1
|
Ciuciulkaite I, Herrmann K, Lahner H. [Importance of peptide receptor radionuclide therapy for the management of neuroendocrine tumours]. RADIOLOGIE (HEIDELBERG, GERMANY) 2025; 65:371-380. [PMID: 40227439 DOI: 10.1007/s00117-025-01452-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
Neuroendocrine tumours (NETs) are rare, heterogeneous neoplasms that often express somatostatin receptors (SSTRs). This allows targeted peptide receptor radionuclide therapy (PRRT) for NETs. PRRT is currently indicated as second- or third-line therapy for metastatic or unresectable, progressive, SSTR-positive NETs of grade (G) 1 or 2. Adequate bone marrow reserves as well as renal and hepatic function are required for PRRT. The most commonly used radiopharmaceutical for PRRT is 177Lu-DOTA-TATE. PRRT prolongs progression-free and overall survival, reduces or stabilises tumour burden, and improves tumour symptoms and quality of life. Adverse events associated with PRRT are mostly mild and transient. Haemato- and nephrotoxicity are the most common toxicities following PRRT. The NETTER‑2 and COMPOSE trials are investigating PRRT with 177Lu-DOTA-TATE/-TOC in G2 and G3 gastroenteropancreatic NETs.
Collapse
Affiliation(s)
- I Ciuciulkaite
- Klinik für Nuklearmedizin, Universitätsmedizin Essen, Universität Duisburg-Essen, Essen, Deutschland.
- Klinik für Nuklearmedizin, Universitätsklinikum Essen, Hufelandstr. 55, 45147, Essen, Deutschland.
| | - K Herrmann
- Klinik für Nuklearmedizin, Universitätsmedizin Essen, Universität Duisburg-Essen, Essen, Deutschland
| | - H Lahner
- Klinik für Endokrinologie, Diabetologie und Stoffwechsel, Universitätsmedizin Essen, Universität Duisburg-Essen, Essen, Deutschland
| |
Collapse
|
2
|
Parghane RV, Basu S. Toxicity manifestations encountered in peptide receptor radionuclide therapy setting. J Neuroendocrinol 2025; 37:e13464. [PMID: 39531370 DOI: 10.1111/jne.13464] [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: 08/07/2024] [Revised: 10/10/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
Peptide receptor radionuclide therapy (PRRT) has demonstrated immense promise as a treatment for patients with neuroendocrine tumors (NET) who have somatostatin receptor (SSTR) expression. PRRT significantly reduces tumor growth, stabilizes the disease, and prolongs survival in a significant percentage of patients with metastatic/advanced NET. It produces an important beneficial effect on the quality of life (QOL) and effectively alleviates symptoms in patients with NET. Overall, PRRT is typically well-tolerated and most of the side effects are usually transient and subside on their own. It is, however, crucial to be cognizant of the potential toxicities associated with this treatment. This awareness will enable physicians to promptly detect, effectively manage, and prevent these toxicities by identifying high-risk factors in NET patients. This review provides an in-depth overview for clinicians managing NET about the toxicity of PRRT. The toxicities are stratified into acute, subacute, and long-term based on their onset following PRRT. Potential high-risk factors in order to treat effectively and prevent these toxicities in NET patients are presented including the management strategy. This review also discusses novel insights, perspectives, and recent advancements in predicting, preventing, and managing toxicity associated with PRRT, while offering prospective future research directions to minimize clinical toxicity and maximize the therapeutic benefits of PRRT as a treatment strategy for NET patients.
Collapse
Affiliation(s)
- Rahul V Parghane
- Radiation Medicine Centre (BARC), Tata Memorial Hospital, Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Sandip Basu
- Radiation Medicine Centre (BARC), Tata Memorial Hospital, Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| |
Collapse
|
3
|
Trejtnar F, Bárta P, Kozempel J, Vlk M, Ďurinová A, Kuchařová M, Pávek P. Terbium-161 in nuclear medicine: Preclinical and clinical progress in comparison with lutetium-177. Nucl Med Biol 2025; 144-145:108998. [PMID: 39978166 DOI: 10.1016/j.nucmedbio.2025.108998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 02/05/2025] [Accepted: 02/11/2025] [Indexed: 02/22/2025]
Abstract
The use of new radiopharmaceuticals labeled with lutetium-177 represents a successful translation of experimental results into clinical practice. Recent experimental data suggests that terbium-161 might well follow the example of lutetium-177 regarding applicability in nuclear medicine. Similarly to lutetium-177, the terbium-161 emits beta particles and gamma-radiation, although terbium-161 emits short-ranged conversion and Auger electrons, creating an effect that may eliminate smaller tumor metastases more effectively than lutetium-177. Terbium-161 may exert a higher radiobiological effect in the target tissues in comparison with lutetium-177, a difference which makes possible a reduction in the doses of radioactivity administered. Further, due to the similar chemical properties of lutetium-177 and terbium-161, similar radiolabeling techniques can be used. The differences found in preclinical experiments on radiotoxicity of the counterparts seem to be minor. Despite intensive progress, the number of preclinical studies on 161Tb-labeled agents is still not comparable to studies on lutetium-177. Clinical trials with 161Tb-labeled radiopharmaceuticals focused on the treatment of prostate cancer and selected neuroendocrine tumors have already begun, although none of them have been completed yet.
