1
|
Gawel J, Rogulski Z. The Challenge of Single-Photon Emission Computed Tomography Image Segmentation in the Internal Dosimetry of 177Lu Molecular Therapies. J Imaging 2024; 10:27. [PMID: 38276319 PMCID: PMC10817423 DOI: 10.3390/jimaging10010027] [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: 11/27/2023] [Revised: 01/03/2024] [Accepted: 01/05/2024] [Indexed: 01/27/2024] Open
Abstract
The aim of this article is to review the single photon emission computed tomography (SPECT) segmentation methods used in patient-specific dosimetry of 177Lu molecular therapy. Notably, 177Lu-labelled radiopharmaceuticals are currently used in molecular therapy of metastatic neuroendocrine tumours (ligands for somatostatin receptors) and metastatic prostate adenocarcinomas (PSMA ligands). The proper segmentation of the organs at risk and tumours in targeted radionuclide therapy is an important part of the optimisation process of internal patient dosimetry in this kind of therapy. Because this is the first step in dosimetry assessments, on which further dose calculations are based, it is important to know the level of uncertainty that is associated with this part of the analysis. However, the robust quantification of SPECT images, which would ensure accurate dosimetry assessments, is very hard to achieve due to the intrinsic features of this device. In this article, papers on this topic were collected and reviewed to weigh up the advantages and disadvantages of the segmentation methods used in clinical practice. Degrading factors of SPECT images were also studied to assess their impact on the quantification of 177Lu therapy images. Our review of the recent literature gives an insight into this important topic. However, based on the PubMed and IEEE databases, only a few papers investigating segmentation methods in 177Lumolecular therapy were found. Although segmentation is an important step in internal dose calculations, this subject has been relatively lightly investigated for SPECT systems. This is mostly due to the inner features of SPECT. What is more, even when studies are conducted, they usually utilise the diagnostic radionuclide 99mTc and not a therapeutic one like 177Lu, which could be of concern regarding SPECT camera performance and its overall outcome on dosimetry.
Collapse
Affiliation(s)
- Joanna Gawel
- Faculty of Chemistry, University of Warsaw, 02-093 Warsaw, Poland
| | | |
Collapse
|
2
|
Katugampola S, Hobbs RF, Howell RW. Generalized methods for predicting biological response to mixed radiation types and calculating equieffective doses (EQDX). Med Phys 2024; 51:637-649. [PMID: 37558637 PMCID: PMC11330299 DOI: 10.1002/mp.16650] [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: 03/23/2023] [Revised: 06/23/2023] [Accepted: 07/10/2023] [Indexed: 08/11/2023] Open
Abstract
BACKGROUND Predicting biological responses to mixed radiation types is of considerable importance when combining radiation therapies that use multiple radiation types and delivery regimens. These may include the use of both low- and high-linear energy transfer (LET) radiations. A number of theoretical models have been developed to address this issue. However, model predictions do not consistently match published experimental data for mixed radiation exposures. Furthermore, the models are often computationally intensive. Accordingly, there is a need for efficient analytical models that can predict responses to mixtures of low- and high-LET radiations. Additionally, a general formalism to calculate equieffective dose (EQDX) for mixed radiations is needed. PURPOSE To develop a computationally efficient analytical model that can predict responses to complex mixtures of low- and high-LET radiations as a function of either absorbed dose or EQDX. METHODS The Zaider-Rossi model (ZRM) was modified by replacing the geometric mean of the quadratic coefficients in the interaction term with the arithmetic mean. This modified ZRM model (mZRM) was then further generalized to any number of radiation types and its validity was tested against published experimental observations. Comparisons between the predictions of the ZRM and mZRM, and other models, were made using two and three radiation types. In addition, a generalized formalism for calculating EQDX for mixed radiations was developed within the context of mZRM and validated with published experimental results. RESULTS The predictions of biological responses to mixed-LET radiations calculated with the mZRM are in better agreement with experimental observations than ZRM, especially when high- and low-LET radiations are mixed. In these situations, the ZRM overestimated the surviving fraction. Furthermore, the EQDX calculated with mZRM are in better agreement with experimental observations. CONCLUSION The mZRM is a computationally efficient model that can be used to predict biological response to mixed radiations that have low- and high-LET characteristics. Importantly, interaction terms are retained in the calculation of EQDX for mixed radiation exposures within the mZRM framework. The mZRM has application in a wide range of radiation therapies, including radiopharmaceutical therapy.
Collapse
Affiliation(s)
- Sumudu Katugampola
- Department of Radiology and Center for Cell Signaling, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | - Robert F Hobbs
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Roger W Howell
- Department of Radiology and Center for Cell Signaling, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| |
Collapse
|
3
|
Grkovski M, O'Donoghue JA, Imber BS, Andl G, Tu C, Lafontaine D, Schwartz J, Thor M, Zelefsky MJ, Humm JL, Bodei L. Lesion Dosimetry for [ 177Lu]Lu-PSMA-617 Radiopharmaceutical Therapy Combined with Stereotactic Body Radiotherapy in Patients with Oligometastatic Castration-Sensitive Prostate Cancer. J Nucl Med 2023; 64:1779-1787. [PMID: 37652541 PMCID: PMC10626375 DOI: 10.2967/jnumed.123.265763] [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: 03/21/2023] [Revised: 07/11/2023] [Indexed: 09/02/2023] Open
Abstract
A single-institution prospective pilot clinical trial was performed to demonstrate the feasibility of combining [177Lu]Lu-PSMA-617 radiopharmaceutical therapy (RPT) with stereotactic body radiotherapy (SBRT) for the treatment of oligometastatic castration-sensitive prostate cancer. Methods: Six patients with 9 prostate-specific membrane antigen (PSMA)-positive oligometastases received 2 cycles of [177Lu]Lu-PSMA-617 RPT followed by SBRT. After the first intravenous infusion of [177Lu]Lu-PSMA-617 (7.46 ± 0.15 GBq), patients underwent SPECT/CT at 3.2 ± 0.5, 23.9 ± 0.4, and 87.4 ± 12.0 h. Voxel-based dosimetry was performed with calibration factors (11.7 counts per second/MBq) and recovery coefficients derived from in-house phantom experiments. Lesions were segmented on baseline PSMA PET/CT (50% SUVmax). After a second cycle of [177Lu]Lu-PSMA-617 (44 ± 3 d; 7.50 ± 0.10 GBq) and an interim PSMA PET/CT scan, SBRT (27 Gy in 3 fractions) was delivered to all PSMA-avid oligometastatic sites, followed by post-PSMA PET/CT. RPT and SBRT voxelwise dose maps were scaled (α/β = 3 Gy; repair half-time, 1.5 h) to calculate the biologically effective dose (BED). Results: All patients completed the combination therapy without complications. No grade 3+ toxicities were noted. The median of the lesion SUVmax as measured on PSMA PET was 16.8 (interquartile range [IQR], 11.6) (baseline), 6.2 (IQR, 2.7) (interim), and 2.9 (IQR, 1.4) (post). PET-derived lesion volumes were 0.4-1.7 cm3 The median lesion-absorbed dose (AD) from the first cycle of [177Lu]Lu-PSMA-617 RPT (ADRPT) was 27.7 Gy (range, 8.3-58.2 Gy; corresponding to 3.7 Gy/GBq, range, 1.1-7.7 Gy/GBq), whereas the median lesion AD from SBRT was 28.1 Gy (range, 26.7-28.8 Gy). Spearman rank correlation, ρ, was 0.90 between the baseline lesion PET SUVmax and SPECT SUVmax (P = 0.005), 0.74 (P = 0.046) between the baseline PET SUVmax and the lesion ADRPT, and -0.81 (P = 0.022) between the lesion ADRPT and the percent change in PET SUVmax (baseline to interim). The median for the lesion BED from RPT and SBRT was 159 Gy (range, 124-219 Gy). ρ between the BED from RPT and SBRT and the percent change in PET SUVmax (baseline to post) was -0.88 (P = 0.007). Two cycles of [177Lu]Lu-PSMA-617 RPT contributed approximately 40% to the maximum BED from RPT and SBRT. Conclusion: Lesional dosimetry in patients with oligometastatic castration-sensitive prostate cancer undergoing [177Lu]Lu-PSMA-617 RPT followed by SBRT is feasible. Combined RPT and SBRT may provide an efficient method to maximize the delivery of meaningful doses to oligometastatic disease while addressing potential microscopic disease reservoirs and limiting the dose exposure to normal tissues.
