1
|
Vergnaud L, Dewaraja YK, Giraudet AL, Badel JN, Sarrut D. A review of 177Lu dosimetry workflows: how to reduce the imaging workloads? EJNMMI Phys 2024; 11:65. [PMID: 39023648 DOI: 10.1186/s40658-024-00658-8] [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: 01/25/2024] [Accepted: 06/07/2024] [Indexed: 07/20/2024] Open
Abstract
177 Lu radiopharmaceutical therapy is a standardized systemic treatment, with a typical dose of 7.4 GBq per injection, but its response varies from patient to patient. Dosimetry provides the opportunity to personalize treatment, but it requires multiple post-injection images to monitor the radiopharmaceutical's biodistribution over time. This imposes an additional imaging burden on centers with limited resources. This review explores methods to lessen this burden by optimizing acquisition types and minimizing the number and duration of imaging sessions. After summarizing the different steps of dosimetry and providing examples of dosimetric workflows for177 Lu -DOTATATE and177 Lu -PSMA, we examine dosimetric workflows based on a reduced number of acquisitions, or even just one. We provide a non-exhaustive description of simplified methods and their assumptions, as well as their limitations. Next, we detail the specificities of each normal tissue and tumors, before reviewing dose-response relationships in the literature. In conclusion, we will discuss the current limitations of dosimetric workflows and propose avenues for improvement.
Collapse
Affiliation(s)
- Laure Vergnaud
- CREATIS; CNRS UMR 5220; INSERM U 1044, Université de Lyon; INSA-Lyon; Université Lyon 1, Lyon, France.
| | - Yuni K Dewaraja
- Department of Radiology, University of Michigan, Ann Arbor, USA
| | | | - Jean-Noël Badel
- CREATIS; CNRS UMR 5220; INSERM U 1044, Université de Lyon; INSA-Lyon; Université Lyon 1, Lyon, France
- Centre de lutte contre le cancer Léon Bérard, Lyon, France
| | - David Sarrut
- CREATIS; CNRS UMR 5220; INSERM U 1044, Université de Lyon; INSA-Lyon; Université Lyon 1, Lyon, France
- Centre de lutte contre le cancer Léon Bérard, Lyon, France
| |
Collapse
|
2
|
Warfvinge CF, Gustafsson J, Roth D, Tennvall J, Svensson J, Bernhardt P, Åkesson A, Wieslander E, Sundlöv A, Sjögreen Gleisner K. Relationship Between Absorbed Dose and Response in Neuroendocrine Tumors Treated with [ 177Lu]Lu-DOTATATE. J Nucl Med 2024; 65:1070-1075. [PMID: 38724277 DOI: 10.2967/jnumed.123.266991] [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: 11/16/2023] [Accepted: 03/26/2024] [Indexed: 07/03/2024] Open
Abstract
Peptide receptor radionuclide therapy presents the possibility of tracing and quantifying the uptake of the drug in the body and performing dosimetry, potentially allowing individualization of treatment schemes. However, the details of how neuroendocrine tumors (NETs) respond to different absorbed doses are insufficiently known. Here, we investigated the relationship between tumor-absorbed dose and tumor response in a cohort of patients with NETs treated with [177Lu]Lu-DOTATATE. Methods: This was a retrospective study based on 69 tumors in 32 patients treated within a clinical trial. Dosimetry was performed at each cycle of [177Lu]Lu-DOTATATE, rendering 366 individual absorbed dose assessments. Hybrid planar-SPECT/CT imaging using [177Lu]Lu-DOTATATE was used, including quantitative SPECT reconstruction, voxel-based absorbed dose rate calculation, semiautomatic image segmentation, and partial-volume correction. Changes in tumor volume were used to determine tumor response. The volume for each tumor was manually delineated on consecutive CT scans, giving a total of 712 individual tumor volume assessments. Tumors were stratified according to grade. The relationship between absorbed dose and response was investigated using mixed-effects models and logistic regression. Tumors smaller than 4 cm3 were excluded. Results: In grade 2 NETs, a clear relationship between absorbed dose and volume reduction was observed. Our observations suggest a 90% probability of partial tumor response for an accumulated tumor-absorbed dose of at least 135 Gy. Conclusion: Our findings are in accordance with previous observations regarding the relationship between tumor shrinkage and absorbed dose. Moreover, our data suggest an absorbed dose threshold for partial response in grade 2 NETs. These observations provide valuable insights for the design of dosimetry-guided peptide receptor radionuclide therapy schemes.
Collapse
Affiliation(s)
- Carl Fredrik Warfvinge
- Department of Hematology, Oncology, and Radiation Physics, Skåne University Hospital, Lund, Sweden;
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | | | - Daniel Roth
- Medical Radiation Physics, Lund University, Lund, Sweden
| | - Jan Tennvall
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Johanna Svensson
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Peter Bernhardt
- Department of Medical Physics and Biomedical Engineering, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Medical Radiation Sciences, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; and
| | - Anna Åkesson
- Clinical Studies Sweden, Forum South, Skåne University Hospital, Lund, Sweden
| | - Elinore Wieslander
- Department of Hematology, Oncology, and Radiation Physics, Skåne University Hospital, Lund, Sweden
| | - Anna Sundlöv
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | | |
Collapse
|
3
|
Hebert K, Santoro L, Monnier M, Castan F, Berkane I, Assénat E, Fersing C, Gélibert P, Pouget JP, Bardiès M, Kotzki PO, Deshayes E. Absorbed Dose-Response Relationship in Patients with Gastroenteropancreatic Neuroendocrine Tumors Treated with [ 177Lu]Lu-DOTATATE: One Step Closer to Personalized Medicine. J Nucl Med 2024; 65:923-930. [PMID: 38637144 PMCID: PMC11149595 DOI: 10.2967/jnumed.123.267023] [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: 11/15/2023] [Revised: 03/04/2024] [Indexed: 04/20/2024] Open
Abstract
[177Lu]Lu-DOTATATE has been approved for progressive and inoperable gastroenteropancreatic neuroendocrine tumors (GEP-NETs) that overexpress somatostatin receptors. The absorbed doses by limiting organs and tumors can be quantified by serial postinfusion scintigraphy measurements of the γ-emissions from 177Lu. The objective of this work was to explore how postinfusion [177Lu]Lu-DOTATATE dosimetry could influence clinical management by predicting treatment efficacy (tumor shrinkage and survival) and toxicity. Methods: Patients with GEP-NETs treated with [177Lu]Lu-DOTATATE between 2016 and 2022 and who underwent dosimetry were included. Absorbed doses were calculated for healthy organs (liver, kidneys, bone marrow, and spleen) and tumors using PLANET Dose and the local energy deposition method based on serial posttreatment SPECT/CT. Up to 5 lesions per site were selected and measured on images collected at baseline and 3 mo after treatment end (measurement masked to the somatostatin receptor imaging uptake). For toxicity assessment, laboratory parameters were regularly monitored. Clinical data, including time to death or progression, were collected from the patients' health records. Correlations between absorbed doses by organs and toxicity and between absorbed doses by lesions and tumor volume variation were studied using regression models. Results: In total, 35 dosimetric studies were performed in patients with mostly grade 2 (77%) tumors and metastases in liver (89%), lymph nodes (77%), and bone (34%), and 146 lesions were analyzed: 1-9 lesions per patient, mostly liver metastases (65%) and lymph nodes (25%). The median total absorbed dose by tumors was 94.4 Gy. The absorbed doses by tumors significantly decreased between cycles. The absorbed dose by tumors was significantly associated with tumor volume variation (P < 0.001) 3 mo after treatment end, and it was a significant prognostic factor for survival. Toxicity analysis showed a correlation between the decrease of hematologic parameters such as lymphocytes or platelet concentrations and the absorbed doses by the spleen or bone marrow. The mean absorbed dose by the kidneys was not correlated with nephrotoxicity during the studied period. Conclusion: In patients treated with [177Lu]Lu-DOTATATE for GEP-NETs, tumor and healthy organ dosimetry can predict survival and toxicities, thus influencing clinical management.
Collapse
Affiliation(s)
- Kévin Hebert
- Department of Nuclear Medicine, Institut du Cancer de Montpellier, Université de Montpellier, Montpellier, France
| | - Lore Santoro
- 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
| | - Maeva Monnier
- Biometry Unit, Institut du Cancer de Montpellier, Université de Montpellier, Montpellier, France
| | - Florence Castan
- Biometry Unit, Institut du Cancer de Montpellier, Université de Montpellier, Montpellier, France
| | - Ikrame Berkane
- Department of Nuclear Medicine, Institut du Cancer de Montpellier, Université de Montpellier, Montpellier, France
| | - Eric Assénat
- Department of Medical Oncology, CHU de Montpellier, Université de Montpellier, Montpellier, France
| | - Cyril Fersing
- 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
- Biometry Unit, Institut du Cancer de Montpellier, Université de Montpellier, Montpellier, France
- Department of Medical Oncology, CHU de Montpellier, Université de Montpellier, Montpellier, France
- IBMM, Université de Montpellier, CNRS, ENSCM, Montpellier, France; and
| | | | - Jean-Pierre Pouget
- Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Montpellier, France
| | - Manuel Bardiès
- 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
| | - Pierre-Olivier Kotzki
- 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
| | - Emmanuel Deshayes
- 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
|
4
|
Hoogenkamp DS, de Wit-van der Veen LJ, Huizing DMV, Tesselaar MET, van Leeuwaarde RS, Stokkel MPM, Lam MGEH, Braat AJAT. Advances in Radionuclide Therapies for Patients with Neuro-endocrine Tumors. Curr Oncol Rep 2024; 26:551-561. [PMID: 38598035 PMCID: PMC11062977 DOI: 10.1007/s11912-024-01521-w] [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] [Accepted: 03/21/2024] [Indexed: 04/11/2024]
Abstract
PURPOSE OF REVIEW To provide insights into the role of peptide receptor radionuclide therapy (PRRT) in patients with advanced neuroendocrine tumors (NET) and an overview of possible strategies to combine PRRT with locoregional and systemic anticancer treatments. RECENT FINDINGS Research on combining PRRT with other treatments encompasses a wide variety or treatments, both local (transarterial radioembolization) and systemic therapies, chemotherapy (i.e., capecitabine and temozolomide), targeted therapies (i.e., olaparib, everolimus, and sunitinib), and immunotherapies (e.g., nivolumab and pembrolizumab). Furthermore, PRRT shows promising first results as a treatment prior to surgery. There is great demand to enhance the efficacy of PRRT through combination with other anticancer treatments. While research in this area is currently limited, the field is rapidly evolving with numerous ongoing clinical trials aiming to address this need and explore novel therapeutic combinations.
Collapse
Affiliation(s)
- Denise S Hoogenkamp
- Department of Nuclear Medicine, Netherlands Cancer Institute, Amsterdam, The Netherlands
- ENETS Center of Excellence NKI-AVL, The Netherlands Cancer Institute/UMC Utrecht, Amsterdam, The Netherlands
| | - Linda J de Wit-van der Veen
- Department of Nuclear Medicine, Netherlands Cancer Institute, Amsterdam, The Netherlands
- ENETS Center of Excellence NKI-AVL, The Netherlands Cancer Institute/UMC Utrecht, Amsterdam, The Netherlands
| | - Daphne M V Huizing
- Department of Nuclear Medicine, Netherlands Cancer Institute, Amsterdam, The Netherlands
- ENETS Center of Excellence NKI-AVL, The Netherlands Cancer Institute/UMC Utrecht, Amsterdam, The Netherlands
| | - Margot E T Tesselaar
- ENETS Center of Excellence NKI-AVL, The Netherlands Cancer Institute/UMC Utrecht, Amsterdam, The Netherlands
- Department of Gastrointestinal Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Rachel S van Leeuwaarde
- ENETS Center of Excellence NKI-AVL, The Netherlands Cancer Institute/UMC Utrecht, Amsterdam, The Netherlands
- Department of Endocrinology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marcel P M Stokkel
- Department of Nuclear Medicine, Netherlands Cancer Institute, Amsterdam, The Netherlands
- ENETS Center of Excellence NKI-AVL, The Netherlands Cancer Institute/UMC Utrecht, Amsterdam, The Netherlands
| | - Marnix G E H Lam
- ENETS Center of Excellence NKI-AVL, The Netherlands Cancer Institute/UMC Utrecht, Amsterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, 3584 CX, Utrecht, The Netherlands
| | - Arthur J A T Braat
- Department of Nuclear Medicine, Netherlands Cancer Institute, Amsterdam, The Netherlands.
- ENETS Center of Excellence NKI-AVL, The Netherlands Cancer Institute/UMC Utrecht, Amsterdam, The Netherlands.
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, 3584 CX, Utrecht, The Netherlands.
| |
Collapse
|
5
|
Siebinga H, de Wit-van der Veen BJ, de Vries-Huizing DMV, Vogel WV, Hendrikx JJMA, Huitema ADR. Quantification of biochemical PSA dynamics after radioligand therapy with [ 177Lu]Lu-PSMA-I&T using a population pharmacokinetic/pharmacodynamic model. EJNMMI Phys 2024; 11:39. [PMID: 38656678 PMCID: PMC11043318 DOI: 10.1186/s40658-024-00642-2] [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: 09/25/2023] [Accepted: 04/12/2024] [Indexed: 04/26/2024] Open
Abstract
BACKGROUND There is an unmet need for prediction of treatment outcome or patient selection for [177Lu]Lu-PSMA therapy in patients with metastatic castration-resistant prostate cancer (mCRPC). Quantification of the tumor exposure-response relationship is pivotal for further treatment optimization. Therefore, a population pharmacokinetic (PK) model was developed for [177Lu]Lu-PSMA-I&T using SPECT/CT data and, subsequently, related to prostate-specific antigen (PSA) dynamics after therapy in patients with mCRPC using a pharmacokinetic/pharmacodynamic (PKPD) modelling approach. METHODS A population PK model was developed using quantitative SPECT/CT data (406 scans) of 76 patients who received multiple cycles [177Lu]Lu-PSMA-I&T (± 7.4 GBq with either two- or six-week interval). The PK model consisted of five compartments; central, salivary glands, kidneys, tumors and combined remaining tissues. Covariates (tumor volume, renal function and cycle number) were tested to explain inter-individual variability on uptake into organs and tumors. The final PK model was expanded with a PD compartment (sequential fitting approach) representing PSA dynamics during and after treatment. To explore the presence of a exposure-response relationship, individually estimated [177Lu]Lu-PSMA-I&T tumor concentrations were related to PSA changes over time. RESULTS The population PK model adequately described observed data in all compartments (based on visual inspection of goodness-of-fit plots) with adequate precision of parameters estimates (< 36.1% relative standard error (RSE)). A significant declining uptake in tumors (k14) during later cycles was identified (uptake decreased to 73%, 50% and 44% in cycle 2, 3 and 4-7, respectively, compared to cycle 1). Tumor growth (defined by PSA increase) was described with an exponential growth rate (0.000408 h-1 (14.2% RSE)). Therapy-induced PSA decrease was related to estimated tumor concentrations (MBq/L) using both a direct and delayed drug effect. The final model adequately captured individual PSA concentrations after treatment (based on goodness-of-fit plots). Simulation based on the final PKPD model showed no evident differences in response for the two different dosing regimens currently used. CONCLUSIONS Our population PK model accurately described observed [177Lu]Lu-PSMA-I&T uptake in salivary glands, kidneys and tumors and revealed a clear declining tumor uptake over treatment cycles. The PKPD model adequately captured individual PSA observations and identified population response rates for the two dosing regimens. Hence, a PKPD modelling approach can guide prediction of treatment response and thus identify patients in whom radioligand therapy is likely to fail.
Collapse
Affiliation(s)
- Hinke Siebinga
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute: Antoni Van Leeuwenhoek, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands.
- Department of Nuclear Medicine, The Netherlands Cancer Institute: Antoni Van Leeuwenhoek, Amsterdam, The Netherlands.
| | | | - Daphne M V de Vries-Huizing
- Department of Nuclear Medicine, The Netherlands Cancer Institute: Antoni Van Leeuwenhoek, Amsterdam, The Netherlands
| | - Wouter V Vogel
- Department of Nuclear Medicine, The Netherlands Cancer Institute: Antoni Van Leeuwenhoek, Amsterdam, The Netherlands
- Department of Radiation Oncology, The Netherlands Cancer Institute: Antoni Van Leeuwenhoek, Amsterdam, The Netherlands
| | - Jeroen J M A Hendrikx
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute: Antoni Van Leeuwenhoek, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
- Department of Nuclear Medicine, The Netherlands Cancer Institute: Antoni Van Leeuwenhoek, Amsterdam, The Netherlands
| | - Alwin D R Huitema
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute: Antoni Van Leeuwenhoek, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
- Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Pharmacology, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| |
Collapse
|
6
|
Vasić V, Gustafsson J, Nowshahr EY, Stenvall A, Beer AJ, Gleisner KS, Glatting G. A PBPK model for PRRT with [ 177Lu]Lu-DOTA-TATE: Comparison of model implementations in SAAM II and MATLAB/SimBiology. Phys Med 2024; 119:103299. [PMID: 38367588 DOI: 10.1016/j.ejmp.2024.103299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 12/06/2023] [Accepted: 01/23/2024] [Indexed: 02/19/2024] Open
Abstract
Physiologically based pharmacokinetic (PBPK) models offer the ability to simulate and predict the biodistribution of radiopharmaceuticals and have the potential to enable individualised treatment planning in molecular radiotherapy. The objective of this study was to develop and implement a whole-body compartmental PBPK model for peptide receptor radionuclide therapy (PRRT) with [177Lu]Lu-DOTA-TATE in SimBiology to allow for more complex analyses. The correctness of the model implementation was ensured by comparing its outputs, such as the time-integrated activity (TIA), with those of a PBPK model implemented in SAAM II software. METHODS A combined PBPK model for [68Ga]Ga-DOTA-TATE and [177Lu]Lu-DOTA-TATE was developed and implemented in both SAAM II and SimBiology. A retrospective analysis of 12 patients with metastatic neuroendocrine tumours (NETs) was conducted. First, time-activity curves (TACs) and TIAs from the two software were calculated and compared for identical parameter values. Second, pharmacokinetic parameters were fitted to activity concentrations, analysed and compared. RESULTS The PBPK model implemented in SimBiology produced TIA results comparable to those generated by the model implemented in SAAM II, with a relative deviation of less than 0.5% when using the same input parameters. The relative deviation of the fitted TIAs was less than 5% when model parameter values were fitted to the measured activity concentrations. CONCLUSION The proposed PBPK model implemented in SimBiology can be used for dosimetry in radioligand therapy and TIA prediction. Its outputs are similar to those generated by the PBPK model implemented in SAAM II, confirming the correctness of the model implementation in SimBiology.
Collapse
Affiliation(s)
- Valentina Vasić
- Department of Nuclear Medicine, Ulm University, Ulm, Germany; Medical Radiation Physics, Department of Nuclear Medicine, Ulm University, Ulm, Germany.
| | | | - Elham Yousefzadeh Nowshahr
- Department of Nuclear Medicine, Ulm University, Ulm, Germany; Medical Radiation Physics, Department of Nuclear Medicine, Ulm University, Ulm, Germany
| | - Anna Stenvall
- Medical Radiation Physics, Lund University, Lund, Sweden
| | - Ambros J Beer
- Department of Nuclear Medicine, Ulm University, Ulm, Germany
| | | | - Gerhard Glatting
- Department of Nuclear Medicine, Ulm University, Ulm, Germany; Medical Radiation Physics, Department of Nuclear Medicine, Ulm University, Ulm, Germany
| |
Collapse
|
7
|
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
|
8
|
Ha S, Kim YI, Oh JS, Yoo C, Ryoo BY, Ryu JS. Prediction of [ 177Lu]Lu-DOTA-TATE therapy response using the absorbed dose estimated from [ 177Lu]Lu-DOTA-TATE SPECT/CT in patients with metastatic neuroendocrine tumour. EJNMMI Phys 2024; 11:14. [PMID: 38315270 PMCID: PMC10844176 DOI: 10.1186/s40658-024-00620-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 01/29/2024] [Indexed: 02/07/2024] Open
Abstract
BACKGROUND Peptide receptor radionuclide therapy (PRRT) with [177Lu]Lu-DOTA-TATE has shown efficacy in patients with metastatic neuroendocrine tumours (NETs). Personalised dosimetry is crucial to optimise treatment outcomes and minimise adverse events. In this study, we investigated the correlation between the tumour-absorbed dose (TAD) estimated from [177Lu]Lu-DOTA-TATE SPECT/CT and the therapeutic response. METHOD A retrospective analysis was conducted on patients with advanced well-differentiated NETs grades 1-3 who underwent PRRT and exhibited greater uptake than liver on pre-therapeutic [68Ga]Ga-DOTA-TOC PET/CT. Target lesions were selected based on the RECIST 1.1 and PERCIST 1.0 criteria using [177Lu]Lu-DOTA-TATE SPECT/CT and pre-therapeutic contrast-enhanced CT scans. For anatomical image analysis, the sum of the longest diameter (SLD) of the target lesions was measured using the RECIST 1.1 criteria for patient-based analysis and the longest diameter (LD) of the target lesion using the RECIST-L criteria for lesion-based analysis. Standardised uptake values (SUVs) were measured on SPECT/CT images, and TADs were calculated based on the SUVs. Dosimetry was performed using a single SPECT/CT imaging time point at day 4-5 post-therapy. Statistical analyses were conducted to investigate correlations and determine the target lesion responses. RESULTS Twenty patients with primary tumour sites and hepatic metastases were included. Fifty-five target lesions, predominantly located in the pancreas and liver, were analysed. The cumulative TAD (lesion-based analysis: r = 0.299-0.301, p = 0.025-0.027), but not the cycle 1 SUV (lesion-based analysis: r = 0.198-0.206, p = 0.131-0.147) or cycle 1 TAD (lesion-based analysis: r = 0.209-0.217, p = 0.112-0.126), exhibited a significant correlation with the change in LD of the target lesion. Binary logistic regression analysis identified the significance of the cumulative TAD in predicting disease control according to the RECIST-L criteria (odds ratio = 1.031-1.051, p = 0.024-0.026). CONCLUSIONS The cumulative TAD estimated from [177Lu]Lu-DOTA-TATE SPECT/CT revealed a significant correlation with change in LD, which was significantly higher for the cumulative TAD than for the cycle 1 SUV or TAD. A higher cumulative TAD was associated with disease control in the target lesion. However, considering the limitations inherent to a confined sample size, careful interpretation of these findings is required. Estimation of the cumulative TAD of [177Lu]Lu-DOTA-TATE therapy could guide the platform towards personalised therapy.
Collapse
Affiliation(s)
- Sejin Ha
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Yong-Il Kim
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
- Theranostics Center, Asan Cancer Institute, Asan Medical Center, Seoul, Republic of Korea.
| | - Jungsu S Oh
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Theranostics Center, Asan Cancer Institute, Asan Medical Center, Seoul, Republic of Korea
| | - Changhoon Yoo
- Theranostics Center, Asan Cancer Institute, Asan Medical Center, Seoul, Republic of Korea
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Baek-Yeol Ryoo
- Theranostics Center, Asan Cancer Institute, Asan Medical Center, Seoul, Republic of Korea
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jin-Sook Ryu
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Theranostics Center, Asan Cancer Institute, Asan Medical Center, Seoul, Republic of Korea
| |
Collapse
|
9
|
Siebinga H, Hendrikx JJMA, de Vries-Huizing DMV, Huitema ADR, de Wit-van der Veen BJ. The cycle effect quantified: reduced tumour uptake in subsequent cycles of [ 177Lu]Lu-HA-DOTATATE during peptide receptor radionuclide therapy. Eur J Nucl Med Mol Imaging 2024; 51:820-827. [PMID: 37843598 DOI: 10.1007/s00259-023-06463-2] [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: 06/01/2023] [Accepted: 10/01/2023] [Indexed: 10/17/2023]
Abstract
BACKGROUND Clear evidence regarding the effect of reduced tumour accumulation in later peptide receptor radionuclide therapy (PRRT) cycles is lacking. Therefore, we aimed to quantify potential cycle effects for patients treated with [177Lu]Lu-HA-DOTATATE using a population pharmacokinetic (PK) modelling approach. METHODS A population PK model was developed using imaging data from 48 patients who received multiple cycles of [177Lu]Lu-HA-DOTATATE. The five-compartment model included a central, kidney, spleen, tumour and lumped rest compartment. Tumour volume and continued use of long-acting somatostatin analogues (SSAs) were tested as covariates in the model. In addition, the presence of a cycle effect was evaluated by relating the uptake rate in a specific cycle as a fraction of the (tumour or organ) uptake rate in the first cycle. RESULTS The final PK model adequately captured observed radioactivity accumulation in kidney, spleen and tumour. A higher tumour volume was identified to increase the tumour uptake rate, where a twofold increase in tumour volume resulted in a 2.3-fold higher uptake rate. Also, continued use of long-acting SSAs significantly reduced the spleen uptake rate (68.4% uptake compared to SSA withdrawal (10.5% RSE)). Lastly, a cycle effect was significantly identified, where tumour uptake rate decreased to 86.9% (5.3% RSE) in the second cycle and even further to 79.7% (5.6% RSE) and 77.6% (6.2% RSE) in the third and fourth cycle, respectively, compared to cycle one. CONCLUSIONS Using a population PK modelling approach, the cycle effect of reduced tumour uptake in subsequent PRRT cycles was quantified. Our findings implied that downregulation of target receptors is probably not the major cause of the cycle effect, due to a plateau in the decrease of tumour uptake in the fourth cycle.
Collapse
Affiliation(s)
- H Siebinga
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
- Department of Nuclear Medicine, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
| | - J J M A Hendrikx
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Nuclear Medicine, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - D M V de Vries-Huizing
- Department of Nuclear Medicine, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - A D R Huitema
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Pharmacology, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - B J de Wit-van der Veen
- Department of Nuclear Medicine, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
10
|
Al-Toubah T, Strosberg J, Hallanger-Johnson J, El-Haddad G. Targeted radionuclide therapy in endocrine-related cancers: advances in the last decade. Front Endocrinol (Lausanne) 2023; 14:1187870. [PMID: 38053729 PMCID: PMC10694449 DOI: 10.3389/fendo.2023.1187870] [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: 03/16/2023] [Accepted: 10/26/2023] [Indexed: 12/07/2023] Open
Abstract
Targeted radionuclide therapy plays an increasingly important role in managing endocrine-related tumors and significantly advances the therapeutic landscape for patients with these diseases. With increasing FDA-approved therapies and advances in the field, come an increased knowledge of the potential for long-term toxicities associated with these therapies and the field must develop new strategies to increase potency and efficacy while individualizing the selection of patients to those most likely to respond to treatment. Novel agents and modalities of therapy are also being explored. This review will discuss the current landscape and describe the avenues for growth in the field currently being explored.
Collapse
Affiliation(s)
- Taymeyah Al-Toubah
- Department of GI Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
| | - Jonathan Strosberg
- Department of GI Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
| | - Julie Hallanger-Johnson
- Department of Head and Neck - Endocrine Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
| | - Ghassan El-Haddad
- Department of Radiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
- Department of Nuclear Medicine, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
| |
Collapse
|
11
|
Plachouris D, Eleftheriadis V, Nanos T, Papathanasiou N, Sarrut D, Papadimitroulas P, Savvidis G, Vergnaud L, Salvadori J, Imperiale A, Visvikis D, Hazle JD, Kagadis GC. A radiomic- and dosiomic-based machine learning regression model for pretreatment planning in 177 Lu-DOTATATE therapy. Med Phys 2023; 50:7222-7235. [PMID: 37722718 DOI: 10.1002/mp.16746] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 09/01/2023] [Accepted: 09/07/2023] [Indexed: 09/20/2023] Open
Abstract
BACKGROUND Standardized patient-specific pretreatment dosimetry planning is mandatory in the modern era of nuclear molecular radiotherapy, which may eventually lead to improvements in the final therapeutic outcome. Only a comprehensive definition of a dosage therapeutic window encompassing the range of absorbed doses, that is, helpful without being detrimental can lead to therapy individualization and improved outcomes. As a result, setting absorbed dose safety limits for organs at risk (OARs) requires knowledge of the absorbed dose-effect relationship. Data sets of consistent and reliable inter-center dosimetry findings are required to characterize this relationship. PURPOSE We developed and standardized a new pretreatment planning model consisting of a predictive dosimetry procedure for OARs in patients with neuroendocrine tumors (NETs) treated with 177 Lu-DOTATATE (Lutathera). In the retrospective study described herein, we used machine learning (ML) regression algorithms to predict absorbed doses in OARs by exploiting a combination of radiomic and dosiomic features extracted from patients' imaging data. METHODS Pretreatment and posttreatment data for 20 patients with NETs treated with 177 Lu-DOTATATE were collected from two clinical centers. A total of 3412 radiomic and dosiomic features were extracted from the patients' computed tomography (CT) scans and dose maps, respectively. All dose maps were generated using Monte Carlo simulations. An ML regression model was designed based on ML algorithms for predicting the absorbed dose in every OAR (liver, left kidney, right kidney, and spleen) before and after the therapy and between each therapy session, thus predicting any possible radiotoxic effects. RESULTS We evaluated nine ML regression algorithms. Our predictive model achieved a mean absolute dose error (MAE, in Gy) of 0.61 for the liver, 1.58 for the spleen, 1.30 for the left kidney, and 1.35 for the right kidney between pretherapy 68 Ga-DOTATOC positron emission tomography (PET)/CT and posttherapy 177 Lu-DOTATATE single photon emission (SPECT)/CT scans. Τhe best predictive performance observed was based on the gradient boost for the liver, the left kidney and the right kidney, and on the extra tree regressor for the spleen. Evaluation of the model's performance according to its ability to predict the absorbed dose in each OAR in every possible combination of pretherapy 68 Ga-DOTATOC PET/CT and any posttherapy 177 Lu-DOTATATE treatment cycle SPECT/CT scans as well as any 177 Lu-DOTATATE SPECT/CT treatment cycle and the consequent 177 Lu-DOTATATE SPECT/CT treatment cycle revealed mean absorbed dose differences ranges from -0.55 to 0.68 Gy. Incorporating radiodosiomics features from the 68 Ga-DOTATOC PET/CT and first 177 Lu-DOTATATE SPECT/CT treatment cycle scans further improved the precision and minimized the standard deviation of the predictions in nine out of 12 instances. An average improvement of 57.34% was observed (range: 17.53%-96.12%). However, it's important to note that in three instances (i.e., Ga,C.1 → C3 in spleen and left kidney, and Ga,C.1 → C2 in right kidney) we did not observe an improvement (absolute differences of 0.17, 0.08, and 0.05 Gy, respectively). Wavelet-based features proved to have high correlated predictive value, whereas non-linear-based ML regression algorithms proved to be more capable than the linear-based of producing precise prediction in our case. CONCLUSIONS The combination of radiomics and dosiomics has potential utility for personalized molecular radiotherapy (PMR) response evaluation and OAR dose prediction. These radiodosiomic features can potentially provide information on any possible disease recurrence and may be highly useful in clinical decision-making, especially regarding dose escalation issues.
Collapse
Affiliation(s)
- Dimitris Plachouris
- 3DMI Research Group, Department of Medical Physics, School of Medicine, University of Patras, Rion, Greece
| | | | - Thomas Nanos
- 3DMI Research Group, Department of Medical Physics, School of Medicine, University of Patras, Rion, Greece
| | | | | | | | | | | | | | | | | | - John D Hazle
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - George C Kagadis
- 3DMI Research Group, Department of Medical Physics, School of Medicine, University of Patras, Rion, Greece
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
12
|
Alipour R, Jackson P, Bressel M, Hogg A, Callahan J, Hicks RJ, Kong G. The relationship between tumour dosimetry, response, and overall survival in patients with unresectable Neuroendocrine Neoplasms (NEN) treated with 177Lu DOTATATE (LuTate). Eur J Nucl Med Mol Imaging 2023; 50:2997-3010. [PMID: 37184682 PMCID: PMC10382388 DOI: 10.1007/s00259-023-06257-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 04/30/2023] [Indexed: 05/16/2023]
Abstract
Peptide Receptor Radionuclide Therapy (PRRT) delivers targeted radiation to Somatostatin Receptor (SSR) expressing Neuroendocrine Neoplasms (NEN). We sought to assess the predictive and prognostic implications of tumour dosimetry with respect to response by 68 Ga DOTATATE (GaTate) PET/CT molecular imaging tumour volume of SSR (MITVSSR) change and RECIST 1.1, and overall survival (OS). METHODS Patients with gastro-entero-pancreatic (GEP) NEN who received LuTate followed by quantitative SPECT/CT (Q-SPECT/CT) the next day (Jul 2010 to Jan 2019) were retrospectively reviewed. Single time-point (STP) lesional dosimetry was performed for each cycle using population-based pharmacokinetic modelling. MITVSSR and RECIST 1.1 were measured at 3-months post PRRT. RESULTS Median of 4 PRRT cycles were administered to 90 patients (range 2-5 cycles; mean 27.4 GBq cumulative activity; mean 7.6 GBq per cycle). 68% received at least one cycle with radiosensitising chemotherapy (RSC). RECIST 1.1 partial response was 24%, with 70% stable and 7% progressive disease. Cycle 1 radiation dose in measurable lesions was associated with local response (odds ratio 1.5 per 50 Gy [95% CI: 1.1-2.0], p = 0.002) when adjusted by tumour grade and RSC. Median change in MITVSSR was -63% (interquartile range -84 to -29), with no correlation with radiation dose to the most avid lesion on univariable or multivariant analyses (5.6 per 10 Gy [95% CI: -1.6, 12.8], p = 0.133). OS at 5-years was 68% (95% CI: 56-78%). Neither baseline MITVSSR (hazard ratio 1.1 [95% CI: 1.0, 1.2], p = 0.128) nor change in baseline MITVSSR (hazard ratio 1.0 [95% CI: 1.0, 1.1], p = 0.223) were associated with OS when adjusted by tumour grade and RSC but RSC was (95% CI: 0.2, 0.8, p = 0.012). CONCLUSION Radiation dose to tumour during PRRT was predictive of radiologic response but not survival. Survival outcomes may relate to other biological factors. There was no evidence that MITVSSR change was associated with OS, but a larger study is needed.
Collapse
Affiliation(s)
- R Alipour
- Department of Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Australia.
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia.
| | - P Jackson
- Department of Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Australia
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| | - M Bressel
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
- Centre for Biostatistics and Clinical Trials, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - A Hogg
- Department of Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - J Callahan
- Department of Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - R J Hicks
- Department of Medicine, St Vincent's Medical School, The University of Melbourne, Melbourne, Australia
| | - G Kong
- Department of Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Australia
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| |
Collapse
|
13
|
Jahn U, Garske-Román U, Sandström M, Lubberink M, Sundin A. Impact of administered amount of peptide on tumor dosimetry at the first cycle of peptide receptor radionuclide therapy (PRRT) in relation to total tumor somatostatin receptor expression. EJNMMI Res 2023; 13:45. [PMID: 37204528 DOI: 10.1186/s13550-023-00997-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 05/08/2023] [Indexed: 05/20/2023] Open
Abstract
BACKGROUND The accumulation of 177Lu-DOTATATE might be influenced by the amount of administered peptide in relation to the tumor somatostatin receptor expression. The effect of the administered peptide mass on the resulting absorbed dose in tumors and normal organs has not previously been assessed in relation to the patients' tumor load. METHOD Patients with small intestinal (n = 141) and pancreatic (n = 62) neuroendocrine tumors (NETs) who underwent PRRT were selected for retrospective evaluation. All patients had received 7.4 GBq 177Lu-DOTATATE, and the amount of administered peptide in the preparation varied from 93 to 456 µg. The absorbed dose in tumors and normal tissue at the first PRRT cycle was calculated, based on SPECT-measurements at day 1, 4, and 7 post-infusion. The total tumor somatostatin receptor expression (tTSSTRE) was calculated on SPECT after 24 h by multiplying the functional tumor volume, delineated by 42% cut-off VOIs of the highest activity, with the SUVmean for the respective tumor VOIs. Spearman's rank correlation analyzed any relationship between the administered amount of peptide and the absorbed dose in tumors and normal organs, in relation to the patients' tTSSTRE. RESULTS There was no correlation between the amount of peptide and any of the tested parameters in relation to tTSSTRE. CONCLUSION In this retrospective analysis, no correlation between the amount of administered peptide in the 177Lu-DOTATATE preparation and the absorbed radiation doses in tumors and normal tissues was demonstrated in relation to the total tumor SSTR expression.
Collapse
Affiliation(s)
- Ulrika Jahn
- Department of Surgical Sciences, Nuclear Medicine and PET, Uppsala University Hospital, 751 85, Uppsala, Sweden.
- Medical Imaging Centre, Uppsala University Hospital, Uppsala, Sweden.
| | - Ulrike Garske-Román
- Department of Blood and Tumor Diseases, Uppsala University Hospital, Uppsala, Sweden
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Mattias Sandström
- Department of Surgical Sciences, Nuclear Medicine and PET, Uppsala University Hospital, 751 85, Uppsala, Sweden
- Department of Medical Physics, Uppsala University Hospital, Uppsala, Sweden
| | - Mark Lubberink
- Department of Surgical Sciences, Nuclear Medicine and PET, Uppsala University Hospital, 751 85, Uppsala, Sweden
- Department of Medical Physics, Uppsala University Hospital, Uppsala, Sweden
| | - Anders Sundin
- Department of Surgical Sciences, Nuclear Medicine and PET, Uppsala University Hospital, 751 85, Uppsala, Sweden
- Medical Imaging Centre, Uppsala University Hospital, Uppsala, Sweden
| |
Collapse
|
14
|
O'Neill E, Mosley M, Cornelissen B. Imaging DNA damage response by γH2AX in vivo predicts treatment response to Lutetium-177 radioligand therapy and suggests senescence as a therapeutically desirable outcome. Theranostics 2023; 13:1302-1310. [PMID: 36923536 PMCID: PMC10008745 DOI: 10.7150/thno.82101] [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: 12/23/2022] [Accepted: 01/19/2023] [Indexed: 03/14/2023] Open
Abstract
Rationale: An effective absorbed dose response relationship is yet to be established for Lutetium-177 based radionuclide therapies such as 177Lu-DOTATATE and 177Lu-PSMA. The inherent biological heterogeneity of neuroendocrine and prostate cancers may make the prospect of establishing cohort-based dose-response relationships unobtainable. Instead, an individual-based approach, monitoring the dose-response within each tumor could provide the necessary metric to monitor treatment efficacy. Methods: We developed a dual isotope SPECT imaging strategy to monitor the change over time in the relationship between 177Lu-DOTATATE and 111In-anti-γH2AX-TAT, a modified radiolabelled antibody that allows imaging of DNA double strand breaks, in mice bearing rat pancreatic cancer xenografts. The dynamics of γH2AX foci, apoptosis and senescence following exposure to 177Lu-DOTATATE was further investigated in vitro and in ex vivo tumor sections. Results: The change in slope of the 111In-anti-γH2AX-TAT to 177Lu signal between days 5 and 7 was found to be highly predictive of survival (r = 0.955, P < 0.0001). This pivotal timeframe was investigated further in vitro: clonogenic survival correlated with the number of γH2AX foci at day 6 (r = -0.995, P < 0.0005). While there was evidence of continuously low levels of apoptosis, delayed induction of senescence in vitro appeared to better account for the γH2AX response to 177Lu. The induction of senescence was further investigated by ex vivo analysis and corresponded with sustained retention of 177Lu within tumor regions. Conclusions: Dual isotope SPECT imaging can provide individualized tumor dose-responses that can be used to predict lutetium-177 treatment efficacy. This bio-dosimeter metric appears to be dependent upon the extent of senescence induction and suggests an integral role that senescence plays in lutetium-177 treatment efficacy.
Collapse
Affiliation(s)
- Edward O'Neill
- MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Michael Mosley
- MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Bart Cornelissen
- MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
- Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
15
|
Correlations between [ 68Ga]Ga-DOTA-TOC Uptake and Absorbed Dose from [ 177Lu]Lu-DOTA-TATE. Cancers (Basel) 2023; 15:cancers15041134. [PMID: 36831477 PMCID: PMC9954147 DOI: 10.3390/cancers15041134] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/01/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
PURPOSE The aim of this paper was to investigate correlations between pre- therapeutic [68Ga]Ga-DOTA-TOC uptake and absorbed dose to tumours from therapy with [177Lu]Lu-DOTA-TATE. METHODS This retrospective study included 301 tumours from 54 GEP-NET patients. The tumours were segmented on pre-therapeutic [68Ga]Ga-DOTA-TOC PET/CT, and post-therapy [177Lu]Lu-DOTA-TATE SPECT/CT images, using a fixed 40% threshold. The SPECT/CT images were used for absorbed dose calculations by assuming a linear build-up from time zero to day one, and mono-exponential wash-out after that. Both SUVmean and SUVmax were measured from the PET images. A linear absorbed-dose prediction model was formed with SUVmean as the independent variable, and the accuracy was tested with a split 70-30 training-test set. RESULTS Mean SUVmean and SUVmax from [68Ga]Ga-DOTA-TOC PET was 24.0 (3.6-84.4) and 41.0 (6.7-146.5), and the mean absorbed dose from [177Lu]Lu-DOTA-TATE was 26.9 Gy (2.4-101.9). A linear relationship between SUVmean and [177Lu]Lu-DOTA-TATE activity concentration at 24 h post injection was found (R2 = 0.44, p < 0.05). In the prediction model, a root mean squared error and a mean absolute error of 1.77 and 1.33 Gy/GBq, respectively, were found for the test set. CONCLUSIONS There was a high inter- and intra-patient variability in tumour measurements, both for [68Ga]Ga-DOTA-TOC SUVs and absorbed doses from [177Lu]Lu-DOTA-TATE. Depending on the required accuracy, [68Ga]Ga-DOTA-TOC PET imaging may estimate the [177Lu]Lu-DOTA-TATE uptake. However, there could be a high variance between predicted and actual absorbed doses.
Collapse
|
16
|
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
|
17
|
Stenvall A, Gustafsson J, Larsson E, Roth D, Sundlöv A, Jönsson L, Hindorf C, Ohlsson T, Sjögreen Gleisner K. Relationships between uptake of [ 68Ga]Ga-DOTA-TATE and absorbed dose in [ 177Lu]Lu-DOTA-TATE therapy. EJNMMI Res 2022; 12:75. [PMID: 36534192 PMCID: PMC9763525 DOI: 10.1186/s13550-022-00947-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Somatostatin receptor 68Ga PET imaging is standard for evaluation of a patient's suitability for 177Lu peptide receptor radionuclide therapy of neuroendocrine tumours (NETs). The 68Ga PET serves to ensure sufficient somatostatin receptor expression, commonly evaluated qualitatively. The aim of this study is to investigate the quantitative relationships between uptake in 68Ga PET and absorbed doses in 177Lu therapy. METHOD Eighteen patients underwent [68Ga]Ga-DOTA-TATE PET imaging within 20 weeks prior to their first cycle of [177Lu]Lu-DOTA-TATE. Absorbed doses for therapy were estimated for tumours, kidney, spleen, and normal liver parenchyma using a hybrid SPECT/CT-planar method. Gallium-68 activity concentrations were retrieved from PET images and also used to calculate SUVs and normalized SUVs, using blood and tissue for normalization. The 68Ga activity concentrations per injected activity, SUVs, and normalized SUVs were compared with 177Lu activity concentrations 1 d post-injection and 177Lu absorbed doses. For tumours, for which there was a variable number per patient, both inter- and intra-patient correlations were analysed. Furthermore, the prediction of 177Lu tumour absorbed doses based on a combination of tumour-specific 68Ga activity concentrations and group-based estimates of the effective half-lives for grade 1 and 2 NETs was explored. RESULTS For normal organs, only spleen showed a significant correlation between the 68Ga activity concentration and 177Lu absorbed dose (r = 0.6). For tumours, significant, but moderate, correlations were obtained, with respect to both inter-patient (r = 0.7) and intra-patient (r = 0.45) analyses. The correlations to absorbed doses did not improve when using 68Ga SUVs or normalized SUVs. The relationship between activity uptakes for 68Ga PET and 177Lu SPECT was stronger, with correlation coefficients r = 0.8 for both inter- and intra-patient analyses. The 177Lu absorbed dose to tumour could be predicted from the 68Ga activity concentrations with a 95% coverage interval of - 65% to 248%. CONCLUSIONS On a group level, a high uptake of [68Ga]Ga-DOTA-TATE is associated with high absorbed doses at 177Lu-DOTA-TATE therapy, but the relationship has a limited potential with respect to individual absorbed dose planning. Using SUV or SUV normalized to reference tissues do not improve correlations compared with using activity concentration per injected activity.
Collapse
Affiliation(s)
- Anna Stenvall
- grid.4514.40000 0001 0930 2361Medical Radiation Physics, Lund, Lund University, Lund, Sweden ,grid.411843.b0000 0004 0623 9987Radiation Physics, Skåne University Hospital, Lund, Sweden
| | - Johan Gustafsson
- grid.4514.40000 0001 0930 2361Medical Radiation Physics, Lund, Lund University, Lund, Sweden
| | - Erik Larsson
- grid.411843.b0000 0004 0623 9987Radiation Physics, Skåne University Hospital, Lund, Sweden
| | - Daniel Roth
- grid.4514.40000 0001 0930 2361Medical Radiation Physics, Lund, Lund University, Lund, Sweden
| | - Anna Sundlöv
- grid.4514.40000 0001 0930 2361Division of Oncology, Department of Clinical Sciences, Lund, Lund University, Lund, Sweden
| | - Lena Jönsson
- grid.4514.40000 0001 0930 2361Medical Radiation Physics, Lund, Lund University, Lund, Sweden ,grid.411843.b0000 0004 0623 9987Radiation Physics, Skåne University Hospital, Lund, Sweden
| | - Cecilia Hindorf
- grid.4514.40000 0001 0930 2361Medical Radiation Physics, Lund, Lund University, Lund, Sweden ,grid.24381.3c0000 0000 9241 5705Department of Radiation Physics and Nuclear Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Tomas Ohlsson
- grid.411843.b0000 0004 0623 9987Radiation Physics, Skåne University Hospital, Lund, Sweden
| | | |
Collapse
|
18
|
Zwirtz K, Hardt J, Acker G, Baur ADJ, Pavel M, Huang K, Brenner W, Prasad V. Comparison of Choi, RECIST and Somatostatin Receptor PET/CT Based Criteria for the Evaluation of Response and Response Prediction to PRRT. Pharmaceutics 2022; 14:1278. [PMID: 35745849 PMCID: PMC9227135 DOI: 10.3390/pharmaceutics14061278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/07/2022] [Accepted: 06/09/2022] [Indexed: 02/01/2023] Open
Abstract
Aim: The most suitable method for assessment of response to peptide receptor radionuclide therapy (PRRT) of neuroendocrine tumors (NET) is still under debate. In this study we aimed to compare size (RECIST 1.1), density (Choi), Standardized Uptake Value (SUV) and a newly defined ZP combined parameter derived from Somatostatin Receptor (SSR) PET/CT for prediction of both response to PRRT and overall survival (OS). Material and Methods: Thirty-four NET patients with progressive disease (F:M 23:11; mean age 61.2 y; SD ± 12) treated with PRRT using either Lu-177 DOTATOC or Lu-177 DOTATATE and imaged with Ga-68 SSR PET/CT approximately 10-12 weeks prior to and after each treatment cycle were retrospectively analyzed. Median duration of follow-up after the first cycle was 63.9 months (range 6.2-86.2). A total of 77 lesions (2-8 per patient) were analyzed. Response assessment was performed according to RECIST 1.1, Choi and modified EORTC (MORE) criteria. In addition, a new parameter named ZP, the product of Hounsfield unit (HU) and SUVmean (Standard Uptake Value) of a tumor lesion, was tested. Further, SUV values (max and mean) of the tumor were normalized to SUV of normal liver parenchyma. Tumor response was defined as CR, PR, or SD. Gold standard for comparison of baseline parameters for prediction of response of individual target lesions to PRRT was change in size of lesions according to RECIST 1.1. For prediction of overall survival, the response after the first and second PRRT were tested. Results: Based on RECIST 1.1, Choi, MORE, and ZP, 85.3%, 64.7%, 61.8%, and 70.6% achieved a response whereas 14.7%, 35.3%, 38.2%, and 29.4% demonstrated PD (progressive disease), respectively. Baseline ZP and ZPnormalized were found to be the only parameters predictive of lesion progression after three PRRT cycles (AUC ZP 0.753; 95% CI 0.6-0.9, p 0.037; AUC ZPnormalized 0.766; 95% CI 0.6-0.9; p 0.029). Based on a cut-off-value of 1201, ZP achieved a sensitivity of 86% and a specificity of 67%, while ZPnormalized reached a sensitivity of 86% and a specificity of 76% at a cut-off-value of 198. Median OS in the total cohort was not reached. In univariate analysis amongst all parameters, only patients having progressive disease according to MORE after the second cycle of PRRT were found to have significantly shorter overall survival (median OS in objective responders not reached, in PD 29.2 months; p 0.015). Patients progressive after two cycles of PRRT according to ZP had shorter OS compared to those responding (median OS for responders not reached, for PD 47.2 months, p 0.066). Conclusions: In this explorative study, we showed that Choi, RECIST 1.1, and SUVmax-based response evaluation varied significantly from each other. Only patients showing progressive disease after two PRRT cycles according to MORE criteria had a worse prognosis while baseline ZP and ZPnormalized performed best in predicting lesion progression after three cycles of PRRT.
Collapse
Affiliation(s)
- Kevin Zwirtz
- Department of Nuclear Medicine, Charité Universitätsmedizin Berlin, 13353 Berlin, Germany; (K.Z.); (K.H.); (W.B.)
| | - Juliane Hardt
- Institute of Biometry and Clinical Epidemiology, Charité Universitätsmedizin Berlin, 13353 Berlin, Germany;
- Department of Biometry, Epidemiology and Information Processing, WHO Collaborating Centre for Research and Training for Health in the Human-Animal-Environment Interface, University of Veterinary Medicine (Foundation) Hannover (TiHo), 30559 Hannover, Germany
- Medical Information Management, Faculty of Information and Communication, University of Applied Sciences Hannover, 30459 Hannover, Germany
| | - Güliz Acker
- Department of Neurosurgery, Charité Universitätsmedizin Berlin, 13353 Berlin, Germany;
| | - Alexander D. J. Baur
- Department of Radiology, Charité Universitätsmedizin Berlin, 13353 Berlin, Germany;
| | - Marianne Pavel
- Department of Hepatology and Gastroenterology, Charité Universitätsmedizin Berlin, 13353 Berlin, Germany;
- Department of Endocrinology, University Hospital of Erlangen, 91054 Erlangen, Germany
| | - Kai Huang
- Department of Nuclear Medicine, Charité Universitätsmedizin Berlin, 13353 Berlin, Germany; (K.Z.); (K.H.); (W.B.)
| | - Winfried Brenner
- Department of Nuclear Medicine, Charité Universitätsmedizin Berlin, 13353 Berlin, Germany; (K.Z.); (K.H.); (W.B.)
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Vikas Prasad
- Department of Nuclear Medicine, Charité Universitätsmedizin Berlin, 13353 Berlin, Germany; (K.Z.); (K.H.); (W.B.)
- Department of Nuclear Medicine, University Ulm, 89081 Ulm, Germany
- International Centers for Precision Oncology Academy, 65189 Wiesbaden, Germany
| |
Collapse
|
19
|
Puliani G, Chiefari A, Mormando M, Bianchini M, Lauretta R, Appetecchia M. New Insights in PRRT: Lessons From 2021. Front Endocrinol (Lausanne) 2022; 13:861434. [PMID: 35450421 PMCID: PMC9016202 DOI: 10.3389/fendo.2022.861434] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/07/2022] [Indexed: 11/16/2022] Open
Abstract
Peptide receptor radionuclide therapy (PRRT) using radiolabeled somatostatin analogs has been used for over two decades for the treatment of well-differentiated neuroendocrine tumors (NETs), and the publication of the NETTER-1 trials has further strengthened its clinical use. However, many aspects of this treatment are still under discussion. The purpose of this review is to collect and discuss the new available evidence, published in 2021, on the use of 177Lu-Oxodotreotide (DOTATATE) or 90Y-Edotreotide (DOTATOC) in adult patients with NETs focusing on the following hot topics: 1) PRRT use in new clinical settings, broaden its indications; 2) the short- and long-term safety; and 3) the identification of prognostic and predictive factors. The review suggests a possible future increase of PRRT applications, using it in other NETs, as a neoadjuvant treatment, or for rechallenge. Regarding safety, available studies, even those with long follow-up, supported the low rates of adverse events, even though 1.8% of treated patients developed a second malignancy. Finally, there is a lack of prognostic and predictive factors for PRRT, with the exception of the crucial role of nuclear imaging for both patient selection and treatment response estimation.
Collapse
Affiliation(s)
- Giulia Puliani
- Oncological Endocrinology Unit, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) Regina Elena National Cancer Institute, Rome, Italy
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Alfonsina Chiefari
- Oncological Endocrinology Unit, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) Regina Elena National Cancer Institute, Rome, Italy
| | - Marilda Mormando
- Oncological Endocrinology Unit, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) Regina Elena National Cancer Institute, Rome, Italy
| | - Marta Bianchini
- Oncological Endocrinology Unit, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) Regina Elena National Cancer Institute, Rome, Italy
| | - Rosa Lauretta
- Oncological Endocrinology Unit, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) Regina Elena National Cancer Institute, Rome, Italy
| | - Marialuisa Appetecchia
- Oncological Endocrinology Unit, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) Regina Elena National Cancer Institute, Rome, Italy
| |
Collapse
|
20
|
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
|
21
|
Danieli R, Milano A, Gallo S, Veronese I, Lascialfari A, Indovina L, Botta F, Ferrari M, Cicchetti A, Raspanti D, Cremonesi M. Personalized Dosimetry in Targeted Radiation Therapy: A Look to Methods, Tools and Critical Aspects. J Pers Med 2022; 12:205. [PMID: 35207693 PMCID: PMC8874397 DOI: 10.3390/jpm12020205] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/13/2022] [Accepted: 01/14/2022] [Indexed: 12/10/2022] Open
Abstract
Targeted radiation therapy (TRT) is a strategy increasingly adopted for the treatment of different types of cancer. The urge for optimization, as stated by the European Council Directive (2013/59/EURATOM), requires the implementation of a personalized dosimetric approach, similar to what already happens in external beam radiation therapy (EBRT). The purpose of this paper is to provide a thorough introduction to the field of personalized dosimetry in TRT, explaining its rationale in the context of optimization and describing the currently available methodologies. After listing the main therapies currently employed, the clinical workflow for the absorbed dose calculation is described, based on works of the most experienced authors in the literature and recent guidelines. Moreover, the widespread software packages for internal dosimetry are presented and critical aspects discussed. Overall, a selection of the most important and recent articles about this topic is provided.
Collapse
Affiliation(s)
- Rachele Danieli
- Dipartimento di Fisica, Università degli Studi di Pavia, Via Bassi 6, 27100 Pavia, Italy;
| | - Alessia Milano
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo F. Vito 1, 00168 Roma, Italy;
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Roma, Italy
| | - Salvatore Gallo
- Dipartimento di Fisica “Aldo Pontremoli”, Università degli Studi di Milano, Via Celoria 16, 20133 Milano, Italy; (S.G.); (I.V.)
- INFN Sezione di Milano, Via Celoria 16, 20133 Milano, Italy
| | - Ivan Veronese
- Dipartimento di Fisica “Aldo Pontremoli”, Università degli Studi di Milano, Via Celoria 16, 20133 Milano, Italy; (S.G.); (I.V.)
- INFN Sezione di Milano, Via Celoria 16, 20133 Milano, Italy
| | - Alessandro Lascialfari
- INFN-Pavia Unit, Department of Physics, University of Pavia, Via Bassi 6, 27100 Pavia, Italy;
| | - Luca Indovina
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo F. Vito 1, 00168 Roma, Italy;
| | - Francesca Botta
- Medical Physics Unit, European Institute of Oncology IRCCS, Via Giuseppe Ripamonti 435, 20141 Milano, Italy; (F.B.); (M.F.)
| | - Mahila Ferrari
- Medical Physics Unit, European Institute of Oncology IRCCS, Via Giuseppe Ripamonti 435, 20141 Milano, Italy; (F.B.); (M.F.)
| | - Alessandro Cicchetti
- Prostate Cancer Program, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian, 1, 20133 Milano, Italy;
| | - Davide Raspanti
- Temasinergie S.p.A., Via Marcello Malpighi 120, 48018 Faenza, Italy;
| | - Marta Cremonesi
- Radiation Research Unit, European Institute of Oncology IRCCS, Via Giuseppe Ripamonti 435, 20141 Milano, Italy;
| |
Collapse
|
22
|
Dosimetry in radionuclide therapy: the clinical role of measuring radiation dose. Lancet Oncol 2022; 23:e75-e87. [DOI: 10.1016/s1470-2045(21)00657-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 11/03/2021] [Accepted: 11/05/2021] [Indexed: 12/22/2022]
|
23
|
Kim KM, Lee MS, Suh MS, Selvam HSMS, Tan TH, Cheon GJ, Kang KW, Lee JS. Comparison of Voxel S-Value Methods for Personalized Voxel-based Dosimetry of 177 Lu-DOTATATE. Med Phys 2022; 49:1888-1901. [PMID: 35014699 DOI: 10.1002/mp.15444] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 01/11/2021] [Accepted: 01/03/2022] [Indexed: 11/09/2022] Open
Abstract
PURPOSE Voxel-based dosimetry is potentially accurate than organ-based dosimetry because it considers the anatomical variations in each individual and the heterogeneous radioactivity distribution in each organ. Here, voxel-based dosimetry for 177 Lu-DOTATATE therapy was performed using single and multiple voxel S-value (VSV) methods and compared with Monte Carlo simulations. To verify these methods, we adopted sequential 177 Lu-DOTATATE SPECT/CT dataset acquired from Sunway Medical Centre using the major vendor's SPECT/CT scanner (Siemens) METHODS: The administered activity of 177 Lu-DOTATATE was 7.99 ± 0.36 GBq. SPECT/CT images were acquired 0.5, 4, 24, and 48 h after injection in Sunway Medical Centre. For the multiple VSV method VSV kernels of 177 Lu in media with various densities were generated by GATE simulation first. The second step involved the convolution of the time-integrated activity map with each kernel to produce medium-specific dose maps. Third, each medium-specific dose map was masked using binary medium masks, which were generated from CT-based density maps. Finally, all masked dose maps were summed to generate the final dose map. VSV methods with four different VSV sets (1, 4, 10, and 20 VSVs) were compared. Voxel-wise density correction for the single VSV method was also performed. The absorbed doses in the kidneys, bone marrow, and tumors were analyzed, and the relative errors between the VSV and Monte Carlo simulation approaches were estimated. Organ-based dosimetry using OLINDA/EXM was also compared RESULTS: The accuracy of the multiple VSV approach increased with the number of dose kernels. The average dose estimation errors of a single VSV with density correction and 20 VSVs were less than 6% in most cases, although organ-based dosimetry using OLINDA/EXM yielded an error of up to 123%. The advantages of the single VSV method with density correction and the 20 VSVs over organ-based dosimetry were most evident in bone marrow and bone-metastatic tumors with heterogeneous medium properties. CONCLUSION The single VSV method with density correction and multiple VSV method with 20 dose kernels enabled fast and accurate radiation dose estimation. Accordingly, voxel-based dosimetry methods can be useful for managing administration activity and for investigating tumor dose responses to further increase the therapeutic efficacy of 177 Lu-DOTATATE. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Keon Min Kim
- Interdisciplinary Program in Bioengineering, Seoul National University Graduate School, Seoul, 03080, Korea.,Integrated Major in Innovative Medical Science, Seoul National University Graduate School, Seoul, 03080, Korea
| | - Min Sun Lee
- Korea Atomic Energy Research Institute, Daejeon, 34057, Korea
| | - Min Seok Suh
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, 03080, Korea.,Institute of Radiation Medicine, Medical Research Center, Seoul National University, Seoul, 03080, Korea
| | | | - Teik Hin Tan
- Nuclear Medicine Centre, Sunway Medical Centre, Selangor, 47500, csMalaysia
| | - Gi Jeong Cheon
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, 03080, Korea.,Institute of Radiation Medicine, Medical Research Center, Seoul National University, Seoul, 03080, Korea
| | - Keon Wook Kang
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, 03080, Korea.,Institute of Radiation Medicine, Medical Research Center, Seoul National University, Seoul, 03080, Korea.,Bio-MAX Institute, Seoul National University, Seoul, 08826, Korea
| | - Jae Sung Lee
- Interdisciplinary Program in Bioengineering, Seoul National University Graduate School, Seoul, 03080, Korea.,Integrated Major in Innovative Medical Science, Seoul National University Graduate School, Seoul, 03080, Korea.,Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, 03080, Korea.,Institute of Radiation Medicine, Medical Research Center, Seoul National University, Seoul, 03080, Korea
| |
Collapse
|
24
|
Stolniceanu CR, Moscalu M, Azoicai D, Tamba B, Volovat C, Grierosu I, Ionescu T, Jalloul W, Ghizdovat V, Gherasim R, Volovat S, Wang F, Fu J, Moscalu R, Matovic M, Stefanescu C. Improved Personalised Neuroendocrine Tumours' Diagnosis Predictive Power by New Receptor Somatostatin Image Processing Quantification. J Pers Med 2021; 11:jpm11101042. [PMID: 34683183 PMCID: PMC8539645 DOI: 10.3390/jpm11101042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 09/28/2021] [Accepted: 10/13/2021] [Indexed: 11/24/2022] Open
Abstract
Although neuroendocrine tumours (NETs) are intensively studied, their diagnosis and consequently personalised therapy management is still puzzling due to their tumoral heterogeneity. In their theragnosis algorithm, receptor somatostatin scintigraphy takes the central place, the diagnosis receptor somatostatin analogue (RSA) choice depending on laboratory experience and accessibility. However, in all cases, the results depend decisively on correct radiotracer tumoral uptake quantification, where unfortunately there are still unrevealed clues and lack of standardization. We propose an improved method to quantify the biodistribution of gamma-emitting RSA, using tissular corrected uptake indices. We conducted a bi-centric retrospective study on 101 patients with different types of NETs. Three uptake indices obtained after applying new corrections to areas of interest drawn for the tumour and for three reference organs (liver, spleen and lung) were statistically analysed. For the corrected pathological uptake indices, the results showed a significant decrease in the error of estimating the occurrence of errors and an increase in the diagnostic predictive power for NETs, especially in the case of lung-referring corrected index. In conclusion, these results support the importance of corrected uptake indices use in the analysis of 99mTcRSA biodistribution for a better personalised diagnostic accuracy of NETs patients.
Collapse
Affiliation(s)
- Cati Raluca Stolniceanu
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (C.R.S.); (I.G.); (T.I.); (W.J.); (V.G.); (R.G.); (C.S.)
| | - Mihaela Moscalu
- Department of Preventive Medicine and Interdisciplinarity, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Correspondence:
| | - Doina Azoicai
- Department of Epidemiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Bogdan Tamba
- Advanced Center for Research and Development in Experimental Medicine (CEMEX), “Grigore T. Popa” University of Medicine and Pharmacy, 700454 Iasi, Romania;
| | - Constantin Volovat
- Department of Medicine III—Medical Oncology-Radiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (C.V.); (S.V.)
| | - Irena Grierosu
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (C.R.S.); (I.G.); (T.I.); (W.J.); (V.G.); (R.G.); (C.S.)
| | - Teodor Ionescu
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (C.R.S.); (I.G.); (T.I.); (W.J.); (V.G.); (R.G.); (C.S.)
| | - Wael Jalloul
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (C.R.S.); (I.G.); (T.I.); (W.J.); (V.G.); (R.G.); (C.S.)
| | - Vlad Ghizdovat
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (C.R.S.); (I.G.); (T.I.); (W.J.); (V.G.); (R.G.); (C.S.)
| | - Roxana Gherasim
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (C.R.S.); (I.G.); (T.I.); (W.J.); (V.G.); (R.G.); (C.S.)
| | - Simona Volovat
- Department of Medicine III—Medical Oncology-Radiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (C.V.); (S.V.)
| | - Feng Wang
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; (F.W.); (J.F.)
| | - Jingjing Fu
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; (F.W.); (J.F.)
| | - Roxana Moscalu
- Manchester Academic Health Science Centre, School of Medical Sciences Manchester, The University of Manchester, Manchester M139PT, UK;
| | - Milovan Matovic
- Clinical Center Kragujevac, Center for Nuclear Medicine, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Cipriana Stefanescu
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (C.R.S.); (I.G.); (T.I.); (W.J.); (V.G.); (R.G.); (C.S.)
| |
Collapse
|
25
|
Wang K, Tepper JE. Radiation therapy-associated toxicity: Etiology, management, and prevention. CA Cancer J Clin 2021; 71:437-454. [PMID: 34255347 DOI: 10.3322/caac.21689] [Citation(s) in RCA: 149] [Impact Index Per Article: 49.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/25/2021] [Accepted: 05/27/2021] [Indexed: 12/25/2022] Open
Abstract
Radiation therapy (RT) is a curative treatment for many malignancies and provides effective palliation in patients with tumor-related symptoms. However, the biophysical effects of RT are not specific to tumor cells and may produce toxicity due to exposure of surrounding organs and tissues. In this article, the authors review the clinical context, pathophysiology, risk factors, presentation, and management of RT side effects in each human organ system. Ionizing radiation works by producing DNA damage leading to tumor death, but effects on normal tissue may result in acute and/or late toxicity. The manifestation of toxicity depends on both cellular characteristics and affected organs' anatomy and physiology. There is usually a direct relationship between the radiation dose and volume to normal tissues and the risk of toxicity, which has led to guidelines and recommended dose limits for most tissues. Side effects are multifactorial, with contributions from baseline patient characteristics and other oncologic treatments. Technological advances in recent decades have decreased RT toxicity by dramatically improving the ability to deliver RT that maximizes tumor dose and minimizes organ dose. Thus the study of RT-associated toxicity is a complex, core component of radiation oncology training that continues to evolve alongside advances in cancer management. Because RT is used in up to one-half of all patients with cancer, an understanding of its acute and late effects in different organ systems is clinically pertinent to both oncologists and nononcologists.
Collapse
Affiliation(s)
- Kyle Wang
- Department of Radiation Oncology, University of Cincinnati, Cincinnati, Ohio
| | - Joel E Tepper
- Department of Radiation Oncology, University of North Carolina, Chapel Hill, North Carolina
| |
Collapse
|
26
|
Roth D, Gustafsson JR, Warfvinge CF, Sundlöv A, Åkesson A, Tennvall J, Sjögreen Gleisner K. Dosimetric quantities of neuroendocrine tumors over treatment cycles with 177Lu-DOTA-TATE. J Nucl Med 2021; 63:399-405. [PMID: 34272319 DOI: 10.2967/jnumed.121.262069] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 05/25/2021] [Indexed: 11/16/2022] Open
Abstract
Tumor dosimetry was performed for 177Lu-DOTA-TATE with the aims of better understanding i) the range and variation of the tumor absorbed doses (ADs), ii) how different dosimetric quantities evolve over the treatment cycles, and iii) whether this evolution differs depending on the tumor grade. Such information is important for radiobiological interpretation and may inform the design of alternative administration schemes. Methods: Data come from 41 patients with neuroendocrine tumors (NETs) of grade 1 (n = 23) or 2 (n = 18), that had received between 2 and 9 treatment cycles. Dosimetry was performed for 182 individual lesions, giving in total 880 individual AD assessments across all cycles. Hybrid planar-SPECT/CT imaging was used, including quantitative SPECT reconstruction, voxel-based absorbed-dose-rate calculation, semi-automatic image segmentation, and partial-volume correction. Linear mixed-effect models were used to analyze changes over cycles in tumor ADs, absorbed-dose rates and activity concentrations at day-1, effective half-times, and tumor volumes. Tumors smaller than 8 ml were excluded from analyses. Results: Tumor ADs ranged between 2 and 77 Gy per cycle. On average the AD decreased over the cycles, with significantly different rates (P < 0.05) for grade 1 and 2 NETs of 6% and 14% per cycle, respectively. The absorbed-dose rates and activity concentrations at day-1 decreased by similar amounts. The effective half-times were less variable but shorter for grade 2 than grade 1 (P < 0.001). For grade 2 NETS the tumor volumes decreased, with a similar tendency in grade 1. Conclusion: The tumor AD, absorbed-dose rate and activity uptake decrease, in parallel with tumor volumes, between 177Lu-DOTA-TATE treatment cycles, particularly for grade 2 NETs. The effective half-times vary less but are lower for grade 2 than grade 1 NETs. These results may indicate the development of radiation-induced fibrosis and could have implications for the design of future treatment and dosimetry protocols.
Collapse
Affiliation(s)
- Daniel Roth
- Department of Medical Radiation Physics, Clinical Sciences Lund, Lund University, Sweden
| | - Johan Ruben Gustafsson
- Department of Medical Radiation Physics, Clinical Sciences Lund, Lund University, Sweden
| | - Carl Fredrik Warfvinge
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Lund University, Sweden
| | - Anna Sundlöv
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Lund University, Sweden
| | - Anna Åkesson
- Clinical Studies Sweden - Forum South, Skåne University Hospital
| | - Jan Tennvall
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Lund University, Sweden
| | | |
Collapse
|