Collapse
Affiliation(s)
- František Trejtnar
- Charles University, Faculty of Pharmacy in Hradec Králové, Department of Pharmacology and Toxicology, Hradec Králové, Czech Republic.
| | - Pavel Bárta
- Charles University, Faculty of Pharmacy in Hradec Králové, Department of Biophysics and Physical Chemistry, Hradec Králové, Czech Republic.
| | - Ján Kozempel
- Czech Technical University, Faculty of Nuclear Sciences and Physical Engineering, Department of Nuclear Chemistry, Prague, Czech Republic.
| | - Martin Vlk
- Czech Technical University, Faculty of Nuclear Sciences and Physical Engineering, Department of Nuclear Chemistry, Prague, Czech Republic.
| | - Anna Ďurinová
- Charles University, Faculty of Pharmacy in Hradec Králové, Department of Pharmacology and Toxicology, Hradec Králové, Czech Republic.
| | - Monika Kuchařová
- Charles University, Faculty of Pharmacy in Hradec Králové, Department of Biophysics and Physical Chemistry, Hradec Králové, Czech Republic.
| | - Petr Pávek
- Charles University, Faculty of Pharmacy in Hradec Králové, Department of Pharmacology and Toxicology, Hradec Králové, Czech Republic.
| |
Collapse
|
4
|
Piscopo L, Zampella E, Volpe F, Gaudieri V, Nappi C, Di Donna E, Clemente S, Varallo A, Scaglione M, Cuocolo A, Klain M. The Safety and Efficacy of Peptide Receptor Radionuclide Therapy for Gastro-Entero-Pancreatic Neuroendocrine Tumors: A Single Center Experience. Curr Oncol 2024; 31:5617-5629. [PMID: 39330044 PMCID: PMC11431754 DOI: 10.3390/curroncol31090416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/16/2024] [Accepted: 09/18/2024] [Indexed: 09/28/2024] Open
Abstract
The aim of the present study was to evaluate the safety and efficacy of radionuclide therapy with [177Lu]Lu-DOTA-TATE according to our single center experience at the University of Naples Federico II. For the present analysis, we considered 21 patients with progressive, advanced, well-differentiated G1 and G2 in patients with gastro-entero-pancreatic (GEP) neuroendocrine tumors (NETs) treated with [177Lu]Lu-DOTA-TATE according to the decisions of a multidisciplinary team. All patients underwent four cycles of 7-8 GBq of [177Lu]Lu-DOTA-TATE every 8 weeks. A whole-body scan (WBS) was performed 4, 48, and 168 h after each treatment. The dosimetry towards the organ at risk and target lesions was calculated. For each patient, renal and bone marrow parameters were evaluated before, during, and 3 months after the end of the treatment. Follow-up data were obtained and RECIST criteria were considered as the endpoint. Among 21 patients enrolled (mean age 65 ± 9 years); 17 (81%) were men and the small intestine was the most frequent location of disease (n = 12). A mild albeit significant variation (p < 0.05) in both platelets and white blood cell counts among all time points was observed, despite it disappearing 3 months after the end of the therapy. According to the RECIST criteria, 11 (55%) patients had a partial response to therapy and 8 (40%) had stable disease. Only one (5%) patient had disease progression 4 months after treatment. Our data confirm that [177Lu]Lu-DOTA is safe and effective in controlling the burden disease of G1/G2 GEP-NETs patients.
Collapse
Affiliation(s)
- Leandra Piscopo
- Department of Advanced Biomedical Sciences, University of Naples, Federico II, 80131 Naples, Italy; (E.Z.); (F.V.); (V.G.); (C.N.); (E.D.D.); (S.C.); (A.V.); (A.C.); (M.K.)
| | - Emilia Zampella
- Department of Advanced Biomedical Sciences, University of Naples, Federico II, 80131 Naples, Italy; (E.Z.); (F.V.); (V.G.); (C.N.); (E.D.D.); (S.C.); (A.V.); (A.C.); (M.K.)
| | - Fabio Volpe
- Department of Advanced Biomedical Sciences, University of Naples, Federico II, 80131 Naples, Italy; (E.Z.); (F.V.); (V.G.); (C.N.); (E.D.D.); (S.C.); (A.V.); (A.C.); (M.K.)
| | - Valeria Gaudieri
- Department of Advanced Biomedical Sciences, University of Naples, Federico II, 80131 Naples, Italy; (E.Z.); (F.V.); (V.G.); (C.N.); (E.D.D.); (S.C.); (A.V.); (A.C.); (M.K.)
| | - Carmela Nappi
- Department of Advanced Biomedical Sciences, University of Naples, Federico II, 80131 Naples, Italy; (E.Z.); (F.V.); (V.G.); (C.N.); (E.D.D.); (S.C.); (A.V.); (A.C.); (M.K.)
| | - Erica Di Donna
- Department of Advanced Biomedical Sciences, University of Naples, Federico II, 80131 Naples, Italy; (E.Z.); (F.V.); (V.G.); (C.N.); (E.D.D.); (S.C.); (A.V.); (A.C.); (M.K.)
| | - Stefania Clemente
- Department of Advanced Biomedical Sciences, University of Naples, Federico II, 80131 Naples, Italy; (E.Z.); (F.V.); (V.G.); (C.N.); (E.D.D.); (S.C.); (A.V.); (A.C.); (M.K.)
| | - Antonio Varallo
- Department of Advanced Biomedical Sciences, University of Naples, Federico II, 80131 Naples, Italy; (E.Z.); (F.V.); (V.G.); (C.N.); (E.D.D.); (S.C.); (A.V.); (A.C.); (M.K.)
| | - Mariano Scaglione
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy;
| | - Alberto Cuocolo
- Department of Advanced Biomedical Sciences, University of Naples, Federico II, 80131 Naples, Italy; (E.Z.); (F.V.); (V.G.); (C.N.); (E.D.D.); (S.C.); (A.V.); (A.C.); (M.K.)
| | - Michele Klain
- Department of Advanced Biomedical Sciences, University of Naples, Federico II, 80131 Naples, Italy; (E.Z.); (F.V.); (V.G.); (C.N.); (E.D.D.); (S.C.); (A.V.); (A.C.); (M.K.)
| |
Collapse
|
5
|
Kennedy AS, Brown DB, Fakih M, Jeyarajah R, Jones S, Liu D, Pinato DJ, Sangro B, Sharma NK, Sze DY, Van Cutsem E, Wasan HS. Multidisciplinary Delphi Consensus on Safety of Combining Transarterial Radioembolization with Yttrium-90 Microspheres with Systemic Anticancer Agents for the Treatment of Liver Malignancy. J Vasc Interv Radiol 2024; 35:1253-1267.e1. [PMID: 38885899 DOI: 10.1016/j.jvir.2024.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 05/31/2024] [Accepted: 06/09/2024] [Indexed: 06/20/2024] Open
Abstract
PURPOSE To provide guidance, via multidisciplinary consensus statements, on the safety interactions between systemic anticancer agents (such as radiosensitizing chemotherapy, immunotherapy, targeted therapy, and peptide receptor radionuclide therapy) and transarterial radioembolization (TARE) with yttrium-90 (90Y)-labeled microspheres in the treatment of primary and metastatic liver malignancies. MATERIALS AND METHODS A literature search identified 59 references that informed 26 statements on the safety of 90Y TARE combined with systemic therapies. Modified Delphi method was used to develop consensus on statements through online anonymous surveys of the 12 panel members representing the fields of interventional radiology, medical oncology, surgical oncology, hepatology, and pharmacy, focusing on hepatocellular carcinoma (HCC), metastatic colorectal cancer (mCRC), neuroendocrine tumors, metastatic breast cancer, and intrahepatic cholangiocarcinoma. RESULTS High-level evidence was limited. Level 1 data in patients with mCRC suggest that some radiosensitizing chemotherapies (eg, oxaliplatin) require temporary dose reduction when used concomitantly with 90Y TARE, and some targeted therapies (eg, vascular endothelial growth factor inhibitors and antiangiogenic tyrosine kinase inhibitors) should be avoided for at least 4 weeks before 90Y TARE. In patients with HCC, the feasibility of 90Y TARE and immunotherapy has been demonstrated with Level 4 evidence. Data are more limited for other primary and secondary liver malignancies, and consensus statements were driven by expert opinion (Level 5). CONCLUSIONS Given the absence of evidence-based guidelines on the safety of 90Y TARE in combination with systemic anticancer therapy, these consensus statements provide expert guidance on the potential risks when considering specific combinations.
Collapse
Affiliation(s)
- Andrew S Kennedy
- Radiation Oncology, Sarah Cannon Research Institute, Nashville, Tennessee.
| | - Daniel B Brown
- Interventional Radiology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Marwan Fakih
- Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center Duarte, Duarte, California
| | | | - Suzanne Jones
- Drug Development, Sarah Cannon Research Institute, Nashville, Tennessee
| | - David Liu
- Faculty of Medicine, School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | - David J Pinato
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, United Kingdom; Division of Oncology, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Bruno Sangro
- Liver Unit, Clinica Universidad de Navarra and CIBEREHD, Pamplona-Madrid, Spain
| | - Navesh K Sharma
- Department of Radiation Oncology, WellSpan Cancer Center, New York, Pennsylvania
| | - Daniel Y Sze
- Interventional Radiology, Stanford University, Palo Alto, California
| | - Eric Van Cutsem
- Digestive Oncology, University Hospitals Leuven and KU Leuven, Leuven, Belgium
| | - Harpreet S Wasan
- Department of Cancer Medicine, Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, United Kingdom
| |
Collapse
|
6
|
Trikalinos NA, Kim H, Vijayan A, Amurao M, Prasad V. Use of approved Lu-177 radiopharmaceuticals in patients with end-stage renal disease: A review of the literature and proposed treatment algorithm. J Neuroendocrinol 2024; 36:e13393. [PMID: 38622851 DOI: 10.1111/jne.13393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 03/20/2024] [Accepted: 04/03/2024] [Indexed: 04/17/2024]
Abstract
Peptide receptor radionuclide therapy (PRRT) can be a very useful treatment for patients with neuroendocrine neoplasms and metastatic castration-resistant prostate cancer but it is routinely avoided in those with advanced kidney disease because it can adversely affect the renal function. Accordingly, no clear guidelines exist on the use of PRRT for patients on hemodialysis (HD). We performed a literature review to identify publications on HD patients who received PRRT with Lutetium-177 (Lu177) Dotatate and Y-90 and obtained information on Lu177 pharmacokinetics and early testing data from the manufacturer. We also perused the most recent North American Neuroendocrine Tumor Society (NANETS)/European Neuroendocrine Tumor Society (ENETS) recommendations. Seven relevant publications with a total of 15 patients were included. Patients received dose-adjusted fractions of PRRT with HD occurring usually within 24 h. There were no immediate or long-term serious adverse events attributed to the radioligand, although data was limited. Using available evidence and input from a multidisciplinary group, we have created an institutional workflow. Dose-adjusted PRRT can be offered to patients undergoing HD under careful, multidisciplinary supervision.
Collapse
Affiliation(s)
- Nikolaos A Trikalinos
- Department of Medical Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Hyun Kim
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Anitha Vijayan
- Intermountain Kidney Services, Intermountain Health, Murray, Utah, USA
| | - Maxwell Amurao
- Office of Radiation Safety, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Vikas Prasad
- Department of Nuclear Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
7
|
Dubois J, Tosato G, Garrigue P, Taieb D, Guillet B, Nail V. Short-Term Biological Toxicity Prediction of [ 177Lu]Lutetium-Oxodotreotide: An Original Retrospective Analysis. Cancer Biother Radiopharm 2024; 39:381-389. [PMID: 38655905 PMCID: PMC11304756 DOI: 10.1089/cbr.2023.0195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2024] Open
Abstract
Introduction: [177Lu]Lutetium (Lu)-oxodotreotide is a radiopharmaceutical drug used as peptide receptor radionuclide therapy (PRRT) for somatostatin receptor-expressing neuroendocrine neoplasms. It provides an additional effective alternative treatment for these rare cancers. Although well tolerated, its safety profile must continue to be characterized to support its use as a first-line treatment or for additional cycles. This study evaluated factors associated with the occurrence of [177Lu]Lu-oxodotreotide induced short-term toxicity. Materials and Methods: A retrospective observational monocentric study was carried out from July 2013 to October 2021. Inclusion criteria were defined as follows: patients who received at least four cycles of [177Lu]Lu-oxodotreotide and were followed up for 6 months after the last injection. Graduated toxicity was defined using the National Cancer Institute Common Terminology Criteria for Adverse Events 5.0. Cox regression was used in the analysis. Results: Forty patients were included. The most frequent toxicities occurred during the first cycle and were graded as G1 or G2. As expected, toxicities were predominantly hematological and hepatic, with incomplete reversibility after each cycle. The following factors were significantly related to the occurrence of hematological or hepatic toxicity during PRRT: gastrointestinal primary tumor diagnosis, bone metastases, peritoneal metastases, pancreatic metastases or pulmonary metastases, and high tumor grade. Conclusion: Knowledge and consideration of these factors in adjusting [177Lu]Lu-oxodotreotide treatment regimen could help prevent or reduce the severity of these toxicities. Further studies are still warranted to refine these results and improve treatment management.