Collapse
Affiliation(s)
- Milan Grkovski
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York;
| | - Joseph A O'Donoghue
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Brandon S Imber
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - George Andl
- Varian Medical Systems Inc., Palo Alto, California; and
| | - Cheng Tu
- Varian Medical Systems Inc., Palo Alto, California; and
| | - Daniel Lafontaine
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jazmin Schwartz
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Maria Thor
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michael J Zelefsky
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - John L Humm
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Lisa Bodei
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
4
|
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
|
5
|
Resch S, Takayama Fouladgar S, Zacherl M, Sheikh GT, Liubchenko G, Rumiantcev M, Unterrainer LM, Wenter V, Bartenstein P, Ziegler SI, Ilhan H, Beyer L, Böning G, Delker A. Investigation of image-based lesion and kidney dosimetry protocols for 177Lu-PSMA-I&T therapy with and without a late SPECT/CT acquisition. EJNMMI Phys 2023; 10:11. [PMID: 36757516 PMCID: PMC9911578 DOI: 10.1186/s40658-023-00529-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 01/26/2023] [Indexed: 02/10/2023] Open
Abstract
BACKGROUND 177Lu-PSMA therapy has been successfully used to prolong the survival of patients with metastatic castration-resistant prostate cancer. Patient-specific dosimetry based on serial quantitative SPECT/CT imaging can support the understanding of dose-effect relationships. However, multiple SPECT/CT measurements can be challenging for patients, which motivates the investigation of efficient sampling schedules and their impact on dosimetry. In this study, different time samplings with respect to the number and timing of SPECT/CT acquisitions with and without a late measurement were investigated. MATERIALS AND METHODS In total, 43 lesions and 10 kidneys of 5 patients receiving 177Lu-PSMA-I&T therapy were investigated. Whole-body SPECT/CT measurements were performed at 1, 2, 3 and 7 days post-injection. For both lesions (isocontour-based segmentation) and kidneys (CT-based segmentation), a reference model was employed including all four time points. To identify the best-matching fit function out of a pre-defined set of models, visual inspection, coefficients of variation and sum of squared errors were considered as goodness-of-fit criteria. Biologically effective doses (BEDs) calculated with different time samplings (days 1, 2, 3/1, 2, 7/1, 3, 7/2, 3, 7 and 1, 2/1, 3/1, 7) were compared to the reference. RESULTS The best-fit function was found to be a mono-exponential model for lesions and a bi-exponential model with a population-based parameter and two free parameters for kidneys. The BEDs calculated with the time sampling 1, 3, 7 days showed the lowest deviations from the reference for lesions with 4 ± 5%. Without day 7, still 86% of all lesions showed deviations from the reference < 10%. The outlier deviations showed a positive correlation with the effective half-life of the respective lesions. For kidneys, including days 1, 2, 3 achieved the best results with 0 ± 1%. Generally, deviations for kidneys were found to be small for all time samplings (max. 13%). CONCLUSIONS For combined optimization of the SPECT/CT time sampling for kidney and lesion dosimetry during 177Lu-PSMA-I&T therapy, the sampling with days 1, 3, 7 showed the smallest deviation from the reference. Without a late acquisition, using the schedule with days 1, 2, 3 is likewise feasible.
Collapse
Affiliation(s)
- Sandra Resch
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.
| | - Sarah Takayama Fouladgar
- grid.5252.00000 0004 1936 973XDepartment of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Mathias Zacherl
- grid.5252.00000 0004 1936 973XDepartment of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Gabriel T. Sheikh
- grid.5252.00000 0004 1936 973XDepartment of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Grigory Liubchenko
- grid.5252.00000 0004 1936 973XDepartment of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Mikhail Rumiantcev
- grid.5252.00000 0004 1936 973XDepartment of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Lena M. Unterrainer
- grid.5252.00000 0004 1936 973XDepartment of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Vera Wenter
- grid.5252.00000 0004 1936 973XDepartment of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Peter Bartenstein
- grid.5252.00000 0004 1936 973XDepartment of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Sibylle I. Ziegler
- grid.5252.00000 0004 1936 973XDepartment of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Harun Ilhan
- grid.5252.00000 0004 1936 973XDepartment of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Leonie Beyer
- grid.5252.00000 0004 1936 973XDepartment of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Guido Böning
- grid.5252.00000 0004 1936 973XDepartment of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Astrid Delker
- grid.5252.00000 0004 1936 973XDepartment of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
6
|
Kao YH. First Strike personalized predictive radioiodine prescription for inoperable metastatic differentiated thyroid cancer. ASIA OCEANIA JOURNAL OF NUCLEAR MEDICINE & BIOLOGY 2023; 11:158-167. [PMID: 37324232 PMCID: PMC10261697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 01/29/2023] [Accepted: 02/12/2023] [Indexed: 06/17/2023]
Abstract
Objectives The traditional practice of empiric radioiodine (I-131) prescription is scientifically obsolete and inappropriate for inoperable metastatic differentiated thyroid cancer. However, theranostically guided prescription is still years away for many institutions. A personalized predictive method of radioiodine prescription that bridges the gap between empiric and theranostic methods is presented. It is an adaptation of the "maximum tolerated activity" method, where serial blood sampling is replaced by population kinetics carefully chosen by the user. It aims to maximize crossfire benefits within safety constraints to overcome tumour absorbed dose heterogeneity for a safe and effective first radioiodine fraction i.e., the First Strike. Methods The EANM method of blood dosimetry was incorporated with population kinetics, marrow and lung safety constraints, body habitus and clinical assessment of metastatic extent. Population data of whole body and blood kinetics in patients with and without metastases, prepared by recombinant human thyroid stimulating hormone or thyroid hormone withdrawal, and the maximum safe marrow dose rate were deduced from published data. For diffuse lung metastases, the lung safety limit was linearly scaled by height and separated into lung and remainder-of-body components. Results The slowest whole body Time Integrated Activity Coefficient (TIAC) amongst patients with any metastases was 33.5±17.0 h and the highest percentage of whole body TIAC attributed to blood was 16.6±7.9%, prepared by thyroid hormone withdrawal. A variety of other average radioiodine kinetics is tabulated. Maximum safe marrow dose rate was deduced to be 0.265 Gy/h per fraction, where blood TIAC is normalised to administered activity. An easy-to-use calculator was developed which only requires height, weight and gender to populate recommendations for personalized First Strike prescription. The user decides by clinical gestalt whether the prescription is to be constrained by marrow or lung, then selects an activity depending on how extensive the metastases are likely to be. A Standard Female with oligometastasis and good urine output without diffuse lung metastasis is expected to safely tolerate 8.03 GBq of radioiodine as the First Strike. Conclusion This predictive method will help institutions rationalise the First Strike prescription based on radiobiologically sound principles, personalised to individual circumstances.
Collapse
Affiliation(s)
- Yung Hsiang Kao
- Department of Nuclear Medicine, The Royal Melbourne Hospital, Australia
| |
Collapse
|
7
|
Siebinga H, de Wit-van der Veen BJ, Stokkel MD, Huitema AD, Hendrikx JJ. Current use and future potential of (physiologically based) pharmacokinetic modelling of radiopharmaceuticals: a review. Theranostics 2022; 12:7804-7820. [PMID: 36451855 PMCID: PMC9706588 DOI: 10.7150/thno.77279] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 10/27/2022] [Indexed: 12/02/2022] Open
Abstract
Rationale: Physiologically based pharmacokinetic (PBPK) and population pharmacokinetic (PK) modelling approaches are widely accepted in non-radiopharmaceutical drug development and research, while there is no major role for these approaches in radiopharmaceutical development yet. In this review, a literature search was performed to specify different research purposes and questions that have previously been answered using both PBPK and population PK modelling for radiopharmaceuticals. Methods: The literature search was performed using the databases PubMed and Embase. Wide search terms included radiopharmaceutical, tracer, radioactivity, physiologically based pharmacokinetic model, PBPK, population pharmacokinetic model and nonlinear mixed-effects model. Results: Eight articles and twenty articles were included for this review based on this literature search for population PK modelling and PBPK modelling, respectively. Included population PK analyses showed to have an added value to develop predictive models for a population and to describe individual variability sources. Main purposes of PBPK models appeared related to optimizing treatment (planning), or more specifically: to find the optimal combination of peptide amount and radioactivity, to optimize treatment planning by reducing the number of measurements, to individualize treatment, to get insights in differences between pre-therapeutic and therapeutic scans or to understand inter-patient differences. Other main research subjects were regarding radiopharmaceutical comparisons, selecting ligands based on their peptide characteristics and gaining a better understanding of drug-drug interactions. Conclusions: The use of PK modelling approaches in radiopharmaceutical research remains scarce, but can be expanded to obtain a better understanding of PK and whole-body distribution of radiopharmaceuticals in general. PK modelling of radiopharmaceuticals has great potential for the nearby future and could contribute to the evolving research of radiopharmaceuticals.
Collapse
Affiliation(s)
- Hinke Siebinga
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Nuclear Medicine, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Marcel D.M. Stokkel
- Department of Nuclear Medicine, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Alwin 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
| | - Jeroen 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
| |
Collapse
|
8
|
O'Donoghue J, Zanzonico P, Humm J, Kesner A. Dosimetry in Radiopharmaceutical Therapy. J Nucl Med 2022; 63:1467-1474. [PMID: 36192334 DOI: 10.2967/jnumed.121.262305] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 07/14/2022] [Indexed: 11/27/2022] Open
Abstract
The application of radiopharmaceutical therapy for the treatment of certain diseases is well established, and the field is expanding. New therapeutic radiopharmaceuticals have been developed in recent years, and more are in the research pipeline. Concurrently, there is growing interest in the use of internal dosimetry as a means of personalizing, and potentially optimizing, such therapy for patients. Internal dosimetry is multifaceted, and the current state of the art is discussed in this continuing education article. Topics include the context of dosimetry, internal dosimetry methods, the advantages and disadvantages of incorporating dosimetry calculations in radiopharmaceutical therapy, a description of the workflow for implementing patient-specific dosimetry, and future prospects in the field.
Collapse
Affiliation(s)
- Joe O'Donoghue
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Pat Zanzonico
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - John Humm
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Adam Kesner
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
9
|
Gholami YH, Willowson KP, Bailey DL. Towards personalised dosimetry in patients with liver malignancy treated with 90Y-SIRT using in vivo-driven radiobiological parameters. EJNMMI Phys 2022; 9:49. [PMID: 35907097 PMCID: PMC9339072 DOI: 10.1186/s40658-022-00479-7] [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: 01/17/2022] [Accepted: 07/20/2022] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The prediction of response is one of the major challenges in radiation-based therapies. Although the selection of accurate linear-quadratic model parameters is essential for the estimation of radiation response and treatment outcome, there is a limited knowledge about these radiobiological parameters for liver tumours using radionuclide treatments. METHODS The "clinical radiobiological" parameters ([Formula: see text], [Formula: see text], [Formula: see text], [Formula: see text]) for twenty-five patients were derived using the generalised linear-quadratic model, the diagnostic ([18F] FDG PET/CT) and therapeutic ([90Y]-SIR-Spheres PET/CT) images to compute the biological effective dose and tumour control probability (TCP) for each patient. RESULTS It was estimated that the values for [Formula: see text] and [Formula: see text] parameters range in ≈ 0.001-1 Gy-1 and ≈ 1-49 Gy, respectively. We have demonstrated that the time factors, [Formula: see text], [Formula: see text] and [Formula: see text] are the key parameters when evaluating liver malignancy lesional response to [90Y]SIR-Spheres treatment. Patients with cholangiocarcinoma have been shown to have the longest average [Formula: see text] (≈ 236 ± 67 d), highest TCP (≈ 53 ± 17%) and total liver lesion glycolysis response ([Formula: see text] ≈ 64%), while patients with metastatic colorectal cancer tumours have the shortest average [Formula: see text] (≈ 129 ± 19 d), lowest TCP (≈ 28 ± 13%) and [Formula: see text] ≈ 8%, respectively. CONCLUSIONS Tumours with shorter [Formula: see text] have shown a shorter [Formula: see text] and thus poorer TCP and [Formula: see text]. Therefore, these results suggest for such tumours the [90Y]SIR-Spheres will be only effective at higher initial dose rate (e.g. > 50 Gy/day).
Collapse
Affiliation(s)
- Yaser H Gholami
- Faculty of Medicine and Health, The University of Sydney, Sydney, Australia. .,Sydney Vital Translational Cancer Research Centre, University of Sydney, Sydney, Australia. .,Department of Nuclear Medicine, Royal North Shore Hospital, Sydney, Australia.
| | - Kathy P Willowson
- Department of Nuclear Medicine, Royal North Shore Hospital, Sydney, Australia
| | - Dale L Bailey
- Faculty of Medicine and Health, The University of Sydney, Sydney, Australia. .,Sydney Vital Translational Cancer Research Centre, University of Sydney, Sydney, Australia. .,Department of Nuclear Medicine, Royal North Shore Hospital, Sydney, Australia.
| |
Collapse
|
10
|
Therapeutic efficacy of heterogeneously distributed radiolabelled peptides: Influence of radionuclide choice. Phys Med 2022; 96:90-100. [DOI: 10.1016/j.ejmp.2022.02.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 02/10/2022] [Accepted: 02/21/2022] [Indexed: 12/21/2022] Open
|
11
|
Park EA, Graves SA, Menda Y. The Impact of Radiopharmaceutical Therapy on Renal Function. Semin Nucl Med 2022; 52:467-474. [DOI: 10.1053/j.semnuclmed.2022.02.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 02/13/2022] [Accepted: 02/20/2022] [Indexed: 11/11/2022]
|
12
|
I-124 PET/CT image-based dosimetry in patients with differentiated thyroid cancer treated with I-131: correlation of patient-specific lesional dosimetry to treatment response. Ann Nucl Med 2022; 36:213-223. [PMID: 35119623 DOI: 10.1007/s12149-021-01655-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 07/12/2021] [Indexed: 11/01/2022]
Abstract
PURPOSE The objective of this study is to evaluate the lesion absorbed dose (AD), biological effective dose (BED), and equivalent uniform dose (EUD) to clinical-response relationship in lesional dosimetry for 131I therapy. METHODS Nineteen lesions in four patients with metastatic differentiated thyroid cancer (DTC) were evaluated. The patients underwent PET/CT imaging at 2 h, 24 h, 48 h, 72 h, and 96 h post administration of ~ 33-65 MBq (0.89-1.76 mCi) of 124I before undergoing 131I therapy. The 124I PET/CT images were used to perform dosimetry calculations for 131I therapy. Lesion dose-rate values were calculated using the time-activity data and integrated over the measured time points to obtain AD and BED. The Geant4 toolkit was used to run Monte Carlo on spheres the same size as the lesions to estimate EUD. The lesion AD, BED, and EUD values were correlated with response data (i.e. change in lesion size pre- and post-therapy): complete response (CR, i.e. disappearance of the lesion), partial response (PR, i.e. any decrease in lesion length), stable disease (SD, i.e., no change in length), and progressive disease (PD, i.e., any increase in length). RESULTS The lesion responses were CR and PR (58%, 11/19 lesions), SD (21%, 4/19), and PD (21%, 4/19). For CR and PR lesions, the ADs, BEDs and EUDs were > 75 Gy for 82% (9/11) and < 75 Gy for 18% (2/11). The ADs and BEDs were < 75 Gy for SD and PD lesions. CONCLUSION By performing retrospective dosimetry calculations for 131I therapy based on 124I PET/CT imaging, we evaluated the correlation of three dosimetric quantities to lesional response. When lesion AD, BED, and EUD values were > 75 Gy, 47% (9/19) of the lesions had a CR or PR. The AD, BED, and EUD values for SD and PD lesions were < 75 Gy. The data presented herein suggest that the greater the lesion AD, BED, and/or EUD, the higher the probability of a therapeutic response to 131I therapy.
Collapse
|
13
|
Toward three-dimensional patient-specific internal dosimetry using GATE Monte Carlo technique. Radiat Phys Chem Oxf Engl 1993 2022. [DOI: 10.1016/j.radphyschem.2022.110046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
14
|
Prasad V, Prasad S, Lehnert W, Brenner W, Kai H, Bronzel M, Kluge A. Effect of Peptide Dose on Radiation Dosimetry for Peptide Receptor Radionuclide Therapy with 177Lu-DOTATOC: A Pilot Study. Indian J Nucl Med 2021; 36:412-421. [PMID: 35125759 PMCID: PMC8771073 DOI: 10.4103/ijnm.ijnm_15_21] [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: 02/04/2021] [Accepted: 04/06/2021] [Indexed: 11/14/2022] Open
Abstract
Background: Optimal peptide concentration in treatment with 177Lu-DOTATOC/DOTATATE is a matter of debate. Most of the studies with peptide receptor radionuclide therapy mention peptide dose ranging between 100 and 250 μg. The aim of this is to identify possible differences in radiation-absorbed doses (D/Gy) to tumor and kidney as a function of the peptide mass dose in order to identify the most suitable peptide dose for treatment. The therapeutic index (Dtumor/Dkidneys) was assessed as a key parameter for the treatment response. Materials and Methods: Five patients with metastasized Grade 1 to Grade 2 neuroendocrine tumor were analyzed in this study. Patients (n = 4) received two cycles of treatment with intravenously injected 177Lu-DOTATOC containing peptide mass doses of 200 μg and 90 μg, alternatively; one patient was treated with 90 μg peptide mass in both the therapy cycles. Whole-body (head to mid-thigh) three-dimensional single-photon emission computerized tomography (3D SPECT)/CT images were acquired at 1, 4, 24, 48, and 72 h following the injection of 177Lu-DOTATOC. Attenuation correction for 3D SPECT images was performed using CT data acquired and fused with the SPECT data (SPECT/CT). Results: Overall, 28 target lesions (liver n = 17, lung n = 4, lymph nodes n = 1, and bone n = 2) were analyzed after 1st and 2nd therapy cycles. Tumor normalized absorbed doses varied by a factor of 74 between 0.35 and 26 mGy/MBq. Averaged over all patients, a higher normalized mean tumor dose (10.51 mGy/MBq) was achieved for a peptide dose of 200 μg compared to 90 μg (4.58 mGy/MBq). Kidneys doses varied by a factor of up to 4 between patients (0.25–1.0 mGy/MBq) (independent of dose cycle and peptide dose) and by a factor of up to 2 between dose cycles. The mean kidney dose was 13.7% higher for the 90 μg peptide dose compared to 200 μg. Given the higher tumor dose, the mean therapeutic index of a 200 μg mass dose was considerably higher (16.95), compared to a 90 μg mass dose (9.63). This coincided with the observation, that lesion volume reduction was more pronounced after an initial treatment with a 200 μg mass dose. Biologically effective dose was only 5. 1%–19.3% higher than the absorbed dose for individual dose cycles. Conclusions: Higher peptide dose of 200 μg appears to be more suitable than 90 μg in terms of tumor dose, kidney dose, and therapeutic index for treatment with 177Lu-DOTATOC.
Collapse
Affiliation(s)
- Vikas Prasad
- Department of Nuclear Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany.,Department of Nuclear Medicine, University Hospital Ulm, Ulm, Berlin, Germany.,German Cancer Consortium (DKTK), Partner Site Berlin, Berlin, Germany
| | - Sonal Prasad
- Department of Nuclear Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany.,Berlin Experimental Radionuclide Imaging Center, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Wencke Lehnert
- ABX - CRO Advanced Pharmaceutical Services, Dresden, Germany.,Department of Nuclear Medicine, University Hospital Hamburg Eppendorf, Berlin, Germany
| | - Winfried Brenner
- Department of Nuclear Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany.,German Cancer Consortium (DKTK), Partner Site Berlin, Berlin, Germany.,Berlin Experimental Radionuclide Imaging Center, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Huang Kai
- Department of Nuclear Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Marcus Bronzel
- ABX - CRO Advanced Pharmaceutical Services, Dresden, Germany
| | - Andreas Kluge
- ABX - CRO Advanced Pharmaceutical Services, Dresden, Germany
| |
Collapse
|
15
|
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
|
16
|
Nawrocki T, Tritt TC, Neti PVSV, Rosen AS, Dondapati AR, Howell RW. Design and testing of a microcontroller that enables alpha particle irradiators to deliver complex dose rate patterns. Phys Med Biol 2018; 63:245022. [PMID: 30524061 PMCID: PMC8528213 DOI: 10.1088/1361-6560/aaf269] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
There is increasing interest in using alpha particle emitting radionuclides for cancer therapy because of their unique cytotoxic properties which are advantageous for eradicating tumor cells. The high linear energy transfer (LET) of alpha particles produces a correspondingly high density of ionizations along their track. Alpha particle emitting radiopharmaceuticals deposit this energy in tissues over prolonged periods with complex dose rate patterns that depend on the physical half-life of the radionuclide, and the biological uptake and clearance half-times in tumor and normal tissues. We have previously shown that the dose rate increase half-time that arises as a consequence of these biokinetics can have a profound effect on the radiotoxicity of low-LET radiation. The microcontroller hardware and software described here offer a unique way to deliver these complex dose rate patterns with a broad-beam alpha particle irradiator, thereby enabling experiments to study the radiobiology of complex dose rate patterns of alpha particles. Complex dose rate patterns were created by precise manipulation of the timing of opening and closing of the electromechanical shutters of an α-particle irradiator. An Arduino Uno and custom circuitry was implemented to control the shutters. The software that controls the circuits and shutters has a user-friendly Graphic User Interface (GUI). Alpha particle detectors were used to validate the programmed dose rate profiles. Circuit diagrams and downloadable software are provided to facilitate adoption of this technology by other radiobiology laboratories.
Collapse
Affiliation(s)
- Tomer Nawrocki
- Division of Radiation Research, Department of Radiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States of America
| | | | | | | | | | | |
Collapse
|
17
|
Marin G, Vanderlinden B, Karfis I, Guiot T, Wimana Z, Reynaert N, Vandenberghe S, Flamen P. A dosimetry procedure for organs-at-risk in 177Lu peptide receptor radionuclide therapy of patients with neuroendocrine tumours. Phys Med 2018; 56:41-49. [PMID: 30527088 DOI: 10.1016/j.ejmp.2018.11.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 10/05/2018] [Accepted: 11/02/2018] [Indexed: 01/25/2023] Open
Abstract
PURPOSE Peptide receptor radionuclide therapy with 177Lu-DOTATATE has become a standard treatment modality in neuroendocrine tumours (NETs). No consensus has yet been reached however regarding the absorbed dose threshold for lesion response, the absorbed dose limit to organs-at-risk, and the optimal fractionation and activity to be administered. This is partly due to a lack of uniform and comparable dosimetry protocols. The present article details the development of an organ-at-risk dosimetry procedure, which could be implemented and used routinely in a clinical context. METHODS Forty-seven patients with NETs underwent 177Lu-DOTATATE therapy. Three SPECT/CT images were acquired at 4, 24 and 144-192 h post-injection. Three blood samples were obtained together with the SPECT/CT acquisitions and 2 additional samples were obtained around 30 min and 1 h post-injection. A bi-exponential fit was used to compute the source organ time-integrated activity coefficients. Coefficients were introduced into OLINDA/EXM software to compute organ-at-risk absorbed doses. Median values for all patients were computed for absorbed dose coefficient D/A0 and for late effective half-life T1/2eff for kidneys, spleen and red marrow. RESULTS Dosimetry resulted in a median[interquartile range] of 0.78[0.35], 1.07[0.58] and 0.028[0.010] Gy/GBq for D/A0 and of 55[9], 71[9] and 52[18] h for T1/2eff for kidneys, spleen and red marrow respectively. CONCLUSIONS A dosimetry procedure for organs-at-risk in 177Lu-DOTATATE therapy based on serial SPECT/CT images and blood samples can be implemented routinely in a clinical context with limited patient burden. The results obtained were in accordance with those of other centres.
Collapse
Affiliation(s)
- Gwennaëlle Marin
- Department of Medical Physics, Institut Jules Bordet-Université Libre de Bruxelles (ULB), 121 boulevard de Waterloo, 1000 Brussels, Belgium; Medical Imaging and Signal Processing (MEDISIP), Department of Electronics and Information Systems (ELIS), Faculty of Engineering and Architecture (FEA), Ghent University (UGent), 185 De Pintelaan, 9000 Gent, Belgium.
| | - Bruno Vanderlinden
- Department of Medical Physics, Institut Jules Bordet-Université Libre de Bruxelles (ULB), 121 boulevard de Waterloo, 1000 Brussels, Belgium.
| | - Ioannis Karfis
- Department of Nuclear Medicine, Institut Jules Bordet-Université Libre de Bruxelles (ULB), 121 boulevard de Waterloo, 1000 Brussels, Belgium.
| | - Thomas Guiot
- Department of Medical Physics, Institut Jules Bordet-Université Libre de Bruxelles (ULB), 121 boulevard de Waterloo, 1000 Brussels, Belgium.
| | - Zena Wimana
- Department of Nuclear Medicine, Institut Jules Bordet-Université Libre de Bruxelles (ULB), 121 boulevard de Waterloo, 1000 Brussels, Belgium.
| | - Nick Reynaert
- Department of Medical Physics, Institut Jules Bordet-Université Libre de Bruxelles (ULB), 121 boulevard de Waterloo, 1000 Brussels, Belgium.
| | - Stefaan Vandenberghe
- Medical Imaging and Signal Processing (MEDISIP), Department of Electronics and Information Systems (ELIS), Faculty of Engineering and Architecture (FEA), Ghent University (UGent), 185 De Pintelaan, 9000 Gent, Belgium.
| | - Patrick Flamen
- Department of Nuclear Medicine, Institut Jules Bordet-Université Libre de Bruxelles (ULB), 121 boulevard de Waterloo, 1000 Brussels, Belgium.
| |
Collapse
|
18
|
Jeremic MZ, Matovic MD, Krstic DZ, Pantovic SB, Nikezic DR. A five-compartment biokinetic model for 90 Y-DOTATOC therapy. Med Phys 2018; 45:5577-5585. [PMID: 30291717 DOI: 10.1002/mp.13229] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 08/16/2018] [Accepted: 09/22/2018] [Indexed: 12/31/2022] Open
Abstract
PURPOSE Neuroendocrine tumors (NETs) are now routinely treated by radiopeptide targeted therapy using somatostatin receptor-binding peptides such as 90 Y- and 177 Lu-DOTATOC. The objective of this work was to develop a biokinetics model of 90 Y labelled DOTATOC, which is applied in the therapy of NETs to estimate doses in kidney and tumor. METHODS A multi-compartment model described by two sets of differential equations, one set for the actual 30-min infusion and the other set for the post-infusion period was developed and activities were measured by liquid scintillation counting in blood (compartment 1) and the urine (compartment 3). The inter-compartment transfer coefficients, λij , were varied to yield the best fit of the calculated to the measured time-activity data and the 90 Y-DOTATOC time-activity data in the five-compartments comprising the human body were thus determined. The resulting time-activity curves were integrated over the interval from 0 to 72 h post administration to obtain the number of radioactive decays in each compartment and, in case of the kidneys and tumor, then multiplied by the self-dose 90 Y beta particle absorbed fraction, determined by Monte Carlo (MC) simulation, the kidney and tumor absorbed doses. RESULTS Transfer coefficients λij , were determined for five-compartments for all patients. Time- activity curves of 90 Y-DOTATOC in 14 patients were determined, and two typical ones are shown graphically. Absorbed doses in the tumor and kidneys, obtained by the developed method, were determined. The mean absorbed dose in a kidney per unit of administered activity is 1.43 mGy/MBq (range 0.73-2.42 mGy/MBq). The tumor dose was determined as 30.94 mGy/MBq (range 20.05-42.31 mGy/MBq). CONCLUSION Analytical solution of a biokinetic model for 90 Y-DOTATOC therapy enabled determination of the transfer coefficients and derivation of time-activity curves and kidney and tumor absorbed doses for 14 treated patients. The model can be applied to other radionuclides where elimination is predominantly through urine, which is often the case in radiopharmaceuticals.
Collapse
Affiliation(s)
- Marija Z Jeremic
- Department of Nuclear Medicine, Clinical Center Kragujevac, 34000, Kragujevac, Serbia.,Department of Physics, Faculty of Science, University of Kragujevac, 34000, Kragujevac, Serbia
| | - Milovan D Matovic
- Department of Nuclear Medicine, Clinical Center Kragujevac, 34000, Kragujevac, Serbia.,Department of Nuclear Medicine, Faculty of Medical Sciences, University of Kragujevac, 34000, Kragujevac, Serbia
| | - Dragana Z Krstic
- Department of Physics, Faculty of Science, University of Kragujevac, 34000, Kragujevac, Serbia
| | - Suzana B Pantovic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, 34000, Kragujevac, Serbia
| | - Dragoslav R Nikezic
- Department of Physics, Faculty of Science, University of Kragujevac, 34000, Kragujevac, Serbia
| |
Collapse
|
19
|
Eberlein U, Cremonesi M, Lassmann M. Individualized Dosimetry for Theranostics: Necessary, Nice to Have, or Counterproductive? J Nucl Med 2017; 58:97S-103S. [PMID: 28864620 DOI: 10.2967/jnumed.116.186841] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 03/10/2017] [Indexed: 11/16/2022] Open
Abstract
In 2005, the term theragnostics (theranostics) was introduced for describing the use of imaging for therapy planning in radiation oncology. In nuclear medicine, this expression describes the use of tracers for predicting the absorbed doses in molecular radiotherapy and, thus, the safety and efficacy of a treatment. At present, the most successful groups of isotopes for this purpose are 123I/124I/131I, 68Ga/177Lu, and 111In/86Y/90Y. The purpose of this review is to summarize available data on the dosimetry and dose-response relationships of several theranostic compounds, with a special focus on radioiodine therapy for differentiated thyroid cancer and peptide receptor radionuclide therapy. These are treatment modalities for which dose-response relationships for healthy tissues and tumors have been demonstrated. In addition, available data demonstrate that posttherapeutic dosimetry after a first treatment cycle predicts the absorbed doses in further cycles. Both examples show the applicability of the concept of theranostics in molecular radiotherapies. Nevertheless, unanswered questions need to be addressed in clinical trials incorporating dosimetry-related concepts for determining the amount of therapeutic activity to be administered.
Collapse
Affiliation(s)
- Uta Eberlein
- Klinik und Poliklinik für Nuklearmedizin, Universitätsklinikum Würzburg, Würzburg, Germany; and
| | - Marta Cremonesi
- Radiation Research Unit, Istituto Europeo di Oncologia, Milano, Italy
| | - Michael Lassmann
- Klinik und Poliklinik für Nuklearmedizin, Universitätsklinikum Würzburg, Würzburg, Germany; and
| |
Collapse
|
20
|
Solanki JH, Tritt T, Pasternack JB, Kim JJ, Leung CN, Domogauer JD, Colangelo NW, Narra VR, Howell RW. Cellular Response to Exponentially Increasing and Decreasing Dose Rates: Implications for Treatment Planning in Targeted Radionuclide Therapy. Radiat Res 2017; 188:221-234. [PMID: 28541775 PMCID: PMC5669265 DOI: 10.1667/rr14766.1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The treatment of cancer using targeted radionuclide therapy is of interest to nuclear medicine and radiation oncology because of its potential for killing tumor cells while minimizing dose-limiting toxicities to normal tissue. The ionizing radiations emitted by radiopharmaceuticals deliver radiation absorbed doses over protracted periods of time with continuously varying dose rates. As targeted radionuclide therapy becomes a more prominent part of cancer therapy, accurate models for estimating the biologically effective dose (BED) or equieffective dose (EQD2α/β) will become essential for treatment planning. This study examines the radiobiological impact of the dose rate increase half-time during the uptake phase of the radiopharmaceutical. MDA-MB-231 human breast cancer cells and V79 Chinese hamster lung fibroblasts were irradiated chronically with 662 keV γ rays delivered with time-varying dose rates that are clinically relevant. The temporal dose-rate patterns were: 1. acute, 2. exponential decrease with a half-time of 64 h (Td = 64 h), 3. initial exponential increase to a maximum (half time Ti = 2, 8 or 24 h) followed by exponential decrease (Td = 64 h). Cell survival assays were conducted and surviving fractions were determined. There was a marked reduction in biological effect when Ti was increased. Cell survival data were tested against existing dose-response models to assess their capacity to predict response. Currently accepted models that are used in radiation oncology overestimated BED and EQD2α/β at low-dose rates and underestimated them at high-dose rates. This appears to be caused by an adaptive response arising as a consequence of the initial low-dose-rate phase of exposure. An adaptive response function was derived that yields more accurate BED and EQD2α/β values over the spectrum of dose rates and absorbed doses delivered. Our experimental data demonstrate a marked increase in cell survival when the dose-rate-increase half-time is increased, thereby suggesting an adaptive response arising as a consequence of this phase of exposure. We have modified conventional radiobiological models used in the clinic for brachytherapy and external beams of radiation to account for this phenomenon and facilitate their use for treatment planning in targeted radionuclide therapy.
Collapse
Affiliation(s)
- Jay H. Solanki
- Division of Radiation Research, Department of Radiology, New Jersey Medical School Cancer Center, Rutgers, The State University of New Jersey, Newark, New Jersey
| | - Thomas Tritt
- Division of Radiation Research, Department of Radiology, New Jersey Medical School Cancer Center, Rutgers, The State University of New Jersey, Newark, New Jersey
| | - Jordan B. Pasternack
- Division of Radiation Research, Department of Radiology, New Jersey Medical School Cancer Center, Rutgers, The State University of New Jersey, Newark, New Jersey
| | - Julia J. Kim
- Division of Radiation Research, Department of Radiology, New Jersey Medical School Cancer Center, Rutgers, The State University of New Jersey, Newark, New Jersey
| | - Calvin N. Leung
- Division of Radiation Research, Department of Radiology, New Jersey Medical School Cancer Center, Rutgers, The State University of New Jersey, Newark, New Jersey
| | - Jason D. Domogauer
- Division of Radiation Research, Department of Radiology, New Jersey Medical School Cancer Center, Rutgers, The State University of New Jersey, Newark, New Jersey
| | - Nicholas W. Colangelo
- Division of Radiation Research, Department of Radiology, New Jersey Medical School Cancer Center, Rutgers, The State University of New Jersey, Newark, New Jersey
| | - Venkat R. Narra
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - Roger W. Howell
- Division of Radiation Research, Department of Radiology, New Jersey Medical School Cancer Center, Rutgers, The State University of New Jersey, Newark, New Jersey
| |
Collapse
|
21
|
Delker A, Ilhan H, Zach C, Brosch J, Gildehaus FJ, Lehner S, Bartenstein P, Böning G. The Influence of Early Measurements Onto the Estimated Kidney Dose in [(177)Lu][DOTA(0),Tyr(3)]Octreotate Peptide Receptor Radiotherapy of Neuroendocrine Tumors. Mol Imaging Biol 2016; 17:726-34. [PMID: 25790773 DOI: 10.1007/s11307-015-0839-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PURPOSE Multiple measurements have been required to estimate the radiation dose to the kidneys resulting from [(177)Lu]DOTATATE therapy for neuroendocrine tumors. The aim of this study was to investigate the influence of early time-point measurement in the renal dose calculation. PROCEDURES Anterior/posterior whole-body planar scintigraphy images were acquired at approx. 1, 24, 48, and 72 h after administration of [(177)Lu]DOTATATE. Furthermore, we acquired planar 1-bed dynamic recordings in 12 frames (5 min each) during the first hour. We assessed kidney exposure with a three-phase model consisting of a linear increase to the maximum within the initial minutes p.i., followed a bi-exponential decline. This three-phase-model served as reference for evaluating accuracy of dose estimates in 105 kidneys calculated by conventional mono-exponential fitting of the final three and four whole-body images. RESULTS Mean effective half-life times for the reference model were 25.8 ± 12.0 min and 63.9 ± 17.6 h, predicting a mean renal dose of 5.7 ± 2.1 Gy. The effective half-life time was 46.3 ± 15.4 h for the last four and 63.3 ± 17.0 h for the last three data points. The mean start of the first whole-body measurement was 1.2 ± 0.1 h p.i. The ratio of fast to slow phases was 28.1 ± 23.9% at this time point, which caused a mean absolute percentage dose deviation of 12.4% for four data points, compared to 3.1% for three data points. At a mean time of 2.4 h p.i. (max 5.1 h), the ratio of fast to slow phase declined below 5%. CONCLUSIONS Kinetic analysis of renal uptake using dynamic planar scans from the first hour after injection revealed a fast and a slow washout phase. Although the fast phase did not contribute substantially to the estimated renal dose, it could influence planar measurements performed within the first hours. We found that the presence of two clearance phases can hamper accurate dose estimation based on a single-phase model, resulting in approximately 12.4% dose underestimation, thus potentially resulting in overtreatment. In the absence of dynamic initial recordings, the first dosimetry measurements should therefore be obtained later than 3-5 h after [(177)Lu]DOTATATE injection. Omitting the early whole-body image reduced the dose estimation error to 3.1%.
Collapse
Affiliation(s)
- Andreas Delker
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität, Munich, Germany
| | - Harun Ilhan
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität, Munich, Germany
| | - Christian Zach
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität, Munich, Germany
| | - Julia Brosch
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität, Munich, Germany
| | - Franz Josef Gildehaus
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität, Munich, Germany
| | - Sebastian Lehner
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität, Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität, Munich, Germany
| | - Guido Böning
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität, Munich, Germany.
| |
Collapse
|
22
|
Mínguez P, Gustafsson J, Flux G, Gleisner KS. Biologically effective dose in fractionated molecular radiotherapy--application to treatment of neuroblastoma with (131)I-mIBG. Phys Med Biol 2016; 61:2532-51. [PMID: 26948833 DOI: 10.1088/0031-9155/61/6/2532] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
In this work, the biologically effective dose (BED) is investigated for fractionated molecular radiotherapy (MRT). A formula for the Lea-Catcheside G-factor is derived which takes the possibility of combinations of sub-lethal damage due to radiation from different administrations of activity into account. In contrast to the previous formula, the new G-factor has an explicit dependence on the time interval between administrations. The BED of tumour and liver is analysed in MRT of neuroblastoma with (131)I-mIBG, following a common two-administration protocol with a mass-based activity prescription. A BED analysis is also made for modified schedules, when due to local regulations there is a maximum permitted activity for each administration. Modifications include both the simplistic approach of delivering this maximum permitted activity in each of the two administrations, and also the introduction of additional administrations while maintaining the protocol-prescribed total activity. For the cases studied with additional (i.e. more than two) administrations, BED of tumour and liver decreases at most 12% and 29%, respectively. The decrease in BED of the tumour is however modest compared to the two-administration schedule using the maximum permitted activity, where the decrease compared to the original schedule is 47%.
Collapse
Affiliation(s)
- Pablo Mínguez
- Department of Medical Radiation Physics, Clinical Sciences Lund, Lund University, 22185 Lund, Sweden. Department of Medical Physics, Gurutzeta/Cruces University Hospital, 48903 Barakaldo, Spain
| | | | | | | |
Collapse
|
23
|
Kletting P, Kull T, Maaß C, Malik N, Luster M, Beer AJ, Glatting G. Optimized Peptide Amount and Activity for ⁹⁰Y-Labeled DOTATATE Therapy. J Nucl Med 2015; 57:503-8. [PMID: 26678617 DOI: 10.2967/jnumed.115.164699] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 11/23/2015] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED In peptide receptor radionuclide therapy with (90)Y-labeled DOTATATE, the kidney absorbed dose limits the maximum amount of total activity that can be safely administered in many patients. A higher tumor-to-kidney absorbed dose ratio might be achieved by optimizing the amount of injected peptide and activity, as recent studies have shown different degrees of receptor saturation for normal tissue and tumor. The aim of this work was to develop and implement a modeling method for treatment planning to determine the optimal combination of peptide amount and pertaining therapeutic activity for each patient. METHODS A whole-body physiologically based pharmacokinetic (PBPK) model was developed. General physiologic parameters were taken from the literature. Individual model parameters were fitted to a series (n= 12) of planar γ-camera and serum measurements ((111)In-DOTATATE) of patients with meningioma or neuroendocrine tumors (NETs). Using the PBPK model and the individually estimated parameters, we determined the tumor, liver, spleen, and red marrow biologically effective doses (BEDs) for a maximal kidney BED (20 Gy2.5) for different peptide amounts and activities. The optimal combination of peptide amount and activity for maximal tumor BED, considering the additional constraint of a red marrow BED less than 1 Gy15, was individually quantified. RESULTS The PBPK model describes the biokinetic data well considering the criteria of visual inspection, the coefficients of determination, the relative standard errors (<50%), and the correlation of the parameters (<0.8). All fitted parameters were in a physiologically reasonable range but varied considerably between patients, especially tumor perfusion (meningioma, 0.1-1 mL·g(-1)·min(-1), and NETs, 0.02-1 mL·g(-1)·min(-1)) and receptor density (meningioma, 5-34 nmol·L(-1), and NETs, 7-35 nmol·L(-1)). Using the proposed method, we identified the optimal amount and pertaining activity to be 76 ± 46 nmol (118 ± 71 μg) and 4.2 ± 1.8 GBq for meningioma and 87 ± 50 nmol (135 ± 78 μg) and 5.1 ± 2.8 GBq for NET patients. CONCLUSION The presented work suggests that to achieve higher efficacy and safety for (90)Y-DOATATE therapy, both the administered amount of peptide and the activity should be optimized in treatment planning using the proposed method. This approach could also be adapted for therapy with other peptides.
Collapse
Affiliation(s)
- Peter Kletting
- Department of Nuclear Medicine, Ulm University, Ulm, Germany
| | - Thomas Kull
- Department of Nuclear Medicine, Ulm University, Ulm, Germany
| | - Christian Maaß
- Medical Radiation Physics/Radiation Protection, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; and
| | - Noeen Malik
- Department of Nuclear Medicine, Ulm University, Ulm, Germany
| | - Markus Luster
- Klinik für Nuklearmedizin, Universität Marburg, Marburg, Germany
| | - Ambros J Beer
- Department of Nuclear Medicine, Ulm University, Ulm, Germany
| | - Gerhard Glatting
- Medical Radiation Physics/Radiation Protection, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; and
| |
Collapse
|
24
|
Monte Carlo Calculation of Radioimmunotherapy with (90)Y-, (177)Lu-, (131)I-, (124)I-, and (188)Re-Nanoobjects: Choice of the Best Radionuclide for Solid Tumour Treatment by Using TCP and NTCP Concepts. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2015; 2015:284360. [PMID: 26136812 PMCID: PMC4469173 DOI: 10.1155/2015/284360] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 10/27/2014] [Indexed: 12/26/2022]
Abstract
Radioimmunotherapy has shown that the use of monoclonal antibodies combined with a radioisotope like 131I or 90Y still remains ineffective for solid and radioresistant tumour treatment. Previous simulations have revealed that an increase in the number of 90Y labelled to each antibody or nanoobject could be a solution to improve treatment output. It now seems important to assess the treatment output and toxicity when radionuclides such as 90Y, 177Lu, 131I, 124I, and 188Re are used. Tumour control probability (TCP) and normal tissue complication probability (NTCP) curves versus the number of radionuclides per nanoobject were computed with MCNPX to evaluate treatment efficacy for solid tumours and to predict the incidence of surrounding side effects. Analyses were carried out for two solid tumour sizes of 0.5 and 1.0 cm radius and for nanoobject (i.e., a radiolabelled antibody) distributed uniformly or nonuniformly throughout a solid tumour (e.g., Non-small-cell-lung cancer (NSCLC)). 90Y and 188Re are the best candidates for solid tumour treatment when only one radionuclide is coupled to one carrier. Furthermore, regardless of the radionuclide properties, high values of TCP can be reached without toxicity if the number of radionuclides per nanoobject increases.
Collapse
|
25
|
Woliner-van der Weg W, Schoffelen R, Hobbs RF, Gotthardt M, Goldenberg DM, Sharkey RM, Slump CH, van der Graaf WT, Oyen WJ, Boerman OC, Sgouros G, Visser EP. Tumor and red bone marrow dosimetry: comparison of methods for prospective treatment planning in pretargeted radioimmunotherapy. EJNMMI Phys 2015; 2:5. [PMID: 26501807 PMCID: PMC4545615 DOI: 10.1186/s40658-014-0104-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 11/25/2014] [Indexed: 11/20/2022] Open
Abstract
Background Red bone marrow (RBM) toxicity is dose-limiting in (pretargeted) radioimmunotherapy (RIT). Previous blood-based and two-dimensional (2D) image-based methods have failed to show a clear dose-response relationship. We developed a three-dimensional (3D) image-based RBM dosimetry approach using the Monte Carlo-based 3D radiobiological dosimetry (3D-RD) software and determined its additional value for predicting RBM toxicity. Methods RBM doses were calculated for 13 colorectal cancer patients after pretargeted RIT with the two-step administration of an anti-CEA × anti-HSG bispecific monoclonal antibody and a 177Lu-labeled di-HSG-peptide. 3D-RD RBM dosimetry was based on the lumbar vertebrae, delineated on single photon emission computed tomography (SPECT) scans acquired directly, 3, 24, and 72 h after 177Lu administration. RBM doses were correlated to hematologic effects, according to NCI-CTC v3 and compared with conventional 2D cranium-based and blood-based dosimetry results. Tumor doses were calculated with 3D-RD, which has not been possible with 2D dosimetry. Tumor-to-RBM dose ratios were calculated and compared for 177Lu-based pretargeted RIT and simulated pretargeted RIT with 90Y. Results 3D-RD RBM doses of all seven patients who developed thrombocytopenia were higher (range 0.43 to 0.97 Gy) than that of the six patients without thrombocytopenia (range 0.12 to 0.39 Gy), except in one patient (0.47 Gy) without thrombocytopenia but with grade 2 leucopenia. Blood and 2D image-based RBM doses for patients with grade 1 to 2 thrombocytopenia were in the same range as in patients without thrombocytopenia (0.14 to 0.29 and 0.11 to 0.26 Gy, respectively). Blood-based RBM doses for two grade 3 to 4 patients were higher (0.66 and 0.51 Gy, respectively) than the others, and the cranium-based dose of only the grade 4 patient was higher (0.34 Gy). Tumor-to-RBM dose ratios would increase by 25% on average when treating with 90Y instead of 177Lu. Conclusions 3D dosimetry identifies patients at risk of developing any grade of RBM toxicity more accurately than blood- or 2D image-based methods. It has the added value to enable calculation of tumor-to-RBM dose ratios.
Collapse
Affiliation(s)
- Wietske Woliner-van der Weg
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, P.O. Box 9101, 6500, HB, Nijmegen, The Netherlands.
| | - Rafke Schoffelen
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, P.O. Box 9101, 6500, HB, Nijmegen, The Netherlands.
| | - Robert F Hobbs
- Department of Radiology, Johns Hopkins University, Baltimore, MD, USA.
| | - Martin Gotthardt
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, P.O. Box 9101, 6500, HB, Nijmegen, The Netherlands.
| | | | | | | | | | - Wim Jg Oyen
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, P.O. Box 9101, 6500, HB, Nijmegen, The Netherlands.
| | - Otto C Boerman
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, P.O. Box 9101, 6500, HB, Nijmegen, The Netherlands.
| | - George Sgouros
- Department of Radiology, Johns Hopkins University, Baltimore, MD, USA.
| | - Eric P Visser
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, P.O. Box 9101, 6500, HB, Nijmegen, The Netherlands.
| |
Collapse
|
26
|
Cremonesi M, Ferrari M, Botta F, Guerriero F, Garibaldi C, Bodei L, De Cicco C, Grana CM, Pedroli G, Orecchia R. Planning combined treatments of external beam radiation therapy and molecular radiotherapy. Cancer Biother Radiopharm 2014; 29:227-37. [PMID: 25006794 DOI: 10.1089/cbr.2014.1607] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Molecular radiotherapy (MRT) with radiolabeled molecules has being constantly evolving, leading to notable results in cancer treatment. In some cases, the absorbed doses delivered to tumors by MRT are sufficient to obtain complete responses; in other cases, instead, to be effective, MRT needs to be combined with other therapeutic approaches. Recently, several studies proposed the combination of MRT with external beam radiation therapy (EBRT). Some describe the theoretical basis within radiobiological models, others report the results of clinical phase I-II studies aimed to assess the feasibility and tolerability. The latter includes the treatment of various tumors, such as meningiomas, paragangliomas, non-Hodgkin's lymphomas, bone, brain, hepatic, and breast lesions. The underlying principle of combined MRT and EBRT is the possibility of exploiting the full potential of each modality, given the different organs at risk. Target tissues can indeed receive a higher irradiation, while respecting the threshold limits of more than one critical tissue. Nevertheless, clinical trials are empirical and optimization is still a theoretical issue. This article describes the state of the art of combined MRT and EBRT regarding the rationale and the results of clinical studies, with special focus on the possibility of treatment improvement.
Collapse
Affiliation(s)
- Marta Cremonesi
- Department of Medical Imaging and Radiation Sciences, Istituto Europeo di Oncologia , Milan, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Hobbs RF, Howell RW, Song H, Baechler S, Sgouros G. Redefining relative biological effectiveness in the context of the EQDX formalism: implications for alpha-particle emitter therapy. Radiat Res 2014; 181:90-8. [PMID: 24502376 DOI: 10.1667/rr13483.1] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Alpha-particle radiopharmaceutical therapy (αRPT) is currently enjoying increasing attention as a viable alternative to chemotherapy for targeting of disseminated micrometastatic disease. In theory, αRPT can be personalized through pre-therapeutic imaging and dosimetry. However, in practice, given the particularities of α-particle emissions, a dosimetric methodology that accurately predicts the thresholds for organ toxicity has not been reported. This is in part due to the fact that the biological effects caused by α-particle radiation differ markedly from the effects caused by traditional external beam (photon or electron) radiation or β-particle emitting radiopharmaceuticals. The concept of relative biological effectiveness (RBE) is used to quantify the ratio of absorbed doses required to achieve a given biological response with alpha particles versus a reference radiation (typically a beta emitter or external beam radiation). However, as conventionally defined, the RBE varies as a function of absorbed dose and therefore a single RBE value is limited in its utility because it cannot be used to predict response over a wide range of absorbed doses. Therefore, efforts are underway to standardize bioeffect modeling for different fractionation schemes and dose rates for both nuclear medicine and external beam radiotherapy. Given the preponderant use of external beams of radiation compared to nuclear medicine in cancer therapy, the more clinically relevant quantity, the 2 Gy equieffective dose, EQD2(α/β), has recently been proposed by the ICRU. In concert with EQD2(α/β), we introduce a new, redefined RBE quantity, named RBE2(α/β), as the ratio of the two linear coefficients that characterize the α particle absorbed dose-response curve and the low-LET megavoltage photon 2 Gy fraction equieffective dose-response curve. The theoretical framework for the proposed new formalism is presented along with its application to experimental data obtained from irradiation of a breast cancer cell line. Radiobiological parameters are obtained using the linear quadratic model to fit cell survival data for MDA-MB-231 human breast cancer cells that were irradiated with either α particles or a single fraction of low-LET (137)Cs γ rays. From these, the linear coefficient for both the biologically effective dose (BED) and the EQD2(α/β) response lines were derived for fractionated irradiation. The standard RBE calculation, using the traditional single fraction reference radiation, gave RBE values that ranged from 2.4 for a surviving fraction of 0.82-6.0 for a surviving fraction of 0.02, while the dose-independent RBE2(4.6) value was 4.5 for all surviving fraction values. Furthermore, bioeffect modeling with RBE2(α/β) and EQD2(α/β) demonstrated the capacity to predict the surviving fraction of cells irradiated with acute and fractionated low-LET radiation, α particles and chronic exponentially decreasing dose rates of low-LET radiation. RBE2(α/β) is independent of absorbed dose for α-particle emitters and it provides a more logical framework for data reporting and conversion to equieffective dose than the conventional dose-dependent definition of RBE. Moreover, it provides a much needed foundation for the ongoing development of an α-particle dosimetry paradigm and will facilitate the use of tolerance dose data available from external beam radiation therapy, thereby helping to develop αRPT as a single modality as well as for combination therapies.
Collapse
Affiliation(s)
- Robert F Hobbs
- a Department of Radiology, Johns Hopkins University, School of Medicine, Baltimore Maryland
| | | | | | | | | |
Collapse
|
28
|
Van Binnebeek S, Baete K, Vanbilloen B, Terwinghe C, Koole M, Mottaghy FM, Clement PM, Mortelmans L, Haustermans K, Van Cutsem E, Verbruggen A, Bogaerts K, Verslype C, Deroose CM. Individualized dosimetry-based activity reduction of ⁹⁰Y-DOTATOC prevents severe and rapid kidney function deterioration from peptide receptor radionuclide therapy. Eur J Nucl Med Mol Imaging 2014; 41:1141-57. [PMID: 24668274 DOI: 10.1007/s00259-013-2670-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Accepted: 12/10/2013] [Indexed: 12/25/2022]
Abstract
PURPOSE Assessment of kidney function evolution after (90)Y-DOTATOC peptide receptor radionuclide therapy (PRRT) with capped activity administration based on a 37-Gy threshold of biological effective dose (BED) to the kidney. METHODS In a prospective phase II study, patients with metastasized neuroendocrine tumours were evaluated for therapy using 185 MBq (111)In-pentetreotide with amino acid coinfusion. Planar whole-body images were acquired at four time-points after injection and kidney volumes were measured using CT/MRI. BED to the kidneys was estimated using an extended BED formula and biexponential renal clearance. Based on published BED dose-toxicity relationships, we allowed a maximal kidney BED of 37 Gy; if the calculated BED exceeded 37 Gy, treatment activity was reduced accordingly. Kidney function was assessed at baseline and at 18 months, predominantly using (51)Cr-EDTA. The rate of renal function decline was expressed as annual glomerular filtration rate loss (aGFRL). RESULTS Only 22 of 50 patients reached the 18-months time-point, with most missing patients having died due to disease progression. In the 22 patients who reached 18 months, no rapid kidney function deterioration was observed over the 18 months, aGFRL >33% was not seen, and only three patients showed an increase of one toxicity grade and one patient an increase of two grades. No significant correlations between kidney volume (p = 0.35), baseline GFR (p = 0.18), risk factors for renal function loss (p = 0.74) and aGFRL were observed. Among the 28 patients who did not reach 18 months, one developed grade 4 kidney toxicity at 15 months after PRRT. CONCLUSION Prospective dosimetry using a 37 Gy BED as the threshold for kidney toxicity is a good guide for (90)Y-DOTATOC PRRT and is associated with a low risk of rapid renal function deterioration and evolution to severe nephrotoxicity.
Collapse
|
29
|
Fahey F, Zukotynski K, Capala J, Knight N. Targeted radionuclide therapy: proceedings of a joint workshop hosted by the National Cancer Institute and the Society of Nuclear Medicine and Molecular Imaging. J Nucl Med 2014; 55:337-48. [PMID: 24396032 DOI: 10.2967/jnumed.113.135178] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Frederic Fahey
- Boston Children's Hospital, Boston, Massachusetts, and Harvard Medical School, Boston, Massachusetts
| | | | | | | | | |
Collapse
|
30
|
Budiawan H, Salavati A, Kulkarni HR, Baum RP. Peptide receptor radionuclide therapy of treatment-refractory metastatic thyroid cancer using (90)Yttrium and (177)Lutetium labeled somatostatin analogs: toxicity, response and survival analysis. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2013; 4:39-52. [PMID: 24380044 PMCID: PMC3867728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Accepted: 10/27/2013] [Indexed: 06/03/2023]
Abstract
The overall survival rate of non-radioiodine avid differentiated (follicular, papillary, medullary) thyroid carcinoma is significantly lower than for patients with iodine-avid lesions. The purpose of this study was to evaluate toxicity and efficacy (response and survival) of peptide receptor radionuclide therapy (PRRT) in non-radioiodine-avid or radioiodine therapy refractory thyroid cancer patients. Sixteen non-radioiodine-avid and/or radioiodine therapy refractory thyroid cancer patients, including follicular thyroid carcinoma (n = 4), medullary thyroid carcinoma (n = 8), Hürthle cell thyroid carcinoma (n = 3), and mixed carcinoma (n = 1) were treated with PRRT by using (90)Yttrium and/or (177)Lutetium labeled somatostatin analogs. (68)Ga somatostatin receptor PET/CT was used to determine the somatostatin receptor density in the residual tumor/metastatic lesions and to assess the treatment response. Hematological profiles and renal function were periodically examined after treatment. By using fractionated regimen, only mild, reversible hematological toxicity (grade 1) or nephrotoxicity (grade 1) were seen. Response assessment (using EORTC criteria) was performed in 11 patients treated with 2 or more (maximum 5) cycles of PRRT and showed disease stabilization in 4 (36.4%) patients. Two patients (18.2%) showed partial remission, in the remaining 5 patients (45.5%) disease remained progressive. Kaplan-Meier analysis resulted in a mean survival after the first PRRT of 4.2 years (95% CI, range 2.9-5.5) and median progression free survival of 25 months (inter-quartiles: 12-43). In non-radioiodine-avid/radioiodine therapy refractory thyroid cancer patients, PRRT is a promising therapeutic option with minimal toxicity, good response rate and excellent survival benefits.
Collapse
Affiliation(s)
- Hendra Budiawan
- THERANOSTICS Center for Molecular Radiotherapy and Molecular Imaging, ENETS Center of ExcellenceZentralklinik Bad Berka, Germany
- Department of Nuclear Medicine, Mochtar Riady Comprehensive Cancer Centre - Siloam Hospitals SemanggiJakarta, Indonesia
| | - Ali Salavati
- THERANOSTICS Center for Molecular Radiotherapy and Molecular Imaging, ENETS Center of ExcellenceZentralklinik Bad Berka, Germany
- Department of Radiology, University of PennsylvaniaPhiladelphia, USA
| | - Harshad R Kulkarni
- THERANOSTICS Center for Molecular Radiotherapy and Molecular Imaging, ENETS Center of ExcellenceZentralklinik Bad Berka, Germany
| | - Richard P Baum
- THERANOSTICS Center for Molecular Radiotherapy and Molecular Imaging, ENETS Center of ExcellenceZentralklinik Bad Berka, Germany
| |
Collapse
|
31
|
Abstract
Personalized dosimetry in radionuclide therapy has gained much attention in recent years. This attention has also an impact on peptide receptor radionuclide therapy (PRRT). This article reviews the PET-based imaging techniques that can be used for pretherapeutic prediction of doses in PRRT. More specifically the usage of (86)Y, (90)Y, (68)Ga, and (44)Sc are discussed: their characteristics for PET acquisition, the available peptides for labeling, the specifics of the imaging protocols, and the experiences gained from phantom and clinical studies. These techniques are evaluated with regard to their usefulness for dosimetry predictions in PRRT, and future perspectives are discussed.
Collapse
|
32
|
Kidney dosimetry in ¹⁷⁷Lu and ⁹⁰Y peptide receptor radionuclide therapy: influence of image timing, time-activity integration method, and risk factors. BIOMED RESEARCH INTERNATIONAL 2013; 2013:935351. [PMID: 23865075 PMCID: PMC3705840 DOI: 10.1155/2013/935351] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 05/31/2013] [Indexed: 11/17/2022]
Abstract
Kidney dosimetry in 177Lu and 90Y PRRT requires 3 to 6 whole-body/SPECT scans to extrapolate the peptide kinetics, and it is considered time and resource consuming. We investigated the most adequate timing for imaging and time-activity interpolating curve, as well as the performance of a simplified dosimetry, by means of just 1-2 scans. Finally the influence of risk factors and of the peptide (DOTATOC versus DOTATATE) is considered. 28 patients treated at first cycle with 177Lu DOTATATE and 30 with 177Lu DOTATOC underwent SPECT scans at 2 and 6 hours, 1, 2, and 3 days after the radiopharmaceutical injection. Dose was calculated with our simplified method, as well as the ones most used in the clinic, that is, trapezoids, monoexponential, and biexponential functions. The same was done skipping the 6 h and the 3 d points. We found that data should be collected until 100 h for 177Lu therapy and 70 h for 90Y therapy, otherwise the dose calculation is strongly influenced by the curve interpolating the data and should be carefully chosen.
Risk factors (hypertension, diabetes) cause a rather statistically significant 20% increase in dose (t-test, P < 0.10), with DOTATATE affecting an increase of 25% compared to DOTATOC (t-test, P < 0.05).
Collapse
|
33
|
Hobbs RF, Jentzen W, Bockisch A, Sgouros G. Monte Carlo-based 3-dimensional dosimetry of salivary glands in radioiodine treatment of differentiated thyroid cancer estimated using 124I PET. 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... 2013; 57:79-91. [PMID: 23474639 PMCID: PMC4037814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
AIM Salivary gland toxicity is of concern in radioiodine treatment of thyroid cancer. Toxicity is often observed while the estimated radiation absorbed dose (AD) values are below expected toxicity thresholds. Monte Carlo-based voxelized 3-dimensional radiobiological dosimetry (3D-RD) calculations of the salivary glands from eight metastatic thyroid cancer patients treated with 131I are presented with the objective of resolving this discrepancy. METHODS GEANT4 Monte Carlo simulations were performed for 131I, based on pretherapeutic 124I PET/CT imaging corrected for partial volume effect, and the results scaled to the therapeutic administered activities. For patients with external regions of high uptake proximal to the salivary glands, such as thyroid remnants or lymph node metastases, separate simulations were run to quantify the AD contributions from both (A) the salivary glands themselves, and (B) the external proximal region of high uptake (present for five patients). The contribution from the whole body outside the field of view was also estimated using modeling. Voxelized and average ADs and biological effective doses (BEDs) were calculated. RESULTS The estimated average therapeutic ADs were 2.26 Gy considering all contributions and 1.94 Gy from the self-dose component only. The average contribution from the external region of high uptake was 0.54 Gy. This difference was more pronounced for the submandibular glands (2.64 versus 2.10 Gy) compared to the parotid glands (1.88 Gy versus 1.78 Gy). The BED values were on average only 6.6 % higher than (2.41 Gy) the ADs. CONCLUSION The external sources of activity contribute significantly to the salivary gland AD, however neither this contribution, nor the radiobiological effect quantified by the BED are in themselves sufficient to explain the clinically observed toxicity.
Collapse
Affiliation(s)
- R F Hobbs
- Department of Radiology, Johns Hopkins University, Baltimore MD, USA
| | | | | | | |
Collapse
|
34
|
Gustafsson J, Nilsson P, Gleisner KS. On the biologically effective dose (BED)—using convolution for calculating the effects of repair: I. Analytical considerations. Phys Med Biol 2013; 58:1507-27. [DOI: 10.1088/0031-9155/58/5/1507] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
35
|
Gustafsson J, Nilsson P, Gleisner KS. On the biologically effective dose (BED)-using convolution for calculating the effects of repair: II. Numerical considerations. Phys Med Biol 2013; 58:1529-48. [PMID: 23406832 DOI: 10.1088/0031-9155/58/5/1529] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
We have previously shown analytically that the biologically effective dose (BED), including effects of repair during irradiation and of incomplete repair between fractions, can be formulated using a convolution between the absorbed dose rate function and the function describing repair. In this work, a discrete formalism is derived along with its implementation via the fast Fourier transform. The implementation takes the intrinsic periodicity of the discrete Fourier transform into consideration, as well as possible inconsistencies that may arise due to discretization and truncation of the functions describing the absorbed dose rate and repair. Numerically and analytically calculated BED values are compared for various situations in external beam radiotherapy, