Italy
| | - Angelo Ferrante
- Dipartimento di Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza "G. D'Alessandro", UO di Reumatologia, Università degli studi di Palermo, Palermo, Italy
| | - Giorgio Arnaldi
- Clinica di Endocrinologia e Malattie del Metabolismo, Dipartimento di Scienze Cliniche e Molecolari (DISCLIMO), Università Politecnica delle Marche, Ospedali Riuniti di Ancona, Via Conca 71, 60126, Ancona, Italy.
| | - Rosario Pivonello
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università Federico II di Napoli, Via Sergio Pansini 5, 80131, Naples, Italy.
| | - Carla Giordano
- Dipartimento di Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza "G. D'Alessandro", UOC di Malattie endocrine, del Ricambio e della Nutrizione, Università degli studi di Palermo, Piazza delle Cliniche 2, 90127, Palermo, Italy.
| |
Collapse
|
144
|
Marini I, Sansovini M, Bongiovanni A, Nicolini S, Grassi I, Ranallo N, Monti M, DI Iorio V, Germanò L, Caroli P, Sarnelli A, Paganelli G, Severi S. Theragnostic in neuroendocrine tumors. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF... 2021; 65:342-352. [PMID: 34881852 DOI: 10.23736/s1824-4785.21.03426-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
In the last few decades, the incidence and prevalence of neuroendocrine tumors has been increasing. The theragnostic approach, that allows the diagnosis and treatment of different neoplasms with the same ligand, is a typical nuclear medicine tool. Applied for years, is also pivotal in neuroendocrine tumors (NETs) where it has improved the diagnostic accuracy and the therapeutic efficacy with impact on patient's survival. Theragnostic also allows the identification of important prognostic factors such as tumor location and burden, presence of liver metastases and intensity of somatostatin receptors (SSTR) expression to consider in new and possibly combined studies to ameliorate patient's outcome. Moreover, the possibility to evaluate receptor expression even in non-NET malignancies has de facto widened the possible indications for PRRT. We believe that this innovative therapeutic approach will be implemented in next years by radiomics and biological tumors characterization to better address PRRT applications.
Collapse
Affiliation(s)
- Irene Marini
- Unit of Nuclear Medicine, IRCCS Istituto Romagnolo per lo Studio dei Tumori - IRST Dino Amadori, Meldola, Forlì-Cesena, Italy
| | - Maddalena Sansovini
- Unit of Nuclear Medicine, IRCCS Istituto Romagnolo per lo Studio dei Tumori - IRST Dino Amadori, Meldola, Forlì-Cesena, Italy
| | - Alberto Bongiovanni
- Osteoncology and Rare Tumors Center - CDO-TR, IRCCS Istituto Romagnolo per lo Studio dei Tumori - IRST Dino Amadori, Meldola, Forlì-Cesena, Italy
| | - Silvia Nicolini
- Unit of Nuclear Medicine, IRCCS Istituto Romagnolo per lo Studio dei Tumori - IRST Dino Amadori, Meldola, Forlì-Cesena, Italy
| | - Ilaria Grassi
- Unit of Nuclear Medicine, IRCCS Istituto Romagnolo per lo Studio dei Tumori - IRST Dino Amadori, Meldola, Forlì-Cesena, Italy
| | - Nicoletta Ranallo
- Osteoncology and Rare Tumors Center - CDO-TR, IRCCS Istituto Romagnolo per lo Studio dei Tumori - IRST Dino Amadori, Meldola, Forlì-Cesena, Italy
| | - Manuela Monti
- Unit of Biostatistics and Clinical Trials, IRCCS Istituto Romagnolo per lo Studio dei Tumori - IRST Dino Amadori, Meldola, Forlì-Cesena, Italy
| | - Valentina DI Iorio
- Unit of Oncological Pharmacy, IRCCS Istituto Romagnolo per lo Studio dei Tumori - IRST Dino Amadori, Meldola, Forlì-Cesena, Italy
| | - Luca Germanò
- Unit of Nuclear Medicine, IRCCS Istituto Romagnolo per lo Studio dei Tumori - IRST Dino Amadori, Meldola, Forlì-Cesena, Italy
| | - Paola Caroli
- Unit of Nuclear Medicine, IRCCS Istituto Romagnolo per lo Studio dei Tumori - IRST Dino Amadori, Meldola, Forlì-Cesena, Italy
| | - Anna Sarnelli
- Unit of Medical Physics, IRCCS Istituto Romagnolo per lo Studio dei Tumori - IRST Dino Amadori, Meldola, Forlì-Cesena, Italy
| | - Giovanni Paganelli
- Unit of Nuclear Medicine, IRCCS Istituto Romagnolo per lo Studio dei Tumori - IRST Dino Amadori, Meldola, Forlì-Cesena, Italy
| | - Stefano Severi
- Unit of Nuclear Medicine, IRCCS Istituto Romagnolo per lo Studio dei Tumori - IRST Dino Amadori, Meldola, Forlì-Cesena, Italy -
| |
Collapse
|
145
|
Lassmann M, Eberlein U, Gear J, Konijnenberg M, Kunikowska J. Dosimetry for Radiopharmaceutical Therapy: The European Perspective. J Nucl Med 2021; 62:73S-79S. [PMID: 34857624 DOI: 10.2967/jnumed.121.262754] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 10/13/2021] [Indexed: 11/16/2022] Open
Abstract
This review presents efforts in Europe over the last few years with respect to standardization of quantitative imaging and dosimetry and comprises the results of several European research projects on practices regarding radiopharmaceutical therapies (RPTs). Because the European Union has regulatory requirements concerning dosimetry in RPTs, the European Association of Nuclear Medicine released a position paper in 2021 on the use of dosimetry under these requirements. The importance of radiobiology for RPTs is elucidated in another position paper by the European Association of Nuclear Medicine. Furthermore, how dosimetry interacts with clinical requirements is described, with several clinical examples. In the future, more efforts need to be undertaken to increase teaching and standardization efforts and to incorporate radiobiology for further individualizing patient treatment, with the aim of improving the outcome and safety of RPTs.
Collapse
Affiliation(s)
- Michael Lassmann
- Department of Nuclear Medicine, University of Würzburg, Würzburg, Germany
| | - Uta Eberlein
- Department of Nuclear Medicine, University of Würzburg, Würzburg, Germany;
| | - Jonathan Gear
- Joint Department of Physics, Royal Marsden NHS Foundation Trust and Institute of Cancer Research, London, United Kingdom
| | - Mark Konijnenberg
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands; and
| | - Jolanta Kunikowska
- Nuclear Medicine Department, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
146
|
Peptide receptor radionuclide therapy as a tool for the treatment of severe hypoglycemia in patients with primary inoperable insulinoma. BIO-ALGORITHMS AND MED-SYSTEMS 2021. [DOI: 10.1515/bams-2021-0138] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Abstract
Objectives
Severe hypoglycemia in a course of inoperable insulinoma may be life-threating and often it is not well controlled, even by high doses of diazoxide requiring second line treatment. Among available methods PRRT is characterized by relatively low toxicity and is connected with favorable antitumor effect. The aim of the study was an evaluation of the PRRT effectiveness in control of hypoglycemia in patients with primary inoperable insulinoma.
Methods
Three patients (female with metastatic insulinoma, male with primary inoperable pancreatic tumor, female with MEN1 syndrome and hepatic metastases) were treated with PRRT due to severe hypoglycemia poorly controlled by diazoxide in course of primary inoperable insulinoma.
Results
Patient 1 baseline fasting glucose concentration increased from 2.4 mmol/L [3.30–5.60] to 5.9 mmol/L after PRRT. In patient 2 fasting glucose level 2.30 mmol/L increased after PRRT to 7.0 mmol/L, while baseline insulin level initially 31.15 uU/mL [2.6–24.9] decreased to 15.4 uU/mL. In patients 3, baseline fasting glucose level 2.5 mmol/L increased after PRRT to 7.9 mmol/L, and insulin decreased from 57.9 uU/mL to 6.3 uU/mL. In imaging there was partial response (PR) in patient 1 and 2 and stabilization of the tumor size in patient 3. In patient 2 reduction of tumor infiltration let for curative surgery performed 4 months after PPRT.
Conclusions
PRRT may be effective as a first or second line treatment in management of hypoglycemia for patients with hormonally active inoperable insulinoma.
Collapse
|
147
|
de Vries–Huizing DMV, Versleijen MWJ, Sinaasappel M, Walraven I, Geluk–Jonker MM, Tesselaar MET, Hendrikx JJMA, de Wit–van der Veen BJ, Stokkel MPM. Haematotoxicity during peptide receptor radionuclide therapy: Baseline parameters differences and effect on patient's therapy course. PLoS One 2021; 16:e0260073. [PMID: 34793530 PMCID: PMC8601524 DOI: 10.1371/journal.pone.0260073] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 11/02/2021] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Mainly severe (CTCAE grade 3-4) haematotoxicity during peptide receptor radionuclide therapy (PRRT) is reported in literature due to major clinical impact, however moderate (CTCAE grade 2) haematotoxicity is common and could affect therapy management. The aim of this study was to evaluate the haematotoxicity course during PRRT and to compare baseline parameters between haematotoxicity grades. METHODS In this retrospective study, 100 patients with a neuroendocrine tumour treated with PRRT were included. Patients were treated with an aimed number of four cycles with 7.4 GBq [177Lu]Lu-DOTA-TATE administered every 10 weeks. Haematological assessment was performed at baseline and frequently up to 10 weeks after the fourth cycle. The lowest haematological value was graded according to CTCAE v5.0, and patients were classified using the highest observed grade. Differences in baseline parameters, including [68Ga]Ga-DOTA-TATE positive tumour volume, were evaluated between CTCAE grades. RESULTS Four cycles were completed by 86/100 of patients, 4/100 patients discontinued due to haematotoxicity, and 10/100 patients due to progressive disease. The treatment course was adjusted due to haematotoxicity in 24/100 patients, including postponed next cycle (n = 17), reduced administered activity (n = 13), and both adjustments (n = 10). The most observed haematotoxicity grade was grade 0-1 in 54/100 patients, grade 2 in 38/100 and grade 3-4 in 8/100. Significant differences in baseline leucocyte, neutrophil and platelet counts were observed between grade 0-1 and grade 2. However, the correlation between baseline and lowest observed values was poor to moderate. No differences between haematotoxicity grades and baseline parameters or somatostatin receptor positive tumour volume was observed. CONCLUSIONS The incidence of severe haematotoxicity was low with extensive screening and monitoring. The vast majority of patients (96/100) was not restricted in treatment continuation by haematotoxicity; therefore, our selection criteria appeared appropriate for safe PRRT treatment. Baseline parameters showed limited correlation with the degree of decline in haematological values.
Collapse
Affiliation(s)
| | | | - Michiel Sinaasappel
- Department of Clinical Physics and Instrumentation, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Iris Walraven
- Department for Health Evidence, Radboudumc, Nijmegen, The Netherlands
| | | | - Margot E. T. Tesselaar
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jeroen J. M. A. Hendrikx
- Department of Nuclear Medicine, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Pharmacy & Pharmacology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Marcel P. M. Stokkel
- Department of Nuclear Medicine, Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
148
|
Staanum PF, Frellsen AF, Olesen ML, Iversen P, Arveschoug AK. Practical kidney dosimetry in peptide receptor radionuclide therapy using [ 177Lu]Lu-DOTATOC and [ 177Lu]Lu-DOTATATE with focus on uncertainty estimates. EJNMMI Phys 2021; 8:78. [PMID: 34773508 PMCID: PMC8590641 DOI: 10.1186/s40658-021-00422-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 10/25/2021] [Indexed: 12/13/2022] Open
Abstract
Background Kidney dosimetry after peptide receptor radionuclide therapy using 177Lu-labelled somatostatin analogues is a procedure with multiple steps. We present the SPECT/CT-based implementation at Aarhus University Hospital and evaluate the uncertainty of the various steps in order to estimate the total uncertainty and to identify the major sources of uncertainty. Absorbed dose data from 115 treatment fractions are reported.
Results The total absorbed dose with uncertainty is presented for 59 treatments with [177Lu]Lu-DOTATOC and 56 treatments with [177Lu]Lu-DOTATATE. For [177Lu]Lu-DOTATOC the mean and median specific absorbed dose (dose per injected activity) is 0.37 Gy/GBq and 0.38 Gy/GBq, respectively, while for [177Lu]Lu-DOTATATE the median and mean are 0.47 Gy/GBq and 0.46 Gy/GBq, respectively. The uncertainty of the procedure is estimated to be about 13% for a single treatment fraction, where the absorbed dose calculation is based on three SPECT/CT scans 1, 4 and 7 days post-injection, while it increases to about 19% if only a single SPECT/CT scan is performed 1 day post-injection. Conclusions The specific absorbed dose values obtained with the described procedure are comparable to those from other treatment sites for both [177Lu]Lu-DOTATOC and [177Lu]Lu-DOTATATE, but towards the lower end of the range of reported values. The estimated uncertainty is also comparable to that from other reports and judged acceptable for clinical and research use, thus proving the kidney dosimetry procedure a useful tool. The greatest reduction in uncertainty can be obtained by improved activity determination, partial volume correction and additional SPECT/CT scans.