Collapse
Affiliation(s)
- Julien Dubois
- Department of Radiopharmacy, La Timone University Hospital, CERIMED, Aix-Marseille University, Marseille, France
- Department of Radiopharmacy, Montpellier University Hospital, Montpellier University, Montpellier, France
- Cancer Research Institute of Montpellier (IRCM), University of Montpellier, Montpellier, France
| | - Guillaume Tosato
- Montpellier University Hospital, Montpellier University, Montpellier, France
| | - Philippe Garrigue
- Department of Radiopharmacy, La Timone University Hospital, CERIMED, Aix-Marseille University, Marseille, France
| | - David Taieb
- Department of Nuclear Medicine, La Timone University Hospital, CERIMED, Aix-Marseille University, Marseille, France
| | - Benjamin Guillet
- Department of Radiopharmacy, La Timone University Hospital, CERIMED, Aix-Marseille University, Marseille, France
| | - Vincent Nail
- Department of Radiopharmacy, La Timone University Hospital, CERIMED, Aix-Marseille University, Marseille, France
| |
Collapse
|
8
|
Zhang M, Kang F, Xing T, Wang J, Ma T, Li G, Quan Z, Yang W, Chen X, Wang J. First-in-human validation of enzymolysis clearance strategy for decreasing renal radioactivity using modified [ 68Ga]Ga-HER2 Affibody. Eur J Nucl Med Mol Imaging 2024; 51:1713-1724. [PMID: 38216779 DOI: 10.1007/s00259-023-06584-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 12/11/2023] [Indexed: 01/14/2024]
Abstract
PURPOSE Enzymolysis clearance strategy, characterized by releasing the non-reabsorbable radioactive fragment under the specific cleavage of enzymes, is confirmed to be a safe and effective way to reduce the renal radioactivity accumulation in mice. However, the effectiveness of this strategy in humans remains unknown. Human epidermal growth factor receptor 2 (HER2) is overexpressed in various types of tumors, and radiolabeled HER2 Affibody is believed to be an attractive tool for HER2-targeted theranostics. However, its wide application is limited by the high and persistent renal uptake. In this study, we intend to validate the effectiveness of enzymolysis clearance strategy in reducing renal accumulation by using a modified HER2 Affibody. MATERIALS AND METHODS A new HER2 Affibody ligand, NOTA-MVK-ZHER2:2891, containing a cleavable Met-Val-Lys (MVK) linker was synthesized and labeled with 68Ga. The microPET imaging study was performed in SKOV-3 tumor mice to assess the uptakes of the control ligand and the MVK one in tumors and kidneys. Seven healthy volunteers were included for biodistribution and dosimetric studies with both the control and MVK ligands performed 1 week apart. Urine and blood samples from healthy volunteers were collected for in vivo metabolism study of the two ligands. Four HER2-positive and two HER2-negative patients were recruited for [68Ga]Ga-NOTA-MVK-ZHER2:2891 PET/CT imaging at 2 and 4 h post-injection (p.i.). RESULTS [68Ga]Ga-NOTA-MVK-ZHER2:2891 was stable both in PBS and in mouse serum. MicroPET images showed that the tumor uptake of [68Ga]Ga-NOTA-MVK-ZHER2:2891 was comparable to that of [68Ga]Ga-NOTA-ZHER2:2891 at all the time points, while the kidney uptake was significantly reduced 40 min p.i. (P < 0.05). The biodistribution study in healthy volunteers showed that the kidney uptake of MVK ligand was significantly lower than that of the control ligand at 1 h p.i. (P < 0.05), with the SUVmean of 34.3 and 45.8, respectively, while the uptakes of the two ligands in the other organs showed negligible difference. The effective doses of the MVK ligand and the control one were 26.1 and 28.7 µSv/MBq, respectively. The enzymolysis fragment of [68Ga]Ga-NOTA-Met-OH was observed in the urine samples of healthy volunteers injected with the MVK ligand, indicating that the enzymolysis clearance strategy worked in humans. The PET/CT study of patients showed that the range of SUVmax of HER2-positive lesions was 9.4-21, while that of HER2-negative lesions was 2.7-6.2, which suggested that the MVK modification did not affect the ability of ZHER2:2891 structure to bind with HER2. CONCLUSION We for the first time demonstrated that enzymolysis clearance strategy can effectively reduce renal radioactivity accumulation in humans. This strategy is expected to decrease renal radiation dose of peptide and small protein-based radiotracers, especially in the field of radionuclide therapy.
Collapse
Affiliation(s)
- Mingru Zhang
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shannxi, China
| | - Fei Kang
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shannxi, China
| | - Tong Xing
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shannxi, China
| | - Junling Wang
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shannxi, China
| | - Taoqi Ma
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shannxi, China
| | - Guiyu Li
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shannxi, China
| | - Zhiyong Quan
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shannxi, China
| | - Weidong Yang
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shannxi, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 119074, Singapore.
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore.
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore.
| | - Jing Wang
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shannxi, China.
| |
Collapse
|
9
|
Becx MN, Minczeles NS, Brabander T, de Herder WW, Nonnekens J, Hofland J. A Clinical Guide to Peptide Receptor Radionuclide Therapy with 177Lu-DOTATATE in Neuroendocrine Tumor Patients. Cancers (Basel) 2022; 14:cancers14235792. [PMID: 36497273 PMCID: PMC9737149 DOI: 10.3390/cancers14235792] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/18/2022] [Accepted: 11/21/2022] [Indexed: 11/27/2022] Open
Abstract
Peptide receptor radionuclide therapy (PRRT) with [177Lu]Lu-[DOTA0,Tyr3]-octreotate (177Lu-DOTATATE) has become an established second- or third-line treatment option for patients with somatostatin receptor (SSTR)-positive advanced well-differentiated gastroenteropancreatic (GEP) neuroendocrine tumors (NETs). Clinical evidence of the efficacy of PRRT in tumor control has been proven and lower risks of disease progression or death are seen combined with an improved quality of life. When appropriate patient selection is performed, PRRT is accompanied by limited risks for renal and hematological toxicities. Treatment of NET patients with PRRT requires dedicated clinical expertise due to the biological characteristics of PRRT and specific characteristics of NET patients. This review provides an overview for clinicians dealing with NET on the history, molecular characteristics, efficacy, toxicity and relevant clinical specifics of PRRT.
Collapse
Affiliation(s)
- Morticia N. Becx
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
- Correspondence:
| | - Noémie S. Minczeles
- Department of Radiology & Nuclear Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
- Department of Internal Medicine, Section of Endocrinology, ENETS Center of Excellence, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Tessa Brabander
- Department of Radiology & Nuclear Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Wouter W. de Herder
- Department of Internal Medicine, Section of Endocrinology, ENETS Center of Excellence, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Julie Nonnekens
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
- Department of Radiology & Nuclear Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Johannes Hofland
- Department of Internal Medicine, Section of Endocrinology, ENETS Center of Excellence, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| |
Collapse
|
10
|
Spada F, Campana D, Lamberti G, Laudicella R, Dellamano R, Dellamano L, Leeuwenkamp O, Baldari S. [ 177Lu]Lu-DOTA-TATE versus standard of care in adult patients with gastro-enteropancreatic neuroendocrine tumours (GEP-NETs): a cost-consequence analysis from an Italian hospital perspective. Eur J Nucl Med Mol Imaging 2022; 49:2037-2048. [PMID: 34950969 PMCID: PMC9016001 DOI: 10.1007/s00259-021-05656-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 12/12/2021] [Indexed: 12/02/2022]
Abstract
PURPOSE To assess and compare clinical outcomes and costs, to the Italian healthcare system, of three therapeutic options approved in the management of adult patients with gastro-enteropancreatic neuroendocrine tumours (GEP-NETs). METHODS We compared the efficacy, safety, and costs of [177Lu]Lu-DOTA-TATE, everolimus (both originator and generic products), and sunitinib in patients with advanced GEP-NETs (NET G1 and G2) that had progressed following treatment with somatostatin analogs (SSAs). A cost-consequence model was developed and validated by a panel of clinical experts from three NET reference centres in Italy. The clinical outcomes included in the model were median progression-free survival and the incidence of grade 3 or 4 adverse events (AEs), as reported in pivotal clinical trials. The costs for acquisition and administration of each treatment, and of managing AEs, were calculated from the perspective of the Italian national health service. Treatment costs per progression-free month were calculated separately for patients with NETs of pancreatic (PanNETs; all three treatments) and gastrointestinal (GI-NETs; [177Lu]Lu-DOTA-TATE and everolimus only) origin. RESULTS In patients with PanNETs, total costs per progression-free month were €2989 for [177Lu]Lu-DOTA-TATE, €4975 for originator everolimus, €3472 for generic everolimus, and €5337 for sunitinib. In patients with GI-NETs, total costs per progression-free month were €3189 for [177Lu]Lu-DOTA-TATE, €4990 for originator everolimus, and €3483 for generic everolimus. CONCLUSIONS [177Lu]Lu-DOTA-TATE was associated with lower costs per progression-free month versus relevant treatment options in patients with GI-NETs or PanNETs (NET G1-G2; progressed following SSA treatment), although acquisition and administration costs are higher. These findings provide further economic arguments in the overall context of treatment decision-making.
Collapse
Affiliation(s)
- Francesca Spada
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology (IEO), IRCCS, Milan, Italy
| | - Davide Campana
- Division of Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via P. Albertoni 15, 40138, Bologna, Italy
| | - Giuseppe Lamberti
- Division of Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via P. Albertoni 15, 40138, Bologna, Italy
| | - Riccardo Laudicella
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and of Morpho-Functional Imaging, University of Messina, Viale Consolare Valeria n.1, Messina, Italy
| | | | | | - Oscar Leeuwenkamp
- Advanced Accelerator Applications/A Novartis Company, Geneva, Switzerland
| | - Sergio Baldari
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and of Morpho-Functional Imaging, University of Messina, Viale Consolare Valeria n.1, Messina, Italy.
| |
Collapse
|
11
|
Sakulpisuti C, Chamroonrat W, Tepmongkol S. Cutaneous Management after Extravasation of High-Concentrated Amino Acid Solution Administered for Renal Protection in PRRT. Tomography 2022; 8:356-363. [PMID: 35202194 PMCID: PMC8880062 DOI: 10.3390/tomography8010029] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/24/2022] [Accepted: 01/28/2022] [Indexed: 11/25/2022] Open
Abstract
High-concentrated amino acid solution is used to protect the kidneys during peptide receptor radionuclide therapy (PPRT) in patients with neuroendocrine tumors (NETs). Extravasation of the solution can cause cutaneous complications. In this study, we described a 66-year-old man with metastatic medullary thyroid cancer and a 32-year-old woman with metastatic pancreatic NET who developed cutaneous lesions caused by the extravasation of an amino acid solution (25 g of lysine and 25 g of arginine in 1 L of normal saline) during PRRT with [177Lu]Lu-DOTA-TATE. Both were treated conservatively, and these cutaneous lesions gradually improved. The patient with metastatic pancreatic NET rejected the amino acid infusion in subsequent cycles of PRRT and therefore received [177Lu]Lu-DOTA-TATE alone, and her serum creatinine level and estimated glomerular filtration rate (eGFR) remained normal for 2 months after the last treatment. These two cases revealed cutaneous complications resulting from high-concentrated amino acid solution during PRRT because of hyperosmolarity. Health care providers should be aware of this complication to ensure its prevention and appropriate management. Preserved renal function was demonstrated after [177Lu]Lu-DOTA-TATE treatment in the absence of the infusion of a high-concentrated amino acid solution. However, long-term follow-up of renal function is suggested.
Collapse
Affiliation(s)
- Chaninart Sakulpisuti
- Division of Nuclear Medicine, Department of Diagnostic and Therapeutic Radiology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand;
| | - Wichana Chamroonrat
- Division of Nuclear Medicine, Department of Diagnostic and Therapeutic Radiology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand;
- Correspondence:
| | - Supatporn Tepmongkol
- Division of Nuclear Medicine, Department of Radiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand;
- Chulalongkorn University Biomedical Imaging Group (CUBIG), Department of Radiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
12
|
Stolniceanu CR, Moscalu M, Azoicai D, Tamba B, Volovat C, Grierosu I, Ionescu T, Jalloul W, Ghizdovat V, Gherasim R, Volovat S, Wang F, Fu J, Moscalu R, Matovic M, Stefanescu C. Improved Personalised Neuroendocrine Tumours' Diagnosis Predictive Power by New Receptor Somatostatin Image Processing Quantification. J Pers Med 2021; 11:jpm11101042. [PMID: 34683183 PMCID: PMC8539645 DOI: 10.3390/jpm11101042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 09/28/2021] [Accepted: 10/13/2021] [Indexed: 11/24/2022] Open
Abstract
Although neuroendocrine tumours (NETs) are intensively studied, their diagnosis and consequently personalised therapy management is still puzzling due to their tumoral heterogeneity. In their theragnosis algorithm, receptor somatostatin scintigraphy takes the central place, the diagnosis receptor somatostatin analogue (RSA) choice depending on laboratory experience and accessibility. However, in all cases, the results depend decisively on correct radiotracer tumoral uptake quantification, where unfortunately there are still unrevealed clues and lack of standardization. We propose an improved method to quantify the biodistribution of gamma-emitting RSA, using tissular corrected uptake indices. We conducted a bi-centric retrospective study on 101 patients with different types of NETs. Three uptake indices obtained after applying new corrections to areas of interest drawn for the tumour and for three reference organs (liver, spleen and lung) were statistically analysed. For the corrected pathological uptake indices, the results showed a significant decrease in the error of estimating the occurrence of errors and an increase in the diagnostic predictive power for NETs, especially in the case of lung-referring corrected index. In conclusion, these results support the importance of corrected uptake indices use in the analysis of 99mTcRSA biodistribution for a better personalised diagnostic accuracy of NETs patients.
Collapse
Affiliation(s)
- Cati Raluca Stolniceanu
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (C.R.S.); (I.G.); (T.I.); (W.J.); (V.G.); (R.G.); (C.S.)
| | - Mihaela Moscalu
- Department of Preventive Medicine and Interdisciplinarity, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Correspondence:
| | - Doina Azoicai
- Department of Epidemiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Bogdan Tamba
- Advanced Center for Research and Development in Experimental Medicine (CEMEX), “Grigore T. Popa” University of Medicine and Pharmacy, 700454 Iasi, Romania;
| | - Constantin Volovat
- Department of Medicine III—Medical Oncology-Radiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (C.V.); (S.V.)
| | - Irena Grierosu
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (C.R.S.); (I.G.); (T.I.); (W.J.); (V.G.); (R.G.); (C.S.)
| | - Teodor Ionescu
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (C.R.S.); (I.G.); (T.I.); (W.J.); (V.G.); (R.G.); (C.S.)
| | - Wael Jalloul
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (C.R.S.); (I.G.); (T.I.); (W.J.); (V.G.); (R.G.); (C.S.)
| | - Vlad Ghizdovat
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (C.R.S.); (I.G.); (T.I.); (W.J.); (V.G.); (R.G.); (C.S.)
| | - Roxana Gherasim
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (C.R.S.); (I.G.); (T.I.); (W.J.); (V.G.); (R.G.); (C.S.)
| | - Simona Volovat
- Department of Medicine III—Medical Oncology-Radiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (C.V.); (S.V.)
| | - Feng Wang
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; (F.W.); (J.F.)
| | - Jingjing Fu
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; (F.W.); (J.F.)
| | - Roxana Moscalu
- Manchester Academic Health Science Centre, School of Medical Sciences Manchester, The University of Manchester, Manchester M139PT, UK;
| | - Milovan Matovic
- Clinical Center Kragujevac, Center for Nuclear Medicine, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Cipriana Stefanescu
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (C.R.S.); (I.G.); (T.I.); (W.J.); (V.G.); (R.G.); (C.S.)
| |
Collapse
|
13
|
Xu J. Current treatments and future potential of surufatinib in neuroendocrine tumors (NETs). Ther Adv Med Oncol 2021; 13:17588359211042689. [PMID: 34484432 PMCID: PMC8411625 DOI: 10.1177/17588359211042689] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 08/11/2021] [Indexed: 02/06/2023] Open
Abstract
Neuroendocrine tumors (NETs) are rare, heterogeneous, often indolent tumors that predominantly originate in the lungs and gastrointestinal tract. An understanding of the biology and tumor microenvironment of NETs has led to the development of molecularly targeted treatment options including somatostatin analogs, tyrosine kinase inhibitors, mammalian target of rapamycin inhibitors and peptide receptor radionuclide therapy. Although increases in progression-free survival have been demonstrated, most currently approved NET therapies are limited by the development of tumor resistance. Surufatinib (HMPL-012, previously known as sulfatinib) is a new, oral, small-molecule tyrosine kinase inhibitor that potently inhibits vascular endothelial growth-factor receptor 1-3, fibroblast growth-factor receptor 1, and colony-stimulating-factor-1 receptor. This unique combination of molecular activities inhibits tumor angiogenesis, regulates tumor-immune evasion, and may decrease tumor resistance. Surufatinib demonstrated statistically significant, clinically meaningful antitumor activity, including tumor shrinkage, in two phase III studies recently completed in China in advanced pancreatic NETs and advanced extrapancreatic NETs. The safety profile of surufatinib in neuroendocrine tumors studies was consistent with previous surufatinib clinical studies. In an ongoing study in United States (US) patients with NETs of pancreatic origin and NETs of extrapancreatic origin previously treated with everolimus or sunitinib, surufatinib has also demonstrated promising efficacy. Furthermore, the pharmacokinetic and safety profile of surufatinib in US patients is similar to data collected in studies done in China. These positive phase III results support the efficacy of surufatinib in patients with advanced, progressive, well-differentiated NETs regardless of tumor origin.
Collapse
Affiliation(s)
- Jianming Xu
- Department of Gastrointestinal Oncology, The
Fifth Medical Center, Chinese PLA General Hospital, No. 8 East Street,
Fengtai District, Beijing 100071, China
| |
Collapse
|
14
|
Liberini V, Huellner MW, Grimaldi S, Finessi M, Thuillier P, Muni A, Pellerito RE, Papotti MG, Piovesan A, Arvat E, Deandreis D. The Challenge of Evaluating Response to Peptide Receptor Radionuclide Therapy in Gastroenteropancreatic Neuroendocrine Tumors: The Present and the Future. Diagnostics (Basel) 2020; 10:E1083. [PMID: 33322819 PMCID: PMC7763988 DOI: 10.3390/diagnostics10121083] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/09/2020] [Accepted: 12/09/2020] [Indexed: 02/07/2023] Open
Abstract
The NETTER-1 study has proven peptide receptor radionuclide therapy (PRRT) to be one of the most effective therapeutic options for metastatic neuroendocrine tumors (NETs), improving progression-free survival and overall survival. However, PRRT response assessment is challenging and no consensus on methods and timing has yet been reached among experts in the field. This issue is owed to the suboptimal sensitivity and specificity of clinical biomarkers, limitations of morphological response criteria in slowly growing tumors and necrotic changes after therapy, a lack of standardized parameters and timing of functional imaging and the heterogeneity of PRRT protocols in the literature. The aim of this article is to review the most relevant current approaches for PRRT efficacy prediction and response assessment criteria in order to provide an overview of suitable tools for safe and efficacious PRRT.
Collapse
Affiliation(s)
- Virginia Liberini
- Nuclear Medicine Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (S.G.); (M.F.); (P.T.); (D.D.)
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland;
| | - Martin W. Huellner
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland;
| | - Serena Grimaldi
- Nuclear Medicine Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (S.G.); (M.F.); (P.T.); (D.D.)
| | - Monica Finessi
- Nuclear Medicine Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (S.G.); (M.F.); (P.T.); (D.D.)
| | - Philippe Thuillier
- Nuclear Medicine Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (S.G.); (M.F.); (P.T.); (D.D.)
- Department of Endocrinology, University Hospital of Brest, 29200 Brest, France
| | - Alfredo Muni
- Department of Nuclear Medicine, S.S. Biagio e Antonio e C. Arrigo Hospital, 15121 Alessandria, Italy;
| | | | - Mauro G. Papotti
- Pathology Unit, City of Health and Science University Hospital, 10126 Turin, Italy;
- Department of Oncology, University of Turin at Molinette Hospital, 10126 Turin, Italy
| | - Alessandro Piovesan
- Department of Endocrinology, A. O. U. Città della Salute della Scienza of Turin, 10126 Turin, Italy;
| | - Emanuela Arvat
- Oncological Endocrinology, Department of Medical Sciences, University of Turin, 10126 Turin, Italy;
| | - Désirée Deandreis
- Nuclear Medicine Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (S.G.); (M.F.); (P.T.); (D.D.)
| |
Collapse
|