1
|
Keane G, Lam M, Braat A, Bruijnen R, Kaufmann N, de Jong H, Smits M. Transarterial Radioembolization (TARE) Global Practice Patterns: An International Survey by the Cardiovascular and Interventional Radiology Society of Europe (CIRSE). Cardiovasc Intervent Radiol 2024; 47:1224-1236. [PMID: 38914769 PMCID: PMC11379766 DOI: 10.1007/s00270-024-03768-z] [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] [Accepted: 05/12/2024] [Indexed: 06/26/2024]
Abstract
PURPOSE An international survey was conducted by the Cardiovascular Interventional Radiological Society of Europe (CIRSE) to evaluate radioembolization practice and capture opinions on real-world clinical and technical aspects of this therapy. MATERIALS AND METHODS A survey with 32 multiple choice questions was sent as an email to CIRSE members between November and December 2022. CIRSE group member and sister societies promoted the survey to their local members. The dataset was cleaned of duplicates and entries with missing data, and the resulting anonymized dataset was analysed. Data were presented using descriptive statistics. RESULTS The survey was completed by 133 sites, from 30 countries, spanning 6 continents. Most responses were from European centres (87/133, 65%), followed by centres from the Americas (22/133, 17%). Responding sites had been performing radioembolization for 10 years on average and had completed a total of 20,140 procedures over the last 5 years. Hepatocellular carcinoma treatments constituted 56% of this total, colorectal liver metastasis 17% and cholangiocarcinoma 14%. New sites had opened every year for the past 20 years, indicating the high demand for this therapy. Results showed a trend towards individualized treatment, with 79% of responders reporting use of personalized dosimetry for treatment planning and 97% reporting routine assessment of microsphere distribution post-treatment. Interventional radiologists played an important role in referrals, being present in the referring multi-disciplinary team in 91% of responding centres. CONCLUSION This survey provides insight into the current state of radioembolization practice globally. The results reveal the increasing significance placed on dosimetry, evolving interventional techniques and increased technology integration.
Collapse
Affiliation(s)
- Grace Keane
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, 3508 GA, Utrecht, The Netherlands.
| | - Marnix Lam
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, 3508 GA, Utrecht, The Netherlands
| | - Arthur Braat
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, 3508 GA, Utrecht, The Netherlands
| | - Rutger Bruijnen
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, 3508 GA, Utrecht, The Netherlands
| | - Nathalie Kaufmann
- Next Research, Contract Research Organization, Vienna, Austria
- Clinical Research, Cardiovascular and Interventional Radiological Society of Europe, Vienna, Austria
| | - Hugo de Jong
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, 3508 GA, Utrecht, The Netherlands
| | - Maarten Smits
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, 3508 GA, Utrecht, The Netherlands
| |
Collapse
|
2
|
Flux G, Gleisner KS, Bardies M. Physics for molecular radiotherapy - The battle between science and simplicity. Phys Med 2024; 124:103413. [PMID: 38880679 DOI: 10.1016/j.ejmp.2024.103413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/18/2024] Open
Affiliation(s)
- Glenn Flux
- Joint Dept of Physics, Royal Marsden Hospital & Institute of Cancer Research, Sutton, UK.
| | | | - Manuel Bardies
- Department of Nuclear Medicine, Institut du Cancer de Montpellier, Université de Montpellier, Montpellier, France; Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Montpellier, France
| |
Collapse
|
3
|
Ramonaheng K, Qebetu M, Ndlovu H, Swanepoel C, Smith L, Mdanda S, Mdlophane A, Sathekge M. Activity quantification and dosimetry in radiopharmaceutical therapy with reference to 177Lutetium. FRONTIERS IN NUCLEAR MEDICINE (LAUSANNE, SWITZERLAND) 2024; 4:1355912. [PMID: 39355215 PMCID: PMC11440950 DOI: 10.3389/fnume.2024.1355912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 03/12/2024] [Indexed: 10/03/2024]
Abstract
Radiopharmaceutical therapy has been widely adopted owing primarily to the development of novel radiopharmaceuticals. To fully utilize the potential of these RPTs in the era of precision medicine, therapy must be optimized to the patient's tumor characteristics. The vastly disparate dosimetry methodologies need to be harmonized as the first step towards this. Multiple factors play a crucial role in the shift from empirical activity administration to patient-specific dosimetry-based administrations from RPT. Factors such as variable responses seen in patients with presumably similar clinical characteristics underscore the need to standardize and validate dosimetry calculations. These efforts combined with ongoing initiatives to streamline the dosimetry process facilitate the implementation of radiomolecular precision oncology. However, various challenges hinder the widespread adoption of personalized dosimetry-based activity administration, particularly when compared to the more convenient and resource-efficient approach of empiric activity administration. This review outlines the fundamental principles, procedures, and methodologies related to image activity quantification and dosimetry with a specific focus on 177Lutetium-based radiopharmaceuticals.
Collapse
Affiliation(s)
- Keamogetswe Ramonaheng
- Department of Medical Physics and Radiobiology, Nuclear Medicine Research, Infrastructure (NuMeRI) NPC, Pretoria, South Africa
- Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria, South Africa
- Department of Nuclear Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Milani Qebetu
- Department of Medical Physics and Radiobiology, Nuclear Medicine Research, Infrastructure (NuMeRI) NPC, Pretoria, South Africa
- Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria, South Africa
| | - Honest Ndlovu
- Department of Medical Physics and Radiobiology, Nuclear Medicine Research, Infrastructure (NuMeRI) NPC, Pretoria, South Africa
- Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria, South Africa
- Department of Nuclear Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Cecile Swanepoel
- Department of Medical Physics and Radiobiology, Nuclear Medicine Research, Infrastructure (NuMeRI) NPC, Pretoria, South Africa
- Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria, South Africa
| | - Liani Smith
- Department of Medical Physics and Radiobiology, Nuclear Medicine Research, Infrastructure (NuMeRI) NPC, Pretoria, South Africa
- Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria, South Africa
| | - Sipho Mdanda
- Department of Medical Physics and Radiobiology, Nuclear Medicine Research, Infrastructure (NuMeRI) NPC, Pretoria, South Africa
- Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria, South Africa
- Department of Nuclear Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Amanda Mdlophane
- Department of Medical Physics and Radiobiology, Nuclear Medicine Research, Infrastructure (NuMeRI) NPC, Pretoria, South Africa
- Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria, South Africa
- Department of Nuclear Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Mike Sathekge
- Department of Medical Physics and Radiobiology, Nuclear Medicine Research, Infrastructure (NuMeRI) NPC, Pretoria, South Africa
- Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria, South Africa
- Department of Nuclear Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
4
|
Yadav S, Lawhn-Heath C, Paciorek A, Lindsay S, Mirro R, Bergsland EK, Hope TA. The Impact of Posttreatment Imaging in Peptide Receptor Radionuclide Therapy. J Nucl Med 2024; 65:409-415. [PMID: 38428966 DOI: 10.2967/jnumed.123.266614] [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: 09/05/2023] [Accepted: 12/20/2023] [Indexed: 03/03/2024] Open
Abstract
Posttreatment imaging of γ-emissions after peptide receptor radionuclide therapy (PRRT) can be used to perform quantitative dosimetry as well as assessment response using qualitative measures. We aimed to assess the impact of qualitative posttreatment imaging on the management of patients undergoing PRRT. Methods: In this retrospective study, we evaluated 100 patients with advanced well-differentiated neuroendocrine tumors undergoing PRRT, who had posttreatment SPECT/CT imaging at 24 h. First, we evaluated the qualitative assessment of response at each cycle. Then using a chart review, we determined the impact on management from the posttreatment imaging. The changes in management were categorized as major or minor, and the cycles at which these changes occurred were noted. Additionally, tumor grade was also evaluated. Results: Of the 100 sequential patients reviewed, most (80% after cycle 2, 79% after cycle 3, and 73% after cycle 4) showed qualitatively stable disease during PRRT. Management changes were observed in 27% (n = 27) of patients; 78% of those (n = 21) were major, and 30% (n = 9) were minor. Most treatment changes occurred after cycle 2 (33% major, 67% minor) and cycle 3 (62% major, 33% minor). Higher tumor grade correlated with increased rate of changes in management (P = 0.006). Conclusion: In this retrospective study, qualitative analysis of posttreatment SPECT/CT imaging informed changes in management in 27% of patients. Patients with higher-grade tumors had a higher rate of change in management, and most of the management changes occurred after cycles 2 and 3. Incorporating posttreatment imaging into standard PRRT workflows could potentially enhance patient management.
Collapse
Affiliation(s)
- Surekha Yadav
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Courtney Lawhn-Heath
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Alan Paciorek
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California
| | - Sheila Lindsay
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
- Department of Medicine, Division of Medical Oncology, University of California San Francisco, San Francisco, California
| | - Rebecca Mirro
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Emily K Bergsland
- Department of Medicine, Division of Medical Oncology, University of California San Francisco, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California; and
| | - Thomas A Hope
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California;
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California; and
- Department of Radiology, San Francisco VA Medical Center, San Francisco, California
| |
Collapse
|
5
|
Kayal G, Barbosa N, Marín CC, Ferrer L, Fragoso-Negrín JA, Grosev D, Gupta SK, Hidayati NR, Moalosi TCG, Poli GL, Thakral P, Tsapaki V, Vauclin S, Vergara-Gil A, Knoll P, Hobbs RF, Bardiès M. Quality Assurance Considerations in Radiopharmaceutical Therapy Dosimetry Using PLANETDose: An International Atomic Energy Agency Study. J Nucl Med 2024; 65:125-131. [PMID: 37884334 PMCID: PMC10755524 DOI: 10.2967/jnumed.122.265340] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 07/25/2023] [Indexed: 10/28/2023] Open
Abstract
Implementation of radiopharmaceutical therapy dosimetry varies depending on the clinical application, dosimetry protocol, software, and ultimately the operator. Assessing clinical dosimetry accuracy and precision is therefore a challenging task. This work emphasizes some pitfalls encountered during a structured analysis, performed on a single-patient dataset consisting of SPECT/CT images by various participants using a standard protocol and clinically approved commercial software. Methods: The clinical dataset consisted of the dosimetric study of a patient administered with [177Lu]Lu-DOTATATE at Tygerberg Hospital, South Africa, as a part of International Atomic Energy Agency-coordinated research project E23005. SPECT/CT images were acquired at 5 time points postinjection. Patient and calibration images were reconstructed on a workstation, and a calibration factor of 122.6 Bq/count was derived independently and provided to the participants. A standard dosimetric protocol was defined, and PLANETDose (version 3.1.1) software was installed at 9 centers to perform the dosimetry of 3 treatment cycles. The protocol included rigid image registration, segmentation (semimanual for organs, activity threshold for tumors), and dose voxel kernel convolution of activity followed by absorbed dose (AD) rate integration to obtain the ADs. Iterations of the protocol were performed by participants individually and within collective training, the results of which were analyzed for dosimetric variability, as well as for quality assurance and error analysis. Intermediary checkpoints were developed to understand possible sources of variation and to differentiate user error from legitimate user variability. Results: Initial dosimetric results for organs (liver and kidneys) and lesions showed considerable interoperator variability. Not only was the generation of intermediate checkpoints such as total counts, volumes, and activity required, but also activity-to-count ratio, activity concentration, and AD rate-to-activity concentration ratio to determine the source of variability. Conclusion: When the same patient dataset was analyzed using the same dosimetry procedure and software, significant disparities were observed in the results despite multiple sessions of training and feedback. Variations due to human error could be minimized or avoided by performing intensive training sessions, establishing intermediate checkpoints, conducting sanity checks, and cross-validating results across physicists or with standardized datasets. This finding promotes the development of quality assurance in clinical dosimetry.
Collapse
Affiliation(s)
- Gunjan Kayal
- CRCT, UMR 1037, INSERM, Université Toulouse III Paul Sabatier, Toulouse, France
- SCK CEN, Belgian Nuclear Research Centre, Mol, Belgium
| | | | | | - Ludovic Ferrer
- Medical Physics Department, ICO René Gauducheau, Nantes, France
- CRCINA, UMR 1232, INSERM, France
| | - José-Alejandro Fragoso-Negrín
- DOSIsoft SA, Cachan, France
- IRCM, UMR 1194 INSERM, Universite de Montpellier and Institut Regional du Cancer de Montpellier, Montpellier, France
| | - Darko Grosev
- Department of Nuclear Medicine and Radiation Protection, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Santosh Kumar Gupta
- Department of Nuclear Medicine and PET, Mahamana Pandit Madanmohan Malviya Cancer Centre and Homi Bhabha Cancer Centre, Varanasi, India
| | - Nur Rahmah Hidayati
- Research Center and Technology for Radiation Safety and Metrology-National Research and Innovation Agency, Jakarta, Indonesia
| | - Tumelo C G Moalosi
- Department of Medical Imaging and Clinical Oncology, Medical Physics, Nuclear Medicine Division, Faculty of Medicine and Health Science, Stellenbosch University, Tygerberg Hospital, Cape Town, South Africa
| | - Gian Luca Poli
- Department of Medical Physics, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Parul Thakral
- Department of Nuclear Medicine, Fortis Memorial Research Institute, Gurugram, India
| | - Virginia Tsapaki
- Dosimetry and Medical Radiation Physics, International Atomic Energy Agency, Vienna, Austria
| | | | - Alex Vergara-Gil
- CRCT, UMR 1037, INSERM, Université Toulouse III Paul Sabatier, Toulouse, France
| | - Peter Knoll
- Dosimetry and Medical Radiation Physics, International Atomic Energy Agency, Vienna, Austria
| | - Robert F Hobbs
- Johns Hopkins Medical Institute, Baltimore, Maryland; and
| | - Manuel Bardiès
- IRCM, UMR 1194 INSERM, Universite de Montpellier and Institut Regional du Cancer de Montpellier, Montpellier, France;
- Département de Médecine Nucléaire, Institut Régional du Cancer de Montpellier, Montpellier, France
| |
Collapse
|
6
|
di Santo G, Santo G, Sviridenko A, Virgolini I. Peptide receptor radionuclide therapy combinations for neuroendocrine tumours in ongoing clinical trials: status 2023. Theranostics 2024; 14:940-953. [PMID: 38250038 PMCID: PMC10797289 DOI: 10.7150/thno.91268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 12/04/2023] [Indexed: 01/23/2024] Open
Abstract
A growing body of literature reports on the combined use of peptide receptor radionuclide therapy (PRRT) with other anti-tumuor therapies in order to anticipate synergistic effects with perhaps increased safety issues. Combination treatments to enhance PRRT outcome are based on improved tumour perfusion, upregulation of somatostatin receptors (SSTR), radiosensitization with DNA damaging agents or targeted therapies. Several Phase 1 or 2 trials are currently recruiting patients in combined regimens. The combination of PRRT with cytotoxic chemotherapy, capecitabine and temozolomide (CAPTEM), seems to become clinically useful especially in pancreatic neuroendocrine tumours (pNETs) with acceptable safety profile. Neoadjuvant PRRT prior to surgery, PRRT combinations of intravenous and intraarterial routes of application, combinations of PRRT with differently radiolabelled (alpha, beta, Auger) SSTR-targeting agonists and antagonists, inhibitors of immune checkpoints (ICIs), poly (ADP-ribose) polymerase-1 (PARP1i), tyrosine kinase (TKI), DNA-dependent protein kinase, ribonucleotide reductase or DNA methyltransferase (DMNT) are tested in currently ongoing clinical trials. The combination with [131I]I-MIBG in rare NETs (such as paraganglioma, pheochromocytoma) and new non-SSTR-targeting radioligands are used in the personalization process of treatment. The present review will provide an overview of the current status of ongoing PRRT combination treatments.