Collapse
Affiliation(s)
- Peter Frøhlich Staanum
- Department of Nuclear Medicine and PET-Centre, Aarhus University Hospital, Palle Juul-Jensens Boulevard 165, 8200, Aarhus N, Denmark.
| | - Anders Floor Frellsen
- Department of Nuclear Medicine and PET-Centre, Aarhus University Hospital, Palle Juul-Jensens Boulevard 165, 8200, Aarhus N, Denmark
| | - Marie Louise Olesen
- Department of Nuclear Medicine and PET-Centre, Aarhus University Hospital, Palle Juul-Jensens Boulevard 165, 8200, Aarhus N, Denmark
| | - Peter Iversen
- Department of Nuclear Medicine and PET-Centre, Aarhus University Hospital, Palle Juul-Jensens Boulevard 165, 8200, Aarhus N, Denmark
| | - Anne Kirstine Arveschoug
- Department of Nuclear Medicine and PET-Centre, Aarhus University Hospital, Palle Juul-Jensens Boulevard 165, 8200, Aarhus N, Denmark
| |
Collapse
|
149
|
Geenen L, Nonnekens J, Konijnenberg M, Baatout S, De Jong M, Aerts A. Overcoming nephrotoxicity in peptide receptor radionuclide therapy using [ 177Lu]Lu-DOTA-TATE for the treatment of neuroendocrine tumours. Nucl Med Biol 2021; 102-103:1-11. [PMID: 34242948 DOI: 10.1016/j.nucmedbio.2021.06.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 06/21/2021] [Accepted: 06/21/2021] [Indexed: 12/29/2022]
Abstract
Peptide receptor radionuclide therapy (PRRT) is used for the treatment of patients with unresectable or metastasized somatostatin receptor type 2 (SSTR2)-expressing gastroenteropancreatic neuroendocrine tumours (GEP-NETs). The radiolabelled somatostatin analogue [177Lu]Lu-DOTA-TATE delivers its radiation dose to SSTR2-overexpressing tumour cells, resulting in selective cell killing during radioactive decay. While tumour control can be achieved in many patients, complete remissions remain rare, causing the majority of patients to relapse after a certain period of time. This raises the question whether the currently fixed treatment regime (4 × 7.4 GBq) leaves room for dose escalation as a means of improving therapy efficacy. The kidneys have shown to play an important role in defining a patient's tolerability to PRRT. As a consequence of the proximal tubular reabsorption of [177Lu]Lu-DOTA-TATE, via the endocytic megalin/cubilin receptor complex, the radionuclides are retained in the renal interstitium. This results in extended retention of radioactivity in the kidneys, generating a risk for the development of radiation nephropathy. In addition, a decreased kidney function has shown to be associated with a prolonged circulation of [177Lu]Lu-DOTA-TATE, causing increased irradiation to the bone marrow. This can on its turn lead to myelosuppression and haematological toxicity, owing to the marked radio sensitivity of the rapidly proliferating cells in the bone marrow. In contrast to external beam radiotherapy (EBRT), the exact absorbed dose limits for these critical organs (kidneys and bone marrow) in PRRT with [177Lu]Lu-DOTA-TATE are still unclear. Better insights into these uncertainties, can help in optimizing PRRT to reach its maximum therapeutic potential, while avoiding severe adverse events, like nephropathy and hematologic toxicities. In this review we focus on the nephrotoxic effects of PRRT with [177Lu]Lu-DOTA-TATE for the treatment of GEP-NETs. If the absorbed dose to the kidneys can be lowered, higher activities can be administered, enlarging the therapeutic window for PRRT. Therefore, we evaluated the renal protective potential of current and promising future strategies and discuss the importance of (renal) dosimetry in PRRT.
Collapse
Affiliation(s)
- Lorain Geenen
- Radiobiology Unit, Interdisciplinary Biosciences, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium; Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, the Netherlands
| | - Julie Nonnekens
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, the Netherlands; Department of Molecular Genetics, Erasmus MC, Rotterdam, the Netherlands; Oncode Institute, Erasmus MC, Rotterdam, the Netherlands
| | - Mark Konijnenberg
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, the Netherlands; Department of Medical Imaging, Radboud UMC, Nijmegen, the Netherlands
| | - Sarah Baatout
- Radiobiology Unit, Interdisciplinary Biosciences, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium; Department of Molecular Biotechnology, Faculty of Bioengineering Sciences, Ghent University, Belgium.
| | - Marion De Jong
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, the Netherlands
| | - An Aerts
- Radiobiology Unit, Interdisciplinary Biosciences, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
| |
Collapse
|
150
|
|