Collapse
Affiliation(s)
- Gianpaolo di Santo
- Department of Nuclear Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Giulia Santo
- Department of Nuclear Medicine, Medical University of Innsbruck, Innsbruck, Austria
- Department of Experimental and Clinical Medicine, “Magna Graecia” University of Catanzaro, Catanzaro, Italy
| | - Anna Sviridenko
- Department of Nuclear Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Irene Virgolini
- Department of Nuclear Medicine, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
7
|
Peters S, Tran-Gia J, Agius S, Ivashchenko OV, Badel JN, Cremonesi M, Kurth J, Gabiña PM, Richetta E, Gleisner KS, Tipping J, Bardiès M, Stokke C. Implementation of dosimetry for molecular radiotherapy; results from a European survey. Phys Med 2024; 117:103196. [PMID: 38104033 DOI: 10.1016/j.ejmp.2023.103196] [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: 06/19/2023] [Revised: 10/13/2023] [Accepted: 12/07/2023] [Indexed: 12/19/2023] Open
Abstract
PURPOSE The use of molecular radiotherapy (MRT) has been rapidly evolving over the last years. The aim of this study was to assess the current implementation of dosimetry for MRTs in Europe. METHODS A web-based questionnaire was open for treating centres between April and June 2022, and focused on 2020-2022. Questions addressed the application of 16 different MRTs, the availability and involvement of medical physicists, software used, quality assurance, as well as the target regions for dosimetry, whether treatment planning and/or verification were performed, and the dosimetric methods used. RESULTS A total of 173 responses suitable for analysis was received from centres performing MRT, geographically distributed over 27 European countries. Of these, 146 centres (84 %) indicated to perform some form of dosimetry, and 97 % of these centres had a medical physicist available and almost always involved in dosimetry. The most common MRTs were 131I-based treatments for thyroid diseases and thyroid cancer, and [223Ra]RaCl2 for bone metastases. The implementation of dosimetry varied widely between therapies, from almost all centres performing dosimetry-based planning for microsphere treatments to none for some of the less common treatments (like 32P sodium-phosphate for myeloproliferative disease and [89Sr]SrCl2 for bone metastases). CONCLUSIONS Over the last years, implementation of dosimetry, both for pre-therapeutic treatment planning and post-therapy absorbed dose verification, increased for several treatments, especially for microsphere treatments. For other treatments that have moved from research to clinical routine, the use of dosimetry decreased in recent years. However, there are still large differences both across and within countries.
Collapse
Affiliation(s)
- Steffie Peters
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Johannes Tran-Gia
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Sam Agius
- Medical Imaging Department and Radioisotope Unit, Mater Dei Hospital, Msida, Malta
| | - Oleksandra V Ivashchenko
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, Groningen, The Netherlands
| | - Jean Noël Badel
- Centre de Lutte Contre le Cancer Léon-Bérard, CREATIS CNRS UMR 5220 INSERM U 1044, Université de Lyon, INSA-Lyon, Lyon, France
| | - Marta Cremonesi
- Unit of Radiation Research, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Jens Kurth
- Department of Nuclear Medicine, Rostock University Medical Center, Rostock, Germany
| | - Pablo Minguez Gabiña
- Department of Medical Physics and Radiation Protection, Gurutzeta-Cruces University Hospital/Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | - Elisa Richetta
- Medical Physics Department, AO Ordine Mauriziano, Turin, Italy
| | | | - Jill Tipping
- The Christie NHS Foundation Trust, Nuclear Medicine, Manchester, UK
| | - Manuel Bardiès
- IRCM, UMR 1194 INSERM, Université de Montpellier and Institut Régional du Cancer de Montpellier (ICM), France & Département de Médecine Nucléaire, Institut Régional du Cancer de Montpellier (ICM), France
| | - Caroline Stokke
- Department of Physics and Computational Radiology, Oslo University Hospital, Oslo, Norway; Department of Physics, University of Oslo, Oslo, Norway.
| |
Collapse
|
8
|
Cicone F, Sjögreen Gleisner K, Sarnelli A, Indovina L, Gear J, Gnesin S, Kraeber-Bodéré F, Bischof Delaloye A, Valentini V, Cremonesi M. The contest between internal and external-beam dosimetry: The Zeno's paradox of Achilles and the tortoise. Phys Med 2024; 117:103188. [PMID: 38042710 DOI: 10.1016/j.ejmp.2023.103188] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/06/2023] [Accepted: 11/23/2023] [Indexed: 12/04/2023] Open
Abstract
Radionuclide therapy, also called molecular radiotherapy (MRT), has come of age, with several novel radiopharmaceuticals being approved for clinical use or under development in the last decade. External beam radiotherapy (EBRT) is a well-established treatment modality, with about half of all oncologic patients expected to receive at least one external radiation treatment over their disease course. The efficacy and the toxicity of both types of treatment rely on the interaction of radiation with biological tissues. Dosimetry played a fundamental role in the scientific and technological evolution of EBRT, and absorbed doses to the target and to the organs at risk are calculated on a routine basis. In contrast, in MRT the usefulness of internal dosimetry has long been questioned, and a structured path to include absorbed dose calculation is missing. However, following a similar route of development as EBRT, MRT treatments could probably be optimized in a significant proportion of patients, likely based on dosimetry and radiobiology. In the present paper we describe the differences and the similarities between internal and external-beam dosimetry in the context of radiation treatments, and we retrace the main stages of their development over the last decades.
Collapse
Affiliation(s)
- Francesco Cicone
- Department of Experimental and Clinical Medicine, "Magna Graecia" University of Catanzaro, Catanzaro, Italy; Nuclear Medicine Unit, "Mater Domini" University Hospital, Catanzaro, Italy.
| | | | - Anna Sarnelli
- Medical Physics Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Luca Indovina
- Department of Diagnostic Imaging, Oncological Radiotherapy and Hematology, Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Rome, Italy
| | - Jonathan Gear
- Joint Department of Physics, Royal Marsden NHSFT & Institute of Cancer Research, Sutton, UK
| | - Silvano Gnesin
- Institute of Radiation Physics, Lausanne University Hospital, Lausanne, Switzerland; University of Lausanne, Lausanne, Switzerland
| | - Françoise Kraeber-Bodéré
- Nantes Université, Université Angers, CHU Nantes, INSERM, CNRS, CRCI2NA, Médecine Nucléaire, F-44000 Nantes, France
| | | | - Vincenzo Valentini
- Department of Diagnostic Imaging, Oncological Radiotherapy and Hematology, Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Rome, Italy; Università Cattolica del Sacro Cuore, Rome, Italy
| | - Marta Cremonesi
- Unit of Radiation Research, IEO, European Institute of Oncology IRCCS, Milan, Italy
| |
Collapse
|
9
|
Sjögreen-Gleisner K, Flux G, Bacher K, Chiesa C, de Nijs R, Kagadis GC, Lima T, Georgosopoulou ML, Gabiña PM, Nekolla S, Peters S, Santos J, Sattler B, Stokke C, Tran-Gia J, Gilligan P, Bardiès M. EFOMP policy statement NO. 19: Dosimetry in nuclear medicine therapy - Molecular radiotherapy. Phys Med 2023; 116:103166. [PMID: 37926641 DOI: 10.1016/j.ejmp.2023.103166] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 10/22/2023] [Indexed: 11/07/2023] Open
Abstract
The European Council Directive 2013/59/Euratom (BSS Directive) includes optimisation of treatment with radiotherapeutic procedures based on patient dosimetry and verification of the absorbed doses delivered. The present policy statement summarises aspects of three directives relating to the therapeutic use of radiopharmaceuticals and medical devices, and outlines the steps needed for implementation of patient dosimetry for radioactive drugs. To support the transition from administrations of fixed activities to personalised treatments based on patient-specific dosimetry, EFOMP presents a number of recommendations including: increased networking between centres and disciplines to support data collection and development of codes-of-practice; resourcing to support an infrastructure that permits routine patient dosimetry; research funding to support investigation into individualised treatments; inter-disciplinary training and education programmes; and support for investigator led clinical trials. Close collaborations between the medical physicist and responsible practitioner are encouraged to develop a similar pathway as is routine for external beam radiotherapy and brachytherapy. EFOMP's policy is to promote the roles and responsibilities of medical physics throughout Europe in the development of molecular radiotherapy to ensure patient benefit. As the BSS directive is adopted throughout Europe, unprecedented opportunities arise to develop informed treatments that will mitigate the risks of under- or over-treatments.
Collapse
Affiliation(s)
| | - Glenn Flux
- Joint Department of Physics, Royal Marsden Hospital and Institute of Cancer Research, Sutton, Surrey, UK
| | - Klaus Bacher
- Medical Physics, Ghent University, Ghent, Belgium
| | - Carlo Chiesa
- Nuclear Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Robin de Nijs
- Department of Clinical Physiology and Nuclear Medicine, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - George C Kagadis
- 3DMI Research Group, Department of Medical Physics, University of Patras, Rion, Greece
| | - Thiago Lima
- Department of Radiology and Nuclear Medicine, Luzerner Kantonsspital, Lucerne, Switzerland/Faculty of Health Sciences and Medicine, University of Lucerne, Switzerland
| | | | - Pablo Minguez Gabiña
- Department of Medical Physics and Radiation Protection, Gurutzeta-Cruces University Hospital /Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | - Stephan Nekolla
- School of Medicine and Health, Department of Nuclear Medicine, Technical University Munich, Munich, Germany
| | - Steffie Peters
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Joao Santos
- Medical Physics, Radiobiology and Radiation Protection Group, IPO Porto Research Center, Portuguese Oncology Institute of Porto/Porto Comprehensive Cancer Center & Health Research Network, Porto, Portugal
| | - Bernhard Sattler
- Department of Nuclear Medicine, University of Leipzig Medical Centre, Leipzig, Germany
| | - Caroline Stokke
- Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway & Department of Physics, University of Oslo, Oslo, Norway
| | - Johannes Tran-Gia
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Paddy Gilligan
- Mater Misericordiae University Hospital, Dublin, Ireland
| | - Manuel Bardiès
- IRCM, UMR 1194 INSERM, Université de Montpellier and Institut Régional du Cancer de Montpellier (ICM), France & Département de Médecine Nucléaire, Institut Régional du Cancer de Montpellier (ICM), France
| |
Collapse
|
10
|
Gustafsson J, Taprogge J. Future trends for patient-specific dosimetry methodology in molecular radiotherapy. Phys Med 2023; 115:103165. [PMID: 37880071 DOI: 10.1016/j.ejmp.2023.103165] [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: 05/31/2023] [Revised: 10/03/2023] [Accepted: 10/17/2023] [Indexed: 10/27/2023] Open
Abstract
Molecular radiotherapy is rapidly expanding, and new radiotherapeutics are emerging. The majority of treatments is still performed using empirical fixed activities and not tailored for individual patients. Molecular radiotherapy dosimetry is often seen as a promising candidate that would allow personalisation of treatments as outcome should ultimately depend on the absorbed doses delivered and not the activities administered. The field of molecular radiotherapy dosimetry has made considerable progress towards the feasibility of routine clinical dosimetry with reasonably accurate absorbed-dose estimates for a range of molecular radiotherapy dosimetry applications. A range of challenges remain with respect to the accurate quantification, assessment of time-integrated activity and absorbed dose estimation. In this review, we summarise a range of technological and methodological advancements, mainly focussed on beta-emitting molecular radiotherapeutics, that aim to improve molecular radiotherapy dosimetry to achieve accurate, reproducible, and streamlined dosimetry. We describe how these new technologies can potentially improve the often time-consuming considered process of dosimetry and provide suggestions as to what further developments might be required.
Collapse
Affiliation(s)
| | - Jan Taprogge
- National Radiotherapy Trials Quality Assurance (RTTQA) Group, Joint Department of Physics, Royal Marsden NHSFT, Downs Road, Sutton SM2 5PT, United Kingdom; The Institute of Cancer Research, 123 Old Brompton Road, London SW7 3RP, United Kingdom
| |
Collapse
|
11
|
Mourik JEM, Derks M, Te Beek ET, Ten Broek MRJ. Gamma camera-specific reference standards for radioactive iodine uptake measurements. EJNMMI Phys 2023; 10:55. [PMID: 37702889 PMCID: PMC10499732 DOI: 10.1186/s40658-023-00575-2] [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: 01/31/2023] [Accepted: 09/05/2023] [Indexed: 09/14/2023] Open
Abstract
BACKGROUND Current guidelines of the radioiodine uptake (RAIU) test allow the use of different equipment, isotopes, activity and region-of-interest (ROI). We evaluated presence and extent of these differences in clinical practice and evaluated the effect of some of these variations on RAIU outcomes. Also, gamma camera-specific reference standards were calculated and retrospectively compared with measurements obtained during clinical RAIU tests. MATERIALS AND METHODS First, questionnaires were sent to Dutch nuclear medicine departments requesting information about equipment usage, isotope, isotope formulation, activity and measurement techniques. Secondly, a neck phantom containing a range of activities in capsule or water-dissolved formulation was scanned. Counts were measured using automatic ROI, square box ROI or all counts in the image. Thirdly, clinical RAIU data were collected during 2015-2018 using three different gamma cameras. Reference standards for each scanner were calculated using regression analysis between reference activity and measured counts. Uptake measurements using this gamma camera-specific reference standard were compared with original measurements. RESULTS The survey demonstrated significant differences in isotope, isotope formulation, activity, use of neck phantoms, frequency and duration of reference measurements, distance to collimator, use of background measurements and ROI delineation. The phantom study demonstrated higher counts for the water-dissolved formulation than capsules using both automatic and square box ROI. Also, higher counts were found using a square box ROI than an automatic ROI. The retrospective study showed feasibility of RAIU calculations using camera-specific reference standards and good correlation with the original RAIU measurements. CONCLUSIONS This study demonstrated considerable technical variation in RAIU measurement in clinical practice. The phantom study demonstrated that these differences could result in differences in count measurements, potentially resulting in different dose calculations for radioactive iodine therapy. Retrospective data suggest that camera-specific reference standards may be used instead of individual reference measurements using separate activity sources, which may thus eliminate some sources of variation.
Collapse
Affiliation(s)
- Jurgen E M Mourik
- Department of Nuclear Medicine, Franciscus Gasthuis & Vlietland Hospital, Kleiweg 500, 3045 PM, Rotterdam, The Netherlands.
| | - Mark Derks
- Department of Nuclear Medicine, IJsselland Hospital, Capelle aan den IJssel, The Netherlands
| | - Erik T Te Beek
- Department of Nuclear Medicine, Reinier de Graaf Hospital, Delft, The Netherlands
| | - Marc R J Ten Broek
- Department of Nuclear Medicine, IJsselland Hospital, Capelle aan den IJssel, The Netherlands
- Department of Nuclear Medicine, Reinier de Graaf Hospital, Delft, The Netherlands
| |
Collapse
|
12
|
Gabiña PM, Gleisner KS, Cremonesi M, Stokke C, Flux G, Cicone F, Konijnenberg M, Aldridge M, Sandstrom M, Chiesa C, Paphiti M, Hippeläinen E, Uribe C, Solny P, Gnesin S, Bernhardt P, Chouin N, Costa PF, Glatting G, Verburg F, Gear J. Results from an EANM survey on time estimates and personnel responsible for main tasks in molecular radiotherapy dosimetry. Eur J Nucl Med Mol Imaging 2023; 50:2595-2604. [PMID: 37129712 DOI: 10.1007/s00259-023-06215-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Affiliation(s)
- Pablo Mínguez Gabiña
- Department of Medical Physics and Radiation Protection, Gurutzeta-Cruces University Hospital/Biocruces Bizkaia Health Research Institute, Plaza Cruces S/N, 48903, Barakaldo, Spain
- Faculty of Engineering, Department of Applied Physics, UPV/EHU, Bilbao, Spain
| | | | - Marta Cremonesi
- Radiation Research Unit, Department of Medical Imaging and Radiation Sciences, Istituto Europeo Di Oncologia, Milan, Italy
| | - Caroline Stokke
- Department of Physics and Computational Radiology, Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
- Department of Physics, University of Oslo, Oslo, Norway
| | - Glenn Flux
- Joint Department of Physics, Royal Marsden NHSFT and Institute of Cancer Research, Sutton, UK
| | - Francesco Cicone
- Department of Experimental and Clinical Medicine, "Magna Graecia" University of Catanzaro, Catanzaro, Italy
- Nuclear Medicine Unit, University Hospital "Mater Domini", Catanzaro, Italy
| | - Mark Konijnenberg
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Matt Aldridge
- Maidstone and Tunbridge Wells NHS Trust, Maidstone Hospital, Maidstone, ME16 9QQ, UK
| | - Mattias Sandstrom
- Section of Nuclear Medicine and PET, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Carlo Chiesa
- Nuclear Medicine Division, Foundation IRCCS Istituto Nazionale Tumori, Via Giacomo Venezian 1, 20133, Milan, Italy
| | - Maria Paphiti
- Medical Physics Department, Pammakaristos Hospital of Divine Providence, Iakovaton 43, 11144, Athens, Greece
| | - Eero Hippeläinen
- Department of Clinical Physiology and Nuclear Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Carlos Uribe
- Functional Imaging, BC Cancer, Vancouver, BC, Canada
- Department of Radiology, University of British Columbia, Vancouver, BC, Canada
| | - Pavel Solny
- National Radiation Protection Institute, Bartoskova 1450/28, 140 00, Praha 4, Nusle, Czech Republic
| | - Silvano Gnesin
- Institute of Radiation Physics, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Peter Bernhardt
- Department of Medical Radiation Sciences, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Gothenburg, University, Gothenburg, Sweden
- Department of Medical Physics and Biomedical Engineering (MFT), Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Nicolas Chouin
- Nantes Université, Inserm, CNRS, Université d'Angers, Oniris, CRCI2NA, Nantes, France
| | - Pedro Fragoso Costa
- Department of Nuclear Medicine, West German Cancer Center, University of Duisburg-Essen, Essen, Germany
- Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
| | - Gerhard Glatting
- Medical Radiation Physics, Department of Nuclear Medicine, Ulm University, Ulm, Germany
| | - Frederik Verburg
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Jonathan Gear
- Joint Department of Physics, Royal Marsden NHSFT and Institute of Cancer Research, Sutton, UK.
| |
Collapse
|
13
|
Strigari L, Marconi R, Solfaroli-Camillocci E. Evolution of Portable Sensors for In-Vivo Dose and Time-Activity Curve Monitoring as Tools for Personalized Dosimetry in Molecular Radiotherapy. SENSORS (BASEL, SWITZERLAND) 2023; 23:2599. [PMID: 36904802 PMCID: PMC10007630 DOI: 10.3390/s23052599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 02/19/2023] [Accepted: 02/21/2023] [Indexed: 06/18/2023]
Abstract
Treatment personalization in Molecular Radiotherapy (MRT) relies on pre- and post-treatment SPECT/ PET-based images and measurements to obtain a patient-specific absorbed dose-rate distribution map and its evolution over time. Unfortunately, the number of time points that are available per patient to investigate individual pharmacokinetics is often reduced by limited patient compliance or SPECT or PET/CT scanner availability for dosimetry in busy departments. The adoption of portable sensors for in-vivo dose monitoring during the entire treatment could improve the assessment of individual biokinetics in MRT and, thus, the treatment personalization. The evolution of portable devices, non-SPECT/PET-based options, already used for monitoring radionuclide activity transit and accumulation during therapy with radionuclides (i.e., MRT or brachytherapy), is presented to identify valuable ones, which combined with conventional nuclear medicine imaging systems could be effective in MRT. External probes, integration dosimeters and active detecting systems were included in the study. The devices and their technology, the range of applications, the features and limitations are discussed. Our overview of the available technologies encourages research and development of portable devices and dedicated algorithms for MRT patient-specific biokinetics study. This would represent a crucial advancement towards personalized treatment in MRT.
Collapse
Affiliation(s)
- Lidia Strigari
- Department of Medical Physics, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Giuseppe Massarenti 9, 40138 Bologna, Italy
| | - Raffaella Marconi
- Scientific Direction, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144 Rome, Italy
| | | |
Collapse
|
14
|
Bly R. Radiation safety of current European practices of therapeutic nuclear medicine: survey results from 20 HERCA countries. JOURNAL OF RADIOLOGICAL PROTECTION : OFFICIAL JOURNAL OF THE SOCIETY FOR RADIOLOGICAL PROTECTION 2023; 43:011507. [PMID: 36599154 DOI: 10.1088/1361-6498/acafef] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 01/04/2023] [Indexed: 06/17/2023]
Abstract
The purpose of this study was to acquire up-to-date information on nuclear medicine treatments in Europe and on the implementation of the requirements of the Basic Safety Standards Directive in HERCA Heads of the European Radiological Protection Competent Authorities (HERCAs) member states. An electronic survey was distributed to competent authorities of 32 HERCA member states. The questionnaire addressed 33 explicitly considered treatments using 13 different radionuclides, and for each treatment, a similar set of questions was included. Questions covered the use of treatments, hospitalisation of patients and radioactive waste management related to therapeutic nuclear medicine involving other radionuclides than the well-known131I. The survey also covered justification of treatments, individual treatment planning, involvement of a medical physics expert (MPE) and radiation protection instructions given to the patient at the time of release. Responses were obtained from 20 HERCA countries. All of these countries used Na[131I]I for benign thyroid diseases and thyroid ablation of adults. 223RaCl2(Xofigo®) for bone metastases,177Lu-somatostatin analogues for neuroendocrine tumours and177Lu-labelled PSMA for castration resistant prostate cancer (PC) and PC-metastases were used in 90%, 65% and 55% of countries, respectively. Only a few countries had specific criteria for hospitalisation and waste management for new therapeutic nuclear medicine. Regulatory requirements for justification of new therapeutic nuclear medicine were in place in almost all countries. Individual treatment planning was required for all therapies in 55% and for some therapies in 28% of the responding countries. Implementation of the requirement for MPEs to be closely involved in nuclear medicine practices varied to a great extend among countries. Almost all responding countries answered that some radiation protection instructions existed for patients released after treatment with radionuclides other than131I treatment, however only few countries had developed specific guidelines in the field.
Collapse
Affiliation(s)
- Ritva Bly
- Radiation and Nuclear Safety Authority (STUK), Helsinki, Finland
| |
Collapse
|
15
|
Pirozzi Palmese V, D'Ambrosio L, Di Gennaro F, Maisto C, de Marino R, Morisco A, Coluccia S, Di Gennaro P, De Lauro F, Raddi M, Gaballo P, Tafuto S, Celentano E, Lastoria S. A comparison of simplified protocols of personalized dosimetry in NEN patients treated by radioligand therapy (RLT) with [ 177Lu]Lu-DOTATATE to favor its use in clinical practice. Eur J Nucl Med Mol Imaging 2023; 50:1753-1764. [PMID: 36688980 PMCID: PMC10119237 DOI: 10.1007/s00259-023-06112-8] [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: 08/10/2022] [Accepted: 01/05/2023] [Indexed: 01/24/2023]
Abstract
The role of internal dosimetry is usually proposed for investigational purposes in patients treated by RLT, even if its application is not yet the standard method in clinical practice. This limited use is partially justified by several concomitant factors that make calculations a complex process. Therefore, simplified dosimetry protocols are required. METHODS In our study, dosimetric evaluations were performed in thirty patients with NENs who underwent RLT with [177Lu]Lu-DOTATATE. The reference method (M0) calculated the cumulative absorbed dose performing dosimetry after each of the four cycles. Obtained data were employed to assess the feasibility of simplified protocols: defining the dosimetry only after the first cycle (M1) and after the first and last one (M2). RESULTS The mean differences of the cumulative absorbed doses between M1 and M0 were - 10% for kidney, - 5% for spleen, + 34% for liver, + 13% for red marrow, and + 37% for tumor lesions. Conversely, differences lower than ± 10% were measured between M2 and M0. CONCLUSION Cumulative absorbed doses obtained with the M2 protocol resembled the doses calculated by M0, while the M1 protocol overestimated the absorbed doses in all organs at risk, except for the spleen.
Collapse
Affiliation(s)
| | - Laura D'Ambrosio
- S.C. Medicina Nucleare E Terapia Radiometabolica, INT IRCCS Fondazione G. Pascale, Naples, Italy
| | - Francesca Di Gennaro
- S.C. Medicina Nucleare E Terapia Radiometabolica, INT IRCCS Fondazione G. Pascale, Naples, Italy
| | - Costantina Maisto
- S.C. Medicina Nucleare E Terapia Radiometabolica, INT IRCCS Fondazione G. Pascale, Naples, Italy
| | - Roberta de Marino
- S.C. Medicina Nucleare E Terapia Radiometabolica, INT IRCCS Fondazione G. Pascale, Naples, Italy
| | - Anna Morisco
- S.C. Medicina Nucleare E Terapia Radiometabolica, INT IRCCS Fondazione G. Pascale, Naples, Italy
| | - Sergio Coluccia
- S.C. Epidemiologia E Biostatistica, INT IRCCS Fondazione G. Pascale, Naples, Italy
| | | | - Francesco De Lauro
- S.C. Medicina Nucleare E Terapia Radiometabolica, INT IRCCS Fondazione G. Pascale, Naples, Italy
| | - Marco Raddi
- S.C. Medicina Nucleare E Terapia Radiometabolica, INT IRCCS Fondazione G. Pascale, Naples, Italy
| | - Paolo Gaballo
- S.C. Medicina Nucleare E Terapia Radiometabolica, INT IRCCS Fondazione G. Pascale, Naples, Italy
| | - Salvatore Tafuto
- S.C. Sarcomi E Tumori Rari, INT IRCCS Fondazione G. Pascale, Naples, Italy
| | - Egidio Celentano
- S.C. Epidemiologia E Biostatistica, INT IRCCS Fondazione G. Pascale, Naples, Italy
| | - Secondo Lastoria
- S.C. Medicina Nucleare E Terapia Radiometabolica, INT IRCCS Fondazione G. Pascale, Naples, Italy.
| |
Collapse
|
16
|
Havlena GT, Kapadia NS, Huang P, Song H, Engles J, Brechbiel M, Sgouros G, Wahl RL. Cure of Micrometastatic B-Cell Lymphoma in a SCID Mouse Model Using 213Bi-Anti-CD20 Monoclonal Antibody. J Nucl Med 2023; 64:109-116. [PMID: 35981897 PMCID: PMC9841256 DOI: 10.2967/jnumed.122.263962] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 05/25/2022] [Accepted: 05/25/2022] [Indexed: 01/28/2023] Open
Abstract
We studied the feasibility of using the α-emitting 213Bi-anti-CD20 therapy with direct bioluminescent tracking of micrometastatic human B-cell lymphoma in a SCID mouse model. Methods: A highly lethal SCID mouse model of minimal-tumor-burden disseminated non-Hodgkin lymphoma (NHL) was established using human Raji lymphoma cells transfected to express the luciferase reporter. In vitro and in vivo radioimmunotherapy experiments were conducted. Single- and multiple-dose regimens were explored, and results with 213Bi-rituximab were compared with various controls, including no treatment, free 213Bi radiometal, unlabeled rituximab, and 213Bi-labeled anti-HER2/neu (non-CD20-specific antibody). 213Bi-rituximab was also compared in vivo with the low-energy β-emitter 131I-tositumomab and the high-energy β-emitter 90Y-rituximab. Results: In vitro studies showed dose-dependent target-specific killing of lymphoma cells with 213Bi-rituximab. Multiple in vivo studies showed significant and specific tumor growth delays with 213Bi-rituximab versus free 213Bi, 213Bi-labeled control antibody, or unlabeled rituximab. Redosing of 213Bi-rituximab was more effective than single dosing. With a single dose of therapy given 4 d after intravenous tumor inoculation, disease in all untreated controls, and in all mice in the 925-kBq 90Y-rituximab group, progressed. With 3,700 kBq of 213Bi-rituximab, 75% of the mice survived and all but 1 survivor was cured. With 2,035 kBq of 131I-tositumomab, 75% of the mice were tumor-free by bioluminescent imaging and 62.5% survived. Conclusion: Cure of micrometastatic NHL is achieved in most animals treated 4 d after intravenous tumor inoculation using either 213Bi-rituximab or 131I-tositumomab, in contrast to the lack of cures with unlabeled rituximab or 90Y-rituximab or if there was a high tumor burden before radioimmunotherapy. α-emitter-labeled anti-CD20 antibodies are promising therapeutics for NHL, although a longer-lived α-emitter may be of greater efficacy.
Collapse
Affiliation(s)
| | | | - Peng Huang
- Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Hong Song
- Section of Nuclear Medicine, Stanford University School of Medicine, Stanford, California
| | - James Engles
- Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - George Sgouros
- Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Richard L. Wahl
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, St. Louis, Missouri
| |
Collapse
|
17
|
Gear J. Milestones in dosimetry for nuclear medicine therapy. Br J Radiol 2022; 95:20220056. [PMID: 35451857 PMCID: PMC10996314 DOI: 10.1259/bjr.20220056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/31/2022] [Accepted: 04/14/2022] [Indexed: 11/05/2022] Open
Abstract
Nuclear Medicine therapy has reached a critical juncture with an unprecedented number of patients being treated and an extensive list of new radiopharmaceuticals under development. Since the early applications of these treatments dosimetry has played a vital role in their development, in both aiding optimisation and enhancing safety and efficacy. To inform the future direction of this field, it is useful to reflect on the scientific and technological advances that have occurred since those early uses. In this review, we explore how dosimetry has evolved over the years and discuss why such initiatives were conceived and the importance of maintaining standards within our practise. Specific milestones and landmark publications are highlighted and a thematic review and significant outcomes during each decade are presented.
Collapse
Affiliation(s)
- Jonathan Gear
- The Joint Department of Physics, The Royal Marsden NHS
Foundation Trust & Institute of Cancer Research,
Sutton, United Kingdom
| |
Collapse
|
18
|
Nilsson JN, Siikanen J, Ihre Lundgren C, Ardenfors O. Dosimetric dependencies on target geometry and size in radioiodine therapy for differentiated thyroid cancer. Phys Med 2022; 99:68-72. [DOI: 10.1016/j.ejmp.2022.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 05/02/2022] [Accepted: 05/25/2022] [Indexed: 11/29/2022] Open
|
19
|
Correa-Alfonso CM, Withrow JD, Domal SJ, Xing S, Shin J, Grassberger C, Paganetti H, Bolch WE. A mesh-based model of liver vasculature: implications for improved radiation dosimetry to liver parenchyma for radiopharmaceuticals. EJNMMI Phys 2022; 9:28. [PMID: 35416550 PMCID: PMC9008118 DOI: 10.1186/s40658-022-00456-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 03/28/2022] [Indexed: 12/05/2022] Open
Abstract
Purpose To develop a model of the internal vasculature of the adult liver and demonstrate its application to the differentiation of radiopharmaceutical decay sites within liver parenchyma from those within organ blood. Method Computer-generated models of hepatic arterial (HA), hepatic venous (HV), and hepatic portal venous (HPV) vascular trees were algorithmically created within individual lobes of the ICRP adult female and male livers (AFL/AML). For each iteration of the algorithm, pressure, blood flow, and vessel radii within each tree were updated as each new vessel was created and connected to a viable bifurcation site. The vascular networks created inside the AFL/AML were then tetrahedralized for coupling to the PHITS radiation transport code. Specific absorbed fractions (SAF) were computed for monoenergetic alpha particles, electrons, positrons, and photons. Dual-region liver models of the AFL/AML were proposed, and particle-specific SAF values were computed assuming radionuclide decays in blood within two locations: (1) sites within explicitly modeled hepatic vessels, and (2) sites within the hepatic blood pool residing outside these vessels to include the capillaries and blood sinuses. S values for 22 and 10 radionuclides commonly used in radiopharmaceutical therapy and imaging, respectively, were computed using the dual-region liver models and compared to those obtained in the existing single-region liver model. Results Liver models with virtual vasculatures of ~ 6000 non-intersecting straight cylinders representing the HA, HPV, and HV circulations were created for the ICRP reference. For alpha emitters and for beta and auger-electron emitters, S values using the single-region models were approximately 11% (AML) to 14% (AFL) and 11% (AML) to 13% (AFL) higher than the S values obtained using the dual-region models, respectively. Conclusions The methodology employed in this study has shown improvements in organ parenchymal dosimetry through explicit consideration of blood self-dose for alpha particles (all energies) and for electrons at energies below ~ 100 keV.
Collapse
Affiliation(s)
- Camilo M Correa-Alfonso
- Medical Physics Program, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Julia D Withrow
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, 32611-6550, USA
| | - Sean J Domal
- Medical Physics Program, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Shu Xing
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Jungwook Shin
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02115, USA.,Radiation Epidemiology Branch, National Cancer Institute, Rockville, MD, 21704, USA
| | - Clemens Grassberger
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Harald Paganetti
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Wesley E Bolch
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, 32611-6550, USA.
| |
Collapse
|
20
|
Kayal G, Clayton N, Vergara-Gil A, Struelens L, Bardiès M. Proof-of-concept of DosiTest: A virtual multicentric clinical trial for assessing uncertainties in molecular radiotherapy dosimetry. Phys Med 2022; 97:25-35. [PMID: 35339863 DOI: 10.1016/j.ejmp.2022.03.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 01/19/2022] [Accepted: 03/14/2022] [Indexed: 11/16/2022] Open
Abstract
Clinical dosimetry in molecular radiotherapy (MRT) is a multi-step procedure, prone to uncertainties at every stage of the dosimetric workflow. These are difficult to assess, especially as some are complex or even impossible to measure experimentally. The DosiTest project was initiated to assess the variability associated with clinical dosimetry, by setting up a 'virtual' multicentric clinical dosimetry trial based on Monte Carlo (MC) modelling. A reference patient model with a realistic geometry and activity input for a specific tracer is considered. Reference absorbed dose rate distribution maps are generated at various time-points from MC modelling, combining precise information on density and activity distributions (voxel wise). Then, centre-specific calibration and patient SPECT/CT datasets are modelled, on which the clinical centres can perform clinical (i.e. image-based) dosimetry. The results of this dosimetric analysis can be benchmarked against the reference dosimetry to assess the variability induced by implementing different clinical dosimetry approaches. The feasibility of DosiTest is presented here for a clinical situation of therapeutic administration of 177Lu-DOTATATE (Lutathera®) peptide receptor radionuclide therapy (PRRT). From a real patient dataset composed of 5 SPECT/CT images and associated calibrations, we generated the reference absorbed dose rate images with GATE. Then, simulated SPECT/CT image generation based on GATE was performed, both for a calibration phantom and virtual patient images. Based on this simulated dataset, image-based dosimetry could be performed, and compared with reference dosimetry. The good agreement, between real and simulated images, and between reference and image-based dosimetry established the proof of concept of DosiTest.
Collapse
Affiliation(s)
- G Kayal
- CRCT, UMR 1037, INSERM, Université Toulouse III Paul Sabatier, Toulouse, France; SCK CEN, Belgian Nuclear Research Centre, Boeretang 200, Mol 2400, Belgium.
| | - N Clayton
- CRCT, UMR 1037, INSERM, Université Toulouse III Paul Sabatier, Toulouse, France
| | - A Vergara-Gil
- CRCT, UMR 1037, INSERM, Université Toulouse III Paul Sabatier, Toulouse, France
| | - L Struelens
- SCK CEN, Belgian Nuclear Research Centre, Boeretang 200, Mol 2400, Belgium
| | - M Bardiès
- ICM, Département de Médecine Nucléaire, Montpellier, France; IRCM, UMR 1194 INSERM, Université de Montpellier and ICM, Montpellier, France
| |
Collapse
|
21
|
Bockhold S, Foley SJ, Rainford LA, Corridori R, Eberstein A, Hoeschen C, Konijnenberg MW, Molyneux-Hodgson S, Paulo G, Santos J, McNulty JP. Exploring the translational challenge for medical applications of ionising radiation and corresponding radiation protection research. J Transl Med 2022; 20:137. [PMID: 35303930 PMCID: PMC8932076 DOI: 10.1186/s12967-022-03344-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 03/06/2022] [Indexed: 01/19/2023] Open
Abstract
Background Medical applications of ionising radiation and associated radiation protection research often encounter long delays and inconsistent implementation when translated into clinical practice. A coordinated effort is needed to analyse the research needs for innovation transfer in radiation-based high-quality healthcare across Europe which can inform the development of an innovation transfer framework tailored for equitable implementation of radiation research at scale. Methods Between March and September 2021 a Delphi methodology was employed to gain consensus on key translational challenges from a range of professional stakeholders. A total of three Delphi rounds were conducted using a series of electronic surveys comprised of open-ended and closed-type questions. The surveys were disseminated via the EURAMED Rocc-n-Roll consortium network and prominent medical societies in the field. Approximately 350 professionals were invited to participate. Participants’ level of agreement with each generated statement was captured using a 6-point Likert scale. Consensus was defined as median ≥ 4 with ≥ 60% of responses in the upper tertile of the scale. Additionally, the stability of responses across rounds was assessed. Results In the first Delphi round a multidisciplinary panel of 20 generated 127 unique statements. The second and third Delphi rounds recruited a broader sample of 130 individuals to rate the extent to which they agreed with each statement as a key translational challenge. A total of 60 consensus statements resulted from the iterative Delphi process of which 55 demonstrated good stability. Ten statements were identified as high priority challenges with ≥ 80% of statement ratings either ‘Agree’ or ‘Strongly Agree’. Conclusion A lack of interoperability between systems, insufficient resources, unsatisfactory education and training, and the need for greater public awareness surrounding the benefits, risks, and applications of ionising radiation were identified as principal translational challenges. These findings will help to inform a tailored innovation transfer framework for medical radiation research. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03344-4.
Collapse
Affiliation(s)
- Sophie Bockhold
- Radiography and Diagnostic Imaging, School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland.
| | - Shane J Foley
- Radiography and Diagnostic Imaging, School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Louise A Rainford
- Radiography and Diagnostic Imaging, School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | | | | | - Christoph Hoeschen
- Institute of Medical Engineering, Otto Von Guericke Universität Magdeburg, Magdeburg, Germany
| | - Mark W Konijnenberg
- Department of Radiology and Nuclear Medicine, Erasmus Medical Centre, Rotterdam, Netherlands
| | | | - Graciano Paulo
- Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Coimbra, Coimbra, Portugal
| | - Joana Santos
- Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Coimbra, Coimbra, Portugal
| | - Jonathan P McNulty
- Radiography and Diagnostic Imaging, School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|
22
|
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: 79] [Impact Index Per Article: 39.5] [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
|
23
|
Kayal G, Chauvin M, Mora-Ramirez E, Clayton N, Vergara-Gil A, Tran-Gia J, Lassmann M, Calvert N, Tipping J, Struelens L, Bardiès M. Modelling SPECT auto-contouring acquisitions for 177Lu & 131I molecular radiotherapy using new developments in Geant4/GATE. Phys Med 2022; 96:101-113. [PMID: 35276403 DOI: 10.1016/j.ejmp.2022.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 02/22/2022] [Accepted: 03/04/2022] [Indexed: 11/17/2022] Open
Abstract
PURPOSE Monte Carlo modelling of SPECT imaging in Molecular Radiotherapy can improve activity quantification. Until now, SPECT modelling with GATE only considered circular orbit (CO) acquisitions. This cannot reproduce auto-contour acquisitions, where the detector head moves close to the patient to improve image resolution. The aim of this work is to develop and validate an auto-contouring step-and-shoot acquisition mode for GATE SPECT modelling. METHODS 177Lu and 131I SPECT experimental acquisitions performed on a Siemens Symbia T2 and GE Discovery 670 gamma camera, respectively, were modelled. SPECT projections were obtained for a cylindrical Jaszczak phantom and a lung and spine phantom. Detector head parameters (radial positions and acquisition angles) were extracted from the experimental projections to model the non-circular orbit (NCO) detector motion. The gamma camera model was validated against the experimental projections obtained with the cylindrical Jaszczak (177Lu) and lung and spine phantom (131I). Then, 177Lu and 131I CO and NCO SPECT projections were simulated to validate the impact of explicit NCO modelling on simulated projections. RESULTS Experimental and simulated SPECT images were compared using the gamma index, and were in good agreement with gamma index passing rate (GIPR) and gammaavg of 96.27%, 0.242 (177Lu) and 92.89%, 0.36 (131I). Then, simulated 177Lu and 131I CO and NCO SPECT projections were compared. The GIPR, gammaavg between the two gamma camera motions was 99.85%, 0.108 for 177Lu and 75.58%, 0.6 for 131I. CONCLUSION This work thereby justifies the need for auto-contouring modelling for isotopes with high septal penetration.
Collapse
Affiliation(s)
- Gunjan Kayal
- CRCT, UMR 1037, INSERM, Université Toulouse III Paul Sabatier, Toulouse, France; SCK CEN, Belgian Nuclear Research Centre, Boeretang 200, Mol 2400, Belgium.
| | - Maxime Chauvin
- CRCT, UMR 1037, INSERM, Université Toulouse III Paul Sabatier, Toulouse, France
| | - Erick Mora-Ramirez
- CRCT, UMR 1037, INSERM, Université Toulouse III Paul Sabatier, Toulouse, France; Universidad de Costa Rica, Escuela de Fisica, CICANUM, San Jose, Costa Rica
| | - Naomi Clayton
- CRCT, UMR 1037, INSERM, Université Toulouse III Paul Sabatier, Toulouse, France
| | - Alex Vergara-Gil
- CRCT, UMR 1037, INSERM, Université Toulouse III Paul Sabatier, Toulouse, France
| | - Johannes Tran-Gia
- Department of Nuclear Medicine, University of Würzburg, Würzburg, Germany
| | - Michael Lassmann
- Department of Nuclear Medicine, University of Würzburg, Würzburg, Germany
| | - Nicholas Calvert
- Christie Medical Physics and Engineering (CMPE), The Christie NHS Foundation Trust, Manchester, UK
| | - Jill Tipping
- Christie Medical Physics and Engineering (CMPE), The Christie NHS Foundation Trust, Manchester, UK
| | - Lara Struelens
- SCK CEN, Belgian Nuclear Research Centre, Boeretang 200, Mol 2400, Belgium
| | | | - Manuel Bardiès
- ICM, Département de Médecine Nucléaire, Montpellier, France; IRCM, UMR 1194 INSERM, Université de Montpellier and ICM, Montpellier, France
| |
Collapse
|
24
|
Ambrosini V, Zanoni L, Filice A, Lamberti G, Argalia G, Fortunati E, Campana D, Versari A, Fanti S. Radiolabeled Somatostatin Analogues for Diagnosis and Treatment of Neuroendocrine Tumors. Cancers (Basel) 2022; 14:1055. [PMID: 35205805 PMCID: PMC8870358 DOI: 10.3390/cancers14041055] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/10/2022] [Accepted: 02/17/2022] [Indexed: 02/04/2023] Open
Abstract
Neuroendocrine neoplasms (NENs) are rare and heterogeneous tumors that require multidisciplinary discussion for optimal care. The theranostic approach (DOTA peptides labelled with 68Ga for diagnosis and with 90Y or 177Lu for therapy) plays a crucial role in the management of NENs to assess disease extension and as a criteria for peptide receptor radionuclide therapy (PRRT) eligibility based on somatostatin receptor (SSTR) expression. On the diagnostic side, [68Ga]Ga-DOTA peptides PET/CT (SSTR PET/CT) is the gold standard for imaging well-differentiated SSTR-expressing neuroendocrine tumors (NETs). [18F]FDG PET/CT is useful in higher grade NENs (NET G2 with Ki-67 > 10% and NET G3; NEC) for more accurate disease characterization and prognostication. Promising emerging radiopharmaceuticals include somatostatin analogues labelled with 18F (to overcome the limits imposed by 68Ga), and SSTR antagonists (for both diagnosis and therapy). On the therapeutic side, the evidence gathered over the past two decades indicates that PRRT is to be considered as an effective and safe treatment option for SSTR-expressing NETs, and is currently included in the therapeutic algorithms of the main scientific societies. The positioning of PRRT in the treatment sequence, as well as treatment personalization (e.g., tailored dosimetry, re-treatment, selection criteria, and combination with other alternative treatment options), is warranted in order to improve its efficacy while reducing toxicity. Although very preliminary (being mostly hampered by lack of methodological standardization, especially regarding feature selection/extraction) and often including small patient cohorts, radiomic studies in NETs are also presented. To date, the implementation of radiomics in clinical practice is still unclear. The purpose of this review is to offer an overview of radiolabeled SSTR analogues for theranostic use in NENs.
Collapse
Affiliation(s)
- Valentina Ambrosini
- Department of Experimental Diagnostic and Specialized Medicine, University of Bologna, 40138 Bologna, Italy; (V.A.); (G.L.); (G.A.); (E.F.); (D.C.); (S.F.)
- Nuclear Medicine Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Lucia Zanoni
- Nuclear Medicine Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Angelina Filice
- Nuclear Medicine Unit, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy; (A.F.); (A.V.)
| | - Giuseppe Lamberti
- Department of Experimental Diagnostic and Specialized Medicine, University of Bologna, 40138 Bologna, Italy; (V.A.); (G.L.); (G.A.); (E.F.); (D.C.); (S.F.)
- Division of Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Giulia Argalia
- Department of Experimental Diagnostic and Specialized Medicine, University of Bologna, 40138 Bologna, Italy; (V.A.); (G.L.); (G.A.); (E.F.); (D.C.); (S.F.)
| | - Emilia Fortunati
- Department of Experimental Diagnostic and Specialized Medicine, University of Bologna, 40138 Bologna, Italy; (V.A.); (G.L.); (G.A.); (E.F.); (D.C.); (S.F.)
| | - Davide Campana
- Department of Experimental Diagnostic and Specialized Medicine, University of Bologna, 40138 Bologna, Italy; (V.A.); (G.L.); (G.A.); (E.F.); (D.C.); (S.F.)
- Division of Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Annibale Versari
- Nuclear Medicine Unit, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy; (A.F.); (A.V.)
| | - Stefano Fanti
- Department of Experimental Diagnostic and Specialized Medicine, University of Bologna, 40138 Bologna, Italy; (V.A.); (G.L.); (G.A.); (E.F.); (D.C.); (S.F.)
- Nuclear Medicine Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| |
Collapse
|
25
|
Graves SA, Martin M, Tiwari A, Merrick MJ, Sunderland JJ. SIR-Spheres activity measurements reveal systematic miscalibration. J Nucl Med 2022; 63:1131-1135. [PMID: 34992155 DOI: 10.2967/jnumed.121.262650] [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: 05/26/2021] [Accepted: 11/16/2021] [Indexed: 11/16/2022] Open
Abstract
Purpose: Accurate dosimetry-guided radiopharmaceutical therapy fundamentally relies on knowledge of the quantity of radioactivity administered to patients. The purpose of this work was to perform an independent and NIST-traceable activity measurement of 90Y SIR-Spheres®. Methods: Gamma spectroscopic measurements of the 90Y internal pair production decay mode were made using a high-purity germanium detector. Un-modified patient SIR-Spheres® vials were placed within a high-density polyethylene source holder positioned at a distance of 210 cm from the detector, with acquisition durations of 3 - 6 hours. Measured annihilation radiation detection rates were corrected for radioactive decay during acquisition, dead time, source attenuation, and source geometry effects. Detection efficiency was determined by two independent and NIST-traceable methods. Resulting 90Y activity measurements were compared against the manufacturer activity calibration. Results: Measured SIR-Sphere® vials (n = 5) were found to contain more activity than specified by the manufacturer calibration - on average the ratio of measured activity to calibrated was 1.233 ± 0.030. Activity measurements made using two distinct efficiency calibration methods were found to agree within 1%. Uncertainty in individual measurements was dominated by counting statistical uncertainty (~2.5%), uncertainty in the internal pair production branching ratio of 90Y (1.5%), and efficiency calibration (1.2% - 1.9%). Conclusion: The primary SIR-Spheres® activity calibration appears to be a significant underestimate of true activity. This mis-calibration has likely been consistent for as long as the SIR-Sphere® product has been available. This finding should be independently verified, and steps should be taken by the manufacturer to establish an accurate and traceable activity standard.
Collapse
|
26
|
Flux G, Leek F, Gape P, Gear J, Taprogge J. Iodine-131 and Iodine-131-Meta-iodobenzylguanidine Dosimetry in Cancer Therapy. Semin Nucl Med 2021; 52:167-177. [PMID: 34961618 DOI: 10.1053/j.semnuclmed.2021.11.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Radioactive iodine was first used for the treatment of benign thyroid disease and thyroid cancer 80 years ago. I-131 mIBG was later developed for the treatment of adult and pediatric neuroendocrine tumors. Physicists were closely involved from the outset to measure retention, to quantify uptake and to calculate radiation dosimetry. As the treatment became widespread, contrasting treatment regimes were followed, either given with empirically derived fixed levels of activity or guided according to the radiation doses delivered. As for external beam radiotherapy, individualized treatments for both thyroid cancer and neuroendocrine tumors were developed based on the aim of maximizing the radiation doses delivered to target volumes while restricting the radiation doses delivered to organs-at-risk, particularly the bone marrow. The challenge of marrow dosimetry has been met by using surrogate measures, often the blood dose for thyroid treatments and the whole-body dose in the case of treatment of neuroblastoma with I-131 mIBG. A number of studies have sought to establish threshold absorbed doses to ensure therapeutic efficacy. Although different values have been postulated, it has nevertheless been conclusively demonstrated that a fixed activity approach leads to a wide range of absorbed doses delivered to target volumes and to normal organs. Personalized treatment planning is now technically feasible with ongoing multicenter clinical trials and investigations into image quantification, biokinetic modelling and radiobiology.
Collapse
Affiliation(s)
- Glenn Flux
- Department of Physics, Royal Marsden Hospital & Institute of Cancer Research, Sutton, UK.
| | - Francesca Leek
- Department of Physics, Royal Marsden Hospital & Institute of Cancer Research, Sutton, UK
| | - Paul Gape
- Department of Physics, Royal Marsden Hospital & Institute of Cancer Research, Sutton, UK
| | - Jonathan Gear
- Department of Physics, Royal Marsden Hospital & Institute of Cancer Research, Sutton, UK
| | - Jan Taprogge
- Department of Physics, Royal Marsden Hospital & Institute of Cancer Research, Sutton, UK
| |
Collapse
|
27
|
Marengo M, Martin CJ, Rubow S, Sera T, Amador Z, Torres L. Radiation Safety and Accidental Radiation Exposures in Nuclear Medicine. Semin Nucl Med 2021; 52:94-113. [PMID: 34916044 DOI: 10.1053/j.semnuclmed.2021.11.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Medical radiation accidents and unintended events may lead to accidental or unintended medical exposure of patients and exposure of staff or the public. Most unintended exposures in nuclear medicine will lead to a small increase in risk; nevertheless, these require investigation and a clinical and dosimetric assessment. Nuclear medicine staff are exposed to radiation emitted directly by radiopharmaceuticals and by patients after administration of radiopharmaceuticals. This is particularly relevant in PET, due to the penetrating 511 keV γ-rays. Dose constraints should be set for planning the exposure of individuals. Staff body doses of 1-25 µSv/GBq are reported for PET imaging, the largest component being from the injection. The preparation and administration of radiopharmaceuticals can lead to high doses to the hands, challenging dose limits for radionuclides such as 90Y and even 18F. The risks of contamination can be minimized by basic precautions, such as carrying out manipulations in purpose-built facilities, wearing protective clothing, especially gloves, and removing contaminated gloves or any skin contamination as quickly as possible. Airborne contamination is a potential problem when handling radioisotopes of iodine or administering radioaerosols. Manipulating radiopharmaceuticals in laminar air flow cabinets, and appropriate premises ventilation are necessary to improve safety levels. Ensuring patient safety and minimizing the risk of incidents require efficient overall quality management. Critical aspects include: the booking process, particularly if qualified medical supervision is not present; administration of radiopharmaceuticals to patients, with the risk of misadministration or extravasation; management of patients' data and images by information technology systems, considering the possibility of misalignment between patient personal data and clinical information. Prevention of possible mistakes in patient identification or in the management of patients with similar names requires particular attention. Appropriate management of pregnant or breast-feeding patients is another important aspect of radiation safety. In radiopharmacy activities, strict quality assurance should be implemented at all operational levels, in addition to adherence to national and international regulations and guidelines. This includes not only administrative aspects, like checking the request/prescription, patient's data and the details of the requested procedure, but also quantitative tests according to national/international pharmacopoeias, and measuring the dispensed activity with a calibrated activity meter prior to administration. In therapy with radionuclides, skin tissue reactions can occur following extravasation, which can result in localized doses of tens of Grays. Other relevant incidents include confusion of products for patients administered at the same time or malfunction of administration devices. Furthermore, errors in internal radiation dosimetry calculations for treatment planning may lead to under or over-treatment. According to literature, proper instructions are fundamental to keep effective dose to caregivers and family members after patient discharge below the Dose constraints. The IAEA Basic Safety Standards require measures to minimize the likelihood of any unintended or accidental medical exposures and reporting any radiation incident. The relative complexity of nuclear medicine practice presents many possibilities for errors. It is therefore important that all activities are performed according to well established procedures, and that all actions are supported by regular quality assurance/QC procedures.
Collapse
Affiliation(s)
- Mario Marengo
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Italy.
| | - Colin J Martin
- Department of Clinical Physics and Bioengineering, University of Glasgow, UK
| | - Sietske Rubow
- Nuclear Medicine Division, Stellenbosch University, Stellenbosch, South Africa
| | - Terez Sera
- Department of Nuclear Medicine, University of Szeged, Szeged, Hungary
| | - Zayda Amador
- Radiation Protection Department, Centre of Isotopes, Havana, Cuba
| | - Leonel Torres
- Nuclear Medicine Department, Centre of Isotopes, Havana, Cuba
| |
Collapse
|
28
|
Cicone F, Gnesin S, Cremonesi M. Dosimetry of nuclear medicine therapies: current controversies and impact on treatment optimization. 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; 65:327-332. [PMID: 34881850 DOI: 10.23736/s1824-4785.21.03418-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Nuclear medicine therapeutic procedures have considerably expanded over the last few years, and their number is expected to grow exponentially in the future. Internal dosimetry has significantly developed as well, but has not yet been uniformly accepted as a valuable tool for prediction of therapeutic efficacy and toxicity. In this paper, we briefly summarize some of the arguments about the implementation of internal dosimetry in clinical practice. In addition, we provide a few examples of radionuclide anticancer therapies for which internal dosimetry demonstrated a significant impact on treatment optimization and patient outcome.
Collapse
Affiliation(s)
- Francesco Cicone
- PET/RM Unit, Department of Experimental and Clinical Medicine, and Neuroscience Research Center, Magna Graecia University of Catanzaro, Catanzaro, Italy - .,Unit of Nuclear Medicine, Mater Domini University Hospital, Catanzaro, Italy - .,University of Lausanne, Lausanne, Switzerland -
| | - Silvano Gnesin
- Institute of Radiation Physics, Lausanne University Hospital, Lausanne, Switzerland
| | - Marta Cremonesi
- Unit of Radiation Research, Department of Medical Imaging and Radiation Sciences, IRCCS European Institute of Oncology, Milan, Italy
| |
Collapse
|
29
|
Ligonnet T, Pistone D, Auditore L, Italiano A, Amato E, Campennì A, Schaefer N, Boughdad S, Baldari S, Gnesin S. Simplified patient-specific renal dosimetry in 177Lu therapy: a proof of concept. Phys Med 2021; 92:75-85. [PMID: 34875425 DOI: 10.1016/j.ejmp.2021.11.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 11/17/2021] [Accepted: 11/20/2021] [Indexed: 12/25/2022] Open
Abstract
PURPOSE The aim of this proof-of-concept study is to propose a simplified personalized kidney dosimetry procedure in 177Lu peptide receptor radionuclide therapy (PRRT) for neuroendocrine tumors and metastatic prostate cancer. It relies on a single quantitative SPECT/CT acquisition and multiple radiometric measurements executed with a collimated external probe, properly directed on kidneys. METHODS We conducted a phantom study involving external count-rate measurements in an abdominal phantom setup filled with activity concentrations of 99mTc, reproducing patient-relevant organ effective half-lives occurring in 177Lu PRRT. GATE Monte Carlo (MC) simulations of the experiment, using 99mTc and 177Lu as sources, were performed. Furthermore, we tested this method via MC on a clinical case of 177Lu-DOTATATE PRRT with SPECT/CT images at three time points (2, 20 and 70 hrs), comparing a simplified kidney dosimetry, employing a single SPECT/CT and probe measurements at three time points, with the complete MC dosimetry. RESULTS The experimentally estimated kidney half-life with background subtraction applied was compatible within 3% with the expected value. The MC simulations of the phantom study, both with 99mTc and 177Lu, confirmed a similar level of accuracy. Concerning the clinical case, the simplified dosimetric method led to a kidney dose estimation compatible with the complete MC dosimetry within 6%, 12% and 2%, using respectively the SPECT/CT at 2, 20 and 70 hrs. CONCLUSIONS The proposed simplified procedure provided a satisfactory accuracy and would reduce the imaging required to derive the kidney absorbed dose to a unique quantitative SPECT/CT, with consequent benefits in terms of clinic workflows and patient comfort.
Collapse
Affiliation(s)
- Thomas Ligonnet
- Institute of Radiation Physics, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Daniele Pistone
- MIFT Department, Università degli Studi di Messina, Messina, Italy; INFN Sezione di Catania, Catania, Italy.
| | - Lucrezia Auditore
- BIOMORF Department, Università degli Studi di Messina, Messina, Italy
| | - Antonio Italiano
- MIFT Department, Università degli Studi di Messina, Messina, Italy; INFN Sezione di Catania, Catania, Italy
| | - Ernesto Amato
- INFN Sezione di Catania, Catania, Italy; BIOMORF Department, Università degli Studi di Messina, Messina, Italy
| | - Alfredo Campennì
- BIOMORF Department, Università degli Studi di Messina, Messina, Italy; Nuclear Medicine Unit, University Hospital "G. Martino", Messina, Italy
| | - Niklaus Schaefer
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Sarah Boughdad
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Sergio Baldari
- BIOMORF Department, Università degli Studi di Messina, Messina, Italy; Nuclear Medicine Unit, University Hospital "G. Martino", Messina, Italy
| | - Silvano Gnesin
- Institute of Radiation Physics, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
30
|
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: 1.0] [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
|
31
|
Pandit-Taskar N, Iravani A, Lee D, Jacene H, Pryma D, Hope T, Saboury B, Capala J, Wahl RL. Dosimetry in Clinical Radiopharmaceutical Therapy of Cancer: Practicality Versus Perfection in Current Practice. J Nucl Med 2021; 62:60S-72S. [PMID: 34857623 DOI: 10.2967/jnumed.121.262977] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 10/22/2021] [Indexed: 12/25/2022] Open
Abstract
The use of radiopharmaceutical therapies (RPTs) in the treatment of cancers is growing rapidly, with more agents becoming available for clinical use in last few years and many new RPTs being in development. Dosimetry assessment is critical for personalized RPT, insofar as administered activity should be assessed and optimized in order to maximize tumor-absorbed dose while keeping normal organs within defined safe dosages. However, many current clinical RPTs do not require patient-specific dosimetry based on current Food and Drug Administration-labeled approvals, and overall, dosimetry for RPT in clinical practice and trials is highly varied and underutilized. Several factors impede rigorous use of dosimetry, as compared with the more convenient and less resource-intensive practice of empiric dosing. We review various approaches to applying dosimetry for the assessment of activity in RPT and key clinical trials, the extent of dosimetry use, the relative pros and cons of dosimetry-based versus fixed activity, and practical limiting factors pertaining to current clinical practice.
Collapse
Affiliation(s)
| | - Amir Iravani
- Washington University School of Medicine, St. Louis, Missouri
| | - Dan Lee
- Ochsner Medical Center, New Orleans, Louisiana
| | | | - Dan Pryma
- Penn Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Thomas Hope
- University of San Francisco, San Francisco, California; and
| | | | - Jacek Capala
- National Institutes of Health, Bethesda, Maryland
| | - Richard L Wahl
- Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
32
|
Chiesa C, Strigari L, Pacilio M, Richetta E, Cannatà V, Stasi M, Marzola MC, Schillaci O, Bagni O, Maccauro M. Dosimetric optimization of nuclear medicine therapy based on the Council Directive 2013/59/EURATOM and the Italian law N. 101/2020. Position paper and recommendations by the Italian National Associations of Medical Physics (AIFM) and Nuclear Medicine (AIMN). Phys Med 2021; 89:317-326. [PMID: 34583307 DOI: 10.1016/j.ejmp.2021.07.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 06/27/2021] [Accepted: 07/01/2021] [Indexed: 10/20/2022] Open
Abstract
This recommendation by the Italian Associations of Nuclear Medicine (AIMN) and Medical Physics (AIFM) focuses on the dosimetric optimization of Nuclear Medicine Therapy (NMT) as clearly requested by the article 56 of the EURATOM Directive 2013/59 and its consequent implementation in article 158 in the Italian Law n. 101/2020. However, this statement must deal with scientific and methodological limits that still exist and, above all, with the currently available limited resources. This paper addresses these specific issues. It distinguishes among many possible kinds of NMT. For each type, dosimetric optimization is recommended or considered optional, according to the general criteria adopted in any human choice, i.e. a check of technical feasibility first, followed by a cost/benefit argument. The classification of therapies as standardized or non-standardized is presented. This is based on the complexity of the type of pathology, on the variability of the treatment outcome, and on the risks involved. According to the present document, which was officially delivered to Italian Health Ministry as necessary interpretation of the law, a therapeutic team can, in science and consciousness, overcome the indications of posology, to optimize and tailoring a treatment with dosimetry, on the basis of published national or international data or guidelines, without need of an Ethics Committee approval. Data collected in this way will provide additional evidence about optimal dosimetric reference values. As conclusion, a formal appeal is made to the European and National regulatory agencies for pharmaceuticals to obtain the official acknowledgment of this principle.
Collapse
Affiliation(s)
- Carlo Chiesa
- Nuclear Medicine, Foundation IRCCS Istituto Nazionale Tumori, Milan, Italy.
| | - Lidia Strigari
- Director of Medical Physics, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Massimiliano Pacilio
- Director of Medical Physics, Azienda Ospedaliero-Universitaria Policlinico Umberto I, Rome, Italy
| | - Elisa Richetta
- Medical Physics, Azienda Ospedaliera Ordine Mauriziano, Turin, Italy
| | - Vittorio Cannatà
- Director of Medical Physics Unit, Medical Physics Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Michele Stasi
- Medical Physics, Azienda Ospedaliera Ordine Mauriziano, Turin, Italy
| | - Maria Cristina Marzola
- Department of Nuclear Medicine PET/CT Centre, S. Maria della Misericordia Hospital, Rovigo, Italy
| | - Orazio Schillaci
- Dean of University Tor Vergata, Rome, Italy; IRCCS Neuromed, Pozzilli, Italy
| | - Oreste Bagni
- Director of Nuclear Medicine, S. Maria Goretti Hospital, Latina, Italy
| | - Marco Maccauro
- Nuclear Medicine, Foundation IRCCS Istituto Nazionale Tumori, Milan, Italy
| |
Collapse
|
33
|
Schreuder N, de Romijn I, Jager PL, Kosterink JGW, van Puijenbroek EP. Safe use of radiopharmaceuticals in patients with chronic kidney disease: a systematic review. EJNMMI Radiopharm Chem 2021; 6:27. [PMID: 34417933 PMCID: PMC8380202 DOI: 10.1186/s41181-021-00145-w] [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: 06/22/2021] [Accepted: 08/09/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Patients with chronic kidney disease (CKD) may need to have their radiopharmaceutical dosage adjusted to prevent adverse effects and poor outcomes, but there are few recommendations on radiopharmaceutical dosing for this group of patients. The aim of this study is to provide an overview of the available information on radiopharmaceutical dose recommendations for patients with CKD. METHODS We performed a systematic literature review according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement. We conducted a literature search in the MEDLINE (PubMed) and Embase databases and screened potentially relevant studies using inclusion and exclusion criteria. We independently assessed the included observational studies' methodologies and extracted relevant data. RESULTS Of the 5795 studies first identified, 34 were included in this systematic review. These studies described three radiopharmaceuticals: [131I]sodium iodine, [18F]fludeoxyglucose, and [131I]iobenguane. Twenty-nine studies (85.3%) reported data on patients with CKD stage 5, while only three studies mentioned CKD patients in other stages (8.8%). CONCLUSION We found no consistent recommendations for radiopharmaceutical dosing in patients with CKD. Although some studies do mention dosing difficulties in patients with CKD, information is available for only a few radiopharmaceuticals, and recommendations are sometimes contradictory. Further research on radiopharmaceutical dosing in patients with CKD is needed to determine whether these patients require specific dosing, especially for therapeutic radiopharmaceuticals where a non-optimised dose may lead to an increased risk of toxicity for non-targeted organs. Including patients with CKD in studies and providing specific information about dosing in these patients should be a priority for the radiopharmaceutical community.
Collapse
Affiliation(s)
- Nanno Schreuder
- Groningen Research Institute of Pharmacy, Unit of PharmacoTherapy, Epidemiology and Economics, University of Groningen, Groningen, The Netherlands.
- GE Healthcare Radiopharmacy Zwolle, Zwolle, The Netherlands.
| | - Iris de Romijn
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Pieter L Jager
- Department of Nuclear Medicine, Isala Hospital, Zwolle, The Netherlands
| | - Jos G W Kosterink
- Groningen Research Institute of Pharmacy, Unit of PharmacoTherapy, Epidemiology and Economics, University of Groningen, Groningen, The Netherlands
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Eugène P van Puijenbroek
- Groningen Research Institute of Pharmacy, Unit of PharmacoTherapy, Epidemiology and Economics, University of Groningen, Groningen, The Netherlands
- Netherlands Pharmacovigilance Centre Lareb, 's-Hertogenbosch, The Netherlands
| |
Collapse
|
34
|
Kristiansson A, Örbom A, Vilhelmsson Timmermand O, Ahlstedt J, Strand SE, Åkerström B. Kidney Protection with the Radical Scavenger α 1-Microglobulin (A1M) during Peptide Receptor Radionuclide and Radioligand Therapy. Antioxidants (Basel) 2021; 10:antiox10081271. [PMID: 34439519 PMCID: PMC8389303 DOI: 10.3390/antiox10081271] [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: 06/15/2021] [Revised: 08/05/2021] [Accepted: 08/07/2021] [Indexed: 02/07/2023] Open
Abstract
α1-Microglobulin (A1M) is an antioxidant found in all vertebrates, including humans. It has enzymatic reductase activity and can scavenge radicals and bind free heme groups. Infused recombinant A1M accumulates in the kidneys and has therefore been successful in protecting kidney injuries in different animal models. In this review, we focus on A1M as a radioprotector of the kidneys during peptide receptor radionuclide/radioligand therapy (PRRT/RLT). Patients with, e.g., neuroendocrine tumors or castration resistant prostate cancer can be treated by administration of radiolabeled small molecules which target and therefore enable the irradiation and killing of cancer cells through specific receptor interaction. The treatment is not curative, and kidney toxicity has been reported as a side effect since the small, radiolabeled substances are retained and excreted through the kidneys. In recent studies, A1M was shown to have radioprotective effects on cell cultures as well as having a similar biodistribution as the somatostatin analogue peptide 177Lu-DOTATATE after intravenous infusion in mice. Therefore, several animal studies were conducted to investigate the in vivo radioprotective potential of A1M towards kidneys. The results of these studies demonstrated that A1M co-infusion yielded protection against kidney toxicity and improved overall survival in mouse models. Moreover, two different mouse studies reported that A1M did not interfere with tumor treatment itself. Here, we give an overview of radionuclide therapy, the A1M physiology and the results from the radioprotector studies of the protein.
Collapse
Affiliation(s)
- Amanda Kristiansson
- Department of Clinical Sciences Lund, Oncology, Lund University, 221 00 Lund, Sweden; (A.Ö.); (O.V.T.); (S.-E.S.)
- Correspondence:
| | - Anders Örbom
- Department of Clinical Sciences Lund, Oncology, Lund University, 221 00 Lund, Sweden; (A.Ö.); (O.V.T.); (S.-E.S.)
| | - Oskar Vilhelmsson Timmermand
- Department of Clinical Sciences Lund, Oncology, Lund University, 221 00 Lund, Sweden; (A.Ö.); (O.V.T.); (S.-E.S.)
| | - Jonas Ahlstedt
- Department of Clinical Sciences Lund, CIPA, Lund University, 221 84 Lund, Sweden;
| | - Sven-Erik Strand
- Department of Clinical Sciences Lund, Oncology, Lund University, 221 00 Lund, Sweden; (A.Ö.); (O.V.T.); (S.-E.S.)
- Department of Clinical Sciences Lund, Medical Radiation Physics, Lund University, 221 00 Lund, Sweden
| | - Bo Åkerström
- Department of Clinical Sciences Lund, Section for Infection Medicine, Lund University, 221 84 Lund, Sweden;
| |
Collapse
|
35
|
Pre-Therapeutic Measurements of Iodine Avidity in Papillary and Poorly Differentiated Thyroid Cancer Reveal Associations with Thyroglobulin Expression, Histological Variants and Ki-67 Index. Cancers (Basel) 2021; 13:cancers13143627. [PMID: 34298840 PMCID: PMC8307105 DOI: 10.3390/cancers13143627] [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: 06/17/2021] [Revised: 07/16/2021] [Accepted: 07/17/2021] [Indexed: 12/29/2022] Open
Abstract
Papillary thyroid cancer (PTC) and poorly differentiated thyroid cancer (PDTC) are treated with radioiodine to reduce recurrence and to treat the spread of disease. Adequate iodine accumulation in cancer tissue, iodine avidity, is important for treatment effect. This study investigated which clinical and histological tumour characteristics correlate with avidity. To quantify avidity in cancer tissue, tracer amounts of iodine-131 were given to 45 patients with cytologically confirmed thyroid cancer. At pathology grossing, representative samples of tumour and lymph nodes were taken and subjected to radioactivity quantification ex vivo to determine avidity. Afterwards, samples underwent extended pathology work-up and analysis. We found that tumoural Tg expression and Ki-67 index were correlated with avidity, whereas tumour size and pT stage were not. The histological variant of thyroid cancer was also correlated with iodine avidity. Variants associated with worse clinical prognoses displayed lower avidity than variants with better prognoses. This work provides new information on which tumours have low iodine avidity. Lower avidity in aggressive histological PTC variants may explain their overall poorer prognoses. Our findings also suggest that radioiodine dosage could be adapted to Tg expression, Ki-67 index or histological variant instead of pT stage, potentially improving the efficacy of radioiodine therapy.
Collapse
|
36
|
Population exposure-response model of 131I in patients with benign thyroid disease. Eur J Pharm Sci 2021; 165:105942. [PMID: 34273482 DOI: 10.1016/j.ejps.2021.105942] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 06/01/2021] [Accepted: 06/20/2021] [Indexed: 11/22/2022]
Abstract
PURPOSE The study aimed to explore the relationship of different exposure measures with 131I therapy response in patients with benign thyroid disease, estimate the variability in the response, investigate possible covariates, and discuss dosing implications of the results. METHODS A population exposure-response analysis was performed using nonlinear mixed-effects modelling. Data from 95 adult patients with benign thyroid disease were analysed. Evaluated exposure parameters were: administered radioactivity dose (Aa) [MBq], total absorbed dose (ABD) [Gy], maximum of absorbed dose-rate (MXR) [Gy/h] and biologically effective dose (BED) [Gy]. The response was modelled as ordered categorical data: hyper-, eu- and hypothyroidism. The final model performance was evaluated by a visual predictive check. RESULTS The probability of the outcome following 131I therapy was best described by a proportional-odds model, including the log-linear model of 131I effect and the exponential model of the response-time relationship. All exposure measures were statistically significant with p<0.001, with BED and ABD being statistically better than the other two. Nevertheless, as BED resulted in the lowest AIC value, it was included in the final model. Accordingly, BED value of 289.7 Gy is associated with 80% probability of successful treatment outcome 12 months after 131I application in patients with median thyroid volume (32.28 mL). The target thyroid volume was a statistically significant covariate. The visual predictive check of the final model showed good model performance. CONCLUSION Our results imply that BED formalism could aid in therapy individualisation. The larger thyroid volume is associated with a lower probability of a successful outcome.
Collapse
|
37
|
Prospective SPECT-CT Organ Dosimetry-Driven Radiation-Absorbed Dose Escalation Using the In-111 ( 111In)/Yttrium 90 ( 90Y) Ibritumomab Tiuxetan (Zevalin ®) Theranostic Pair in Patients with Lymphoma at Myeloablative Dose Levels. Cancers (Basel) 2021; 13:cancers13112828. [PMID: 34204102 PMCID: PMC8201215 DOI: 10.3390/cancers13112828] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 05/17/2021] [Accepted: 05/18/2021] [Indexed: 12/25/2022] Open
Abstract
Simple Summary We prospectively evaluated the feasibility of SPECT-CT/planar organ dosimetry-based radiation dose escalation radioimmunotherapy in patients with recurrent non-Hodgkin’s lymphoma using the theranostic pair of 111In and 90Y anti-CD20 ibritumomab tiuxetan (Zevalin®) at myeloablative radiation-absorbed doses with autologous stem cell support. Unlike most routine dose escalation approaches, our approach used patient-individualized measurements of organ radiation absorbed dose from the tracer study, with patient-specific adjustments of the injected therapy dose to deliver a pre-specified radiation absorbed dose to the liver. Our approach was feasible, stem cell engraftment was swift, resulted in an 89% tumor response rate in treated patients, demonstrated over 3 fold variability in liver dosimetry/injected activity among patients, allowed us to exceed the FDA approved administered activity by over 5 fold and demonstrated the normal liver maximum tolerated dose to exceed 28 Gy. Dose escalation was not continued due to lack of drug availability. With modern dosimetry approaches, patient specific dosimetry-driven radiation dose escalation is feasible, allows adjustment of administered activity for heterogeneous pharmacokinetics and allows marked dose escalation vs. non-dosimetry driven approaches. Abstract Purpose: We prospectively evaluated the feasibility of SPECT-CT/planar organ dosimetry-based radiation dose escalation radioimmunotherapy in patients with recurrent non-Hodgkin’s lymphoma using the theranostic pair of 111In and 90Y anti-CD20 ibritumomab tiuxetan (Zevalin®) at myeloablative radiation-absorbed doses with autologous stem cell support. We also assessed acute non-hematopoietic toxicity and early tumor response in this two-center outpatient study. Methods: 24 patients with CD20-positive relapsed or refractory rituximab-sensitive, low-grade, mantle cell, or diffuse large-cell NHL, with normal organ function, platelet counts > 75,000/mm3, and <35% tumor involvement in the marrow were treated with Rituximab (375 mg/m2) weekly for 4 consecutive weeks, then one dose of cyclophosphamide 2.5 g/m2 with filgrastim 10 mcg/kg/day until stem cell collection. Of these, 18 patients with successful stem cell collection (at least 2 × 106 CD34 cells/kg) proceeded to RIT. A dosimetric administration of 111In ibritumomab tiuxetan (185 MBq) followed by five sequential quantitative planar and one SPECT/CT scan was used to determine predicted organ radiation-absorbed dose. Two weeks later, 90Y ibritumomab tiuxetan was administered in an outpatient setting at a cohort- and patient-specific predicted organ radiation-absorbed dose guided by a Continuous Response Assessment (CRM) methodology with the following cohorts for dose escalation: 14.8 MBq/kg, and targeted 18, 24, 28, and 30.5 Gy to the liver. Autologous stem cell infusion occurred when the estimated marrow radiation-absorbed dose rate was predicted to be <1 cGy/h. Feasibility, short-term toxicities, and tumor response were assessed. Results: Patient-specific hybrid SPECT/CT + planar organ dosimetry was feasible in all 18 cases and used to determine the patient-specific therapeutic dose and guide dose escalation (26.8 ± 7.3 MBq/kg (mean), 26.3 MBq/kg (median) of 90Y (range: 12.1–41.4 MBq/kg)) of ibritumomab tiuxetan that was required to deliver 10 Gy to the liver. Infused stem cells engrafted rapidly. The most common treatment-related toxicities were hematological and were reversible following stem cell infusion. No significant hepatotoxicity was seen. One patient died from probable treatment-related causes—pneumonia at day 27 post-transplant. One patient at dose level 18 Gy developed myelodysplastic syndrome (MDS), 4 patients required admission post-90Y RIT for febrile neutropenia, 16/18 patients receiving 90Y ibritumomab tiuxetan (89%) responded to the therapy, with 13 CR (72%) and 3/18 PR (17%), at 60 days post-treatment. Two patients had progressive disease at sixty days. One patient was lost to follow-up. Median time to progression was estimated to be at least 13 months. MTD to the liver is greater than 28 Gy, but the MTD was not reached as the study was terminated due to unexpected discontinuation of availability of the therapeutic agent. Conclusions: Patient-specific outpatient 90Y ibritumomab tiuxetan RIT with myeloablative doses of RIT up to a targeted 30.5 Gy to the liver is feasible, guided by prospective SPECT/CT + planar imaging with the theranostic pair of 111In and 90Y anti-CD20, with outpatient autologous stem cell transplant support. Administered activity over 5 times the standard FDA-approved activity was well-tolerated. The non-hematopoietic MTD in this study exceeds 28 Gy to the liver. Initial tumor responses were common at all dose levels. This study supports the feasibility of organ dosimetry-driven patient-specific dose escalation in the treatment of NHL with stem cell transplant and provides additional information on the radiation tolerance of the normal liver to radiopharmaceutical therapy.
Collapse
|
38
|
Monserrat Fuertes T, González García F, Peinado Montes M, Domínguez Grande M, Martín Fernández N, Gómez de Iturriaga Piña A, Mínguez Gabiña P. Description of the methodology for dosimetric quantification in treatments with 177Lu-DOTATATE. Rev Esp Med Nucl Imagen Mol 2021. [DOI: 10.1016/j.remnie.2021.03.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
39
|
Jena R, Narain TA, Singh UP, Srivastava A. Role of positron emission tomography/computed tomography in the evaluation of renal cell carcinoma. Indian J Urol 2021; 37:125-132. [PMID: 34103794 PMCID: PMC8173953 DOI: 10.4103/iju.iju_268_20] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 08/12/2020] [Accepted: 10/04/2020] [Indexed: 01/03/2023] Open
Abstract
Introduction: Positron emission tomography (PET) is not a standard recommendation in most of the major guidelines for the evaluation of renal cell carcinoma (RCC). Earlier studies evaluating PET scan in patients with RCC have provided discordant results. However, with the advent of newer hybrid PET/computed tomography (CT) scanning systems, this modality has shown increased efficacy in the evaluation of primary renal masses along with the detection of extrarenal metastases, restaging recurrent RCC, and also in monitoring response to targeted therapy. We performed a systematic review of the existing literature on the role of PET scan in the evaluation of RCC. Methodology: We systematically searched the databases of PubMed/Medline, Embase, and Google Scholar to identify studies on the use of PET scan in RCC. Using Preferred Reporting Items for Systematic Reviews and Meta-analysis guidelines, 94 full-text articles were selected, of which 54 relevant articles were then reviewed, after a consensus by the authors. Results: Several studies have shown similar sensitivity and specificity of fluoro-2-deoxy-2-d-glucose-PET (FDG-PET) scan as compared to conventional CT scan for the initial diagnosis of RCC, and an improved sensitivity and specificity for the detection of metastases and recurrences following curative therapy. The PET scan may also play a role in predicting the initial tumor biology and pathology and predicting the prognosis as well as the response to therapy. Conclusion: The current guidelines do not recommend PET scan in the staging armamentarium of RCCs. However, FDG-PET scan is as efficacious, if not better than conventional imaging alone, in the evaluation of the primary and metastatic RCC, as well as in evaluating the response to therapy, due to its ability to pick up areas of increased metabolic activity early on. Newer tracers such as Ga68 prostate specific membrane antigen-labeled ligands may help in opening up newer avenues of theragnostics.
Collapse
Affiliation(s)
- Rahul Jena
- Department of Urology and Renal Transplant, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Tushar Aditya Narain
- Department of Urology, All India Institute of Medical Sciences, Rishikesh, Uttarkhand, India
| | - Uday Pratap Singh
- Department of Urology and Renal Transplant, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Aneesh Srivastava
- Department of Urology and Renal Transplant, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| |
Collapse
|
40
|
Monserrat Fuertes T, González García FM, Peinado Montes MÁ, Domínguez Grande ML, Martín Fernández N, Gómez de Iturriaga Piña A, Mínguez Gabiña P. Description of the methodology for dosimetric quantification in treatments with 177Lu-DOTATATE. Rev Esp Med Nucl Imagen Mol 2021; 40:167-178. [PMID: 33811003 DOI: 10.1016/j.remn.2021.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/22/2021] [Accepted: 02/24/2021] [Indexed: 11/28/2022]
Abstract
Implementation of dosimetry calculations in the daily practice of Nuclear Medicine Departments is, at this time, a controversial issue, partly due to the lack of a standardized methodology that is accepted by all interested parties (patients, nuclear medicine physicians and medical physicists). However, since the publication of RD 601/2019 there is a legal obligation to implement it, despite the fact that it is a complex and high resource consumption procedure. The aim of this article is to review the theoretical bases of in vivo dosimetry in treatments with 177Lu-DOTATATE. The exposed methodology is the one proposed by the MIRD Committee (Medical Internal Radiation Dose) of the SNMMI (Society of Nuclear Medicine & Molecular Imaging). According to this method, the absorbed dose is obtained as the product of 2factors: the time-integrated activity of the radiopharmaceutical present in a source region and a geometrical factor S. This approach, which a priori seems simple, in practice requires several SPECT/CT acquisitions, several measurements of the whole body activity and taking several blood samples, as well as hours of image processing and computation. The systematic implementation of these calculations, in all the patients we treat, will allow us to obtain homogeneous data to correlate the absorbed doses in the lesions with the biological effect of the treatment. The final purpose of the dosimetry calculations is to be able to maximize the therapeutic effect in the lesions, controlling the radiotoxicity in the organs at risk.
Collapse
Affiliation(s)
- T Monserrat Fuertes
- Servicio de Radiofísica y Protección Radiológica, Hospital Universitario Central de Asturias, Oviedo, Asturias, España; Departamento de Cirugía, Radiología y Medicina Física, UPV/EHU, Leioa, Bizkaia, España.
| | - F M González García
- Servicio de Medicina Nuclear, Hospital Universitario Central de Asturias, Oviedo, Asturias, España
| | - M Á Peinado Montes
- Servicio de Radiofísica y Protección Radiológica, Hospital Universitario Central de Asturias, Oviedo, Asturias, España
| | - M L Domínguez Grande
- Servicio de Medicina Nuclear, Hospital Universitario Central de Asturias, Oviedo, Asturias, España
| | - N Martín Fernández
- Servicio de Medicina Nuclear, Hospital Universitario Central de Asturias, Oviedo, Asturias, España
| | - A Gómez de Iturriaga Piña
- Departamento de Cirugía, Radiología y Medicina Física, UPV/EHU, Leioa, Bizkaia, España; Servicio de Oncología Radioterápica, Hospital Universitario Gurutzeta-Cruces/Instituto de Investigación Sanitaria BioCruces, Barakaldo, Bizkaia, España
| | - P Mínguez Gabiña
- Unidad de Protección Radiológica y Radiofísica, Hospital Universitario Gurutzeta-Cruces/Instituto de Investigación Sanitaria BioCruces, Barakaldo, Bizkaia, España
| |
Collapse
|
41
|
Taprogge J, Wadsley J, Miles E, Flux GD. Recommendations for Multicentre Clinical Trials Involving Dosimetry for Molecular Radiotherapy. Clin Oncol (R Coll Radiol) 2021; 33:131-136. [PMID: 33342617 PMCID: PMC7818526 DOI: 10.1016/j.clon.2020.12.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/06/2020] [Accepted: 12/02/2020] [Indexed: 11/17/2022]
Abstract
Multicentre clinical trials involving a dosimetry component are becoming more prevalent in molecular radiotherapy and are essential to generate the evidence to support individualised approaches to treatment planning and to ensure that sufficient patients are recruited to achieve the statistical significance required. Quality assurance programmes should be considered to support the standardisation required to achieve meaningful results. Trials should be designed to ensure that dosimetry results from image acquisition systems across centres are comparable by incorporating steps to standardise the methodologies used for the quantification of images and dosimetry. Furthermore, it is essential to assess the expertise and resources available at each participating site prior to trial commencement. A quality assurance plan should be drawn up and training provided if necessary. Standardisation of quantification and dosimetry methodologies used in a trial are essential to ensure that results from different centres may be collated. In addition, appropriate uncertainty analysis should be carried out to correct for differences in methodologies between centres. Recommendations are provided to support dosimetry studies based on the experience of several previous and ongoing multicentre trials.
Collapse
Affiliation(s)
- J Taprogge
- National Radiotherapy Trials Quality Assurance (RTTQA) Group, Joint Department of Physics, Royal Marsden NHSFT, Sutton, UK; The Institute of Cancer Research, London, UK.
| | | | - E Miles
- National Radiotherapy Trials Quality Assurance (RTTQA) Group, Mount Vernon Cancer Centre, East and North Hertfordshire NHS Trust, Northwood, UK
| | - G D Flux
- The Institute of Cancer Research, London, UK; Joint Department of Physics, Royal Marsden NHSFT, Sutton, UK
| |
Collapse
|
42
|
Al-Jabri A, Cooke J, Cournane S, Healy ML. Gender differences in estimating I-131 thyroid uptake from Tc-99m thyroid uptake for benign thyroid disease. Br J Radiol 2021; 94:20200700. [PMID: 33242250 DOI: 10.1259/bjr.20200700] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
OBJECTIVE For radioactive Iodine-131 (131I) treatments of thyroid diseases, increased efficacy has been reported for personalized dosimetry treatments. The measurement of Iodine-131 thyroid uptake (131IU) is required in these cases. This study aims to investigate whether 99mTc thyroid uptake (99mTcU) may be used in place of 131IU for implementing personalised treatments. METHODS A retrospective study of 152 benign thyroid disease 131I treatments was carried out during 2012-2020; 117 treatments were for female patients while 35 were for male patients diagnosed with either Graves' disease, multinodular goitre or toxic nodules. RESULTS A statistically significant correlation was found between 131IU and 99mTcU data, with the data more correlated for male than female patients (r = 0.71 vs 0.38, p-value < 0.001). Patient age and time difference between the two respective uptake measurements significantly influenced the uptake correlation in females but not for the male cohort, although there was no significant difference between the parameters across gender. Thyroid diagnosis and hormone levels showed a significant correlation with uptakes in both genders. Estimating 131IU based on 99mTcU was shown to be predictive for male but not in female patients (R2 = 91% vs 16%). CONCLUSION Estimating 131IU based on 99mTcU is not recommended for females at our centre. Males reported good correlation, but a larger sample would be needed for validation. ADVANCES IN KNOWLEDGE The initial findings showed a significant gender difference in benign thyroid uptake parameters at our centre, highlighting the potential need for gender consideration when planning 131IU patient management and when reporting studies results.
Collapse
Affiliation(s)
- Amna Al-Jabri
- School of Medicine, Trinity College, Dublin, Ireland.,St James's Hospital, Dublin, Ireland
| | | | | | | |
Collapse
|
43
|
Konijnenberg M, Herrmann K, Kobe C, Verburg F, Hindorf C, Hustinx R, Lassmann M. EANM position paper on article 56 of the Council Directive 2013/59/Euratom (basic safety standards) for nuclear medicine therapy. Eur J Nucl Med Mol Imaging 2021; 48:67-72. [PMID: 33057773 PMCID: PMC7835146 DOI: 10.1007/s00259-020-05038-9] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 09/11/2020] [Indexed: 11/17/2022]
Abstract
The EC Directive 2013/59/Euratom states in article 56 that exposures of target volumes in nuclear medicine treatments shall be individually planned and their delivery appropriately verified. The Directive also mentions that medical physics experts should always be appropriately involved in those treatments. Although it is obvious that, in nuclear medicine practice, every nuclear medicine physician and physicist should follow national rules and legislation, the EANM considered it necessary to provide guidance on how to interpret the Directive statements for nuclear medicine treatments.For this purpose, the EANM proposes to distinguish three levels in compliance to the optimization principle in the directive, inspired by the indication of levels in prescribing, recording and reporting of absorbed doses after radiotherapy defined by the International Commission on Radiation Units and Measurements (ICRU): Most nuclear medicine treatments currently applied in Europe are standardized. The minimum requirement for those treatments is ICRU level 1 ("activity-based prescription and patient-averaged dosimetry"), which is defined by administering the activity within 10% of the intended activity, typically according to the package insert or to the respective EANM guidelines, followed by verification of the therapy delivery, if applicable. Non-standardized treatments are essentially those in developmental phase or approved radiopharmaceuticals being used off-label with significantly (> 25% more than in the label) higher activities. These treatments should comply with ICRU level 2 ("activity-based prescription and patient-specific dosimetry"), which implies recording and reporting of the absorbed dose to organs at risk and optionally the absorbed dose to treatment regions. The EANM strongly encourages to foster research that eventually leads to treatment planning according to ICRU level 3 ("dosimetry-guided patient-specific prescription and verification"), whenever possible and relevant. Evidence for superiority of therapy prescription on basis of patient-specific dosimetry has not been obtained. However, the authors believe that a better understanding of therapy dosimetry, i.e. how much and where the energy is delivered, and radiobiology, i.e. radiation-related processes in tissues, are keys to the long-term improvement of our treatments.
Collapse
Affiliation(s)
- Mark Konijnenberg
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, Netherlands
| | - Ken Herrmann
- Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
| | - Carsten Kobe
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Frederik Verburg
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, Netherlands
| | - Cecilia Hindorf
- Department of Nuclear Medicine Physics, Skåne University Hospital, Lund, Sweden
| | - Roland Hustinx
- Service de Médecine Nucléaire et d'Imagerie Oncologique, Département de Physique médicale, Université de Liège - GIGA-CRC in vivo imaging, Centre Hospitalier Universitaire de Liège, Liège, Belgium
| | - Michael Lassmann
- Department of Nuclear Medicine, Universitätsklinikum Würzburg, Klinik und Poliklinik für Nuklearmedizin, Oberdürrbacher Str. 6, 97080, Würzburg, Germany.
| |
Collapse
|
44
|
Brosch J, Gosewisch A, Kaiser L, Seidensticker M, Ricke J, Zellmer J, Bartenstein P, Ziegler S, Ilhan H, Todica A, Böning G. 3D image-based dosimetry for Yttrium-90 radioembolization of hepatocellular carcinoma: Impact of imaging method on absorbed dose estimates. Phys Med 2020; 80:317-326. [PMID: 33248338 DOI: 10.1016/j.ejmp.2020.11.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 11/09/2020] [Accepted: 11/11/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND To improve therapy outcome of Yttrium-90 selective internal radiation therapy (90Y SIRT), patient-specific post-therapeutic dosimetry is required. For this purpose, various dosimetric approaches based on different available imaging data have been reported. The aim of this work was to compare post-therapeutic 3D absorbed dose images using Technetium-99m (99mTc) MAA SPECT/CT, Yttrium-90 (90Y) bremsstrahlung (BRS) SPECT/CT, and 90Y PET/CT. METHODS Ten SIRTs of nine patients with unresectable hepatocellular carcinoma (HCC) were investigated. The 99mTc SPECT/CT data, obtained from 99mTc-MAA-based treatment simulation prior to 90Y SIRT, were scaled with the administered 90Y therapy activity. 3D absorbed dose images were generated by dose kernel convolution with scaled 99mTc/90Y SPECT/CT, 90Y BRS SPECT/CT, and 90Y PET/CT data of each patient. Absorbed dose estimates in tumor and healthy liver tissue obtained using the two SPECT/CT methods were compared against 90Y PET/CT. RESULTS The percentage deviation of tumor absorbed dose estimates from 90Y PET/CT values was on average -2 ± 18% for scaled 99mTc/90Y SPECT/CT, whereas estimates from 90Y BRS SPECT/CT differed on average by -50 ± 13%. For healthy liver absorbed dose estimates, all three imaging methods revealed comparable values. CONCLUSION The quantification capabilities of the imaging data influence 90Y SIRT tumor dosimetry, while healthy liver absorbed dose values were comparable for all investigated imaging data. When no 90Y PET/CT image data are available, the proposed scaled 99mTc/90Y SPECT/CT dosimetry method was found to be more appropriate for HCC tumor dosimetry than 90Y BRS SPECT/CT based dosimetry.
Collapse
Affiliation(s)
- Julia Brosch
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany.
| | - Astrid Gosewisch
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Lena Kaiser
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Max Seidensticker
- Department of Radiology, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Jens Ricke
- Department of Radiology, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Johannes Zellmer
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Sibylle Ziegler
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Harun Ilhan
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Andrei Todica
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Guido Böning
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| |
Collapse
|
45
|
Cicone F, Denoël T, Gnesin S, Riggi N, Irving M, Jakka G, Schaefer N, Viertl D, Coukos G, Prior JO. Preclinical Evaluation and Dosimetry of [ 111In]CHX-DTPA-scFv78-Fc Targeting Endosialin/Tumor Endothelial Marker 1 (TEM1). Mol Imaging Biol 2020; 22:979-991. [PMID: 31993928 PMCID: PMC7343747 DOI: 10.1007/s11307-020-01479-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE Endosialin/tumor endothelial marker-1 (TEM1) is an attractive theranostic target expressed by the microenvironment of a wide range of tumors, as well as by sarcoma and neuroblastoma cells. We report on the radiolabeling and preclinical evaluation of the scFv78-Fc, a fully human TEM1-targeting antibody fragment cross-reactive with mouse TEM1. PROCEDURES The scFv78-Fc was conjugated with the chelator p-SCN-Bn-CHX-A"-DTPA, followed by labeling with indium-111. The number of chelators per molecule was estimated by mass spectrometry. A conventional saturation assay, extrapolated to infinite antigen concentration, was used to determine the immunoreactive fraction of the radioimmunoconjugate. The radiopharmaceutical biodistribution was assessed in immunodeficient mice grafted with Ewing's sarcoma RD-ES and neuroblastoma SK-N-AS human TEM1-positive tumors. The full biodistribution studies were preceded by a dose-escalation experiment based on the simultaneous administration of the radiopharmaceutical with increasing amounts of unlabeled scFv78-Fc. Radiation dosimetry extrapolations to human adults were obtained from mouse biodistribution data according to established methodologies and additional assumptions concerning the impact of the tumor antigenic sink in the cross-species translation. RESULTS [111In]CHX-DTPA-scFv78-Fc was obtained with a radiochemical purity > 98 % after 1 h incubation at 42 °C and ultrafiltration. It showed good stability in human serum and > 70 % immunoreactive fraction. Biodistribution data acquired in tumor-bearing mice confirmed fast blood clearance and specific tumor targeting in both xenograft models. The radiopharmaceutical off-target uptake was predominantly abdominal. After a theoretical injection of [111In]CHX-DTPA-scFv78-Fc to the reference person, the organs receiving the highest absorbed dose would be the spleen (0.876 mGy/MBq), the liver (0.570 mGy/MBq) and the kidneys (0.298 mGy/MBq). The total body dose and the effective dose would be 0.058 mGy/MBq and 0.116 mSv/MBq, respectively. CONCLUSIONS [111In]CHX-DTPA-scFv78-Fc binds specifically to endosialin/TEM1 in vitro and in vivo. Dosimetry estimates are in the range of other monoclonal antibodies radiolabeled with indium-111. [111In]CHX-DTPA-scFv78-Fc could be potentially translated into clinic.
Collapse
Affiliation(s)
- Francesco Cicone
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland.
- Department of Experimental and Clinical Medicine, Unit of Nuclear Medicine, "Magna Graecia" University of Catanzaro, Catanzaro, Italy.
| | - Thibaut Denoël
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Silvano Gnesin
- Institute of Radiation Physics, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Nicolo Riggi
- Experimental Pathology Service, Institute of Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Melita Irving
- Department of Oncology, Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, CH-1066, Epalinges, Switzerland
| | - Gopinadh Jakka
- Department of Oncology, Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, CH-1066, Epalinges, Switzerland
| | - Niklaus Schaefer
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - David Viertl
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - George Coukos
- Department of Oncology, Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, CH-1066, Epalinges, Switzerland
| | - John O Prior
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
46
|
PRRT of neuroendocrine tumors: individualized dosimetry or fixed dose scheme? EJNMMI Res 2020; 10:35. [PMID: 32296955 PMCID: PMC7158965 DOI: 10.1186/s13550-020-00623-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 03/26/2020] [Indexed: 01/03/2023] Open
Abstract
Great efforts have been made in dosimetry for individualizing PRRT. However, many centers do not use dosimetry and its results hardly influence treatment. A reason for that is that reliable thresholds for organs-at-risk, kidneys and bone marrow, and treatment response are lacking. The nuclear medicine community must provide solid data from large trials delivering reliable thresholds, which then help to tailor PRRT according to organ doses (in order to reduce toxicity or increase treatment activity) or tumor doses (in order to increase activity to meet the response-threshold). Otherwise, development of radionuclide therapies will be done like big pharmaceutical companies do it currently: classical dose escalation studies and agreement on acceptable toxicity probabilities. Therapeutic radiopharmaceuticals will then be handled like other drugs, which on the other hand will increase availability of radionuclide therapies.
Collapse
|
47
|
Gear J, Chiesa C, Lassmann M, Gabiña PM, Tran-Gia J, Stokke C, Flux G. EANM Dosimetry Committee series on standard operational procedures for internal dosimetry for 131I mIBG treatment of neuroendocrine tumours. EJNMMI Phys 2020; 7:15. [PMID: 32144574 PMCID: PMC7060302 DOI: 10.1186/s40658-020-0282-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 02/13/2020] [Indexed: 11/23/2022] Open
Abstract
The purpose of the EANM Dosimetry Committee Series on "Standard Operational Procedures for Dosimetry" (SOP) is to provide advice to scientists and clinicians on how to perform patient-specific absorbed dose assessments. This SOP describes image and data acquisition parameters and dosimetry calculations to determine the absorbed doses delivered to whole-body, tumour and normal organs following a therapeutic administration of 131I mIBG for the treatment of neuroblastoma or adult neuroendocrine tumours. Recommendations are based on evidence in recent literature where available and on expert opinion within the community. This SOP is intended to promote standardisation of practice within the community and as such is based on the facilities and expertise that should be available to any centre able to perform specialised treatments with radiopharmaceuticals and patient-specific dosimetry. A clinical example is given to demonstrate the application of the absorbed dose calculations.
Collapse
Affiliation(s)
- Jonathan Gear
- Joint Department of Physics, Royal Marsden Hospital & Institute of Cancer Research, Sutton, UK.
| | - Carlo Chiesa
- Nuclear Medicine, Foundation IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Michael Lassmann
- Department of Nuclear Medicine, University of Würzburg, 97080, Würzburg, Germany
| | - Pablo Mínguez Gabiña
- Department of Medical Physics and Radiation Protection, Gurutzeta/Cruces University Hospital, Barakaldo, Spain
| | - Johannes Tran-Gia
- Department of Nuclear Medicine, University of Würzburg, 97080, Würzburg, Germany
| | - Caroline Stokke
- Department of Diagnostic Physics, Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Glenn Flux
- Joint Department of Physics, Royal Marsden Hospital & Institute of Cancer Research, Sutton, UK
| |
Collapse
|
48
|
Pozzessere C, Bassanelli M, Ceribelli A, Rasul S, Li S, Prior JO, Cicone F. Renal Cell Carcinoma: the Oncologist Asks, Can PSMA PET/CT Answer? Curr Urol Rep 2019; 20:68. [PMID: 31605269 DOI: 10.1007/s11934-019-0938-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE OF REVIEW To critically review the potential clinical applications of prostate-specific membrane antigen (PSMA) radioactive ligands in renal cell carcinoma (RCC). RECENT FINDINGS Radioactive probes targeting PSMA hold promise in several malignancies in addition to prostate cancer, owing to the expression of PSMA by tumor neovasculature. The majority of clear cell RCCs (ccRCC), the most malignant RCC subtype, express PSMA on tumor-associated neovasculature. The endothelium of less aggressive RCC subtypes is PSMA positive in a lower, but still significant percentage of cases. PSMA might therefore represent an interesting theragnostic target in RCC. The preliminary data available suggest a potential role for PSMA-targeting radiopharmaceuticals in complementing conventional imaging for staging ccRCC patients at risk of nodal involvement and oligometastatic disease. Additional applications of PSMA imaging may be the selection and the response assessment of patients receiving anti-angiogenic treatments. The effectiveness of PSMA-targeting radionuclide therapy should also be investigated.
Collapse
Affiliation(s)
- Chiara Pozzessere
- Department of Radiology, AUSL Toscana Centro San Giuseppe Hospital, Viale Boccaccio 20, 50053, Empoli, Italy.
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland.
| | - Maria Bassanelli
- Division of Medical Oncology, San Camillo De Lellis Hospital, Rieti, Italy
| | - Anna Ceribelli
- Division of Medical Oncology, San Camillo De Lellis Hospital, Rieti, Italy
| | - Sazan Rasul
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Shuren Li
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - John O Prior
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Francesco Cicone
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
49
|
Hosono M. Perspectives for Concepts of Individualized Radionuclide Therapy, Molecular Radiotherapy, and Theranostic Approaches. Nucl Med Mol Imaging 2019; 53:167-171. [PMID: 31231436 PMCID: PMC6554368 DOI: 10.1007/s13139-019-00586-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 01/18/2019] [Accepted: 01/22/2019] [Indexed: 11/29/2022] Open
Abstract
Radionuclide therapy (RNT) stands on the delivery of radiation to tumors or non-tumor target organs using radiopharmaceuticals that are designed to have specific affinity to targets. RNT is recently called molecular radiotherapy (MRT) by some advocators in order to emphasize its characteristics as radiotherapy and the relevance of dosimetry-guided optimization of treatment. Moreover, RNT requires relevant radiation protection standards because it employs unsealed radionuclides and gives therapeutic radiation doses in humans. On the basis of these radiation protection standards, the development and use of radiopharmaceuticals for combined application through diagnostics and therapeutics lead to theranostic approaches that will enhance the efficacy and safety of treatment by implementing dosimetry-based individualization.
Collapse
Affiliation(s)
- Makoto Hosono
- Institute of Advanced Clinical Medicine and Department of Radiology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka 589-8511 Japan
| |
Collapse
|
50
|
Malcolm J, Falzone N, Lee BQ, Vallis KA. Targeted Radionuclide Therapy: New Advances for Improvement of Patient Management and Response. Cancers (Basel) 2019; 11:E268. [PMID: 30823564 PMCID: PMC6406485 DOI: 10.3390/cancers11020268] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 02/20/2019] [Accepted: 02/21/2019] [Indexed: 12/20/2022] Open
Abstract
Compared to external beam radiotherapy, targeted radionuclide therapy (TRT) allows for systemic radiation treatment of metastatic lesions. Published work on recent strategies to improve patient management and response to TRT through individualising patient treatment, modifying treatment pharmacokinetics and increasing anticancer potency are discussed in this review, with a special focus on the application of clinically evaluated radiolabelled ligands and peptides in the treatment of neuroendocrine and prostate cancers.
Collapse
Affiliation(s)
- Javian Malcolm
- CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford OX37DQ, UK.
| | - Nadia Falzone
- CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford OX37DQ, UK.
| | - Boon Q Lee
- CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford OX37DQ, UK.
| | - Katherine A Vallis
- CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford OX37DQ, UK.
| |
Collapse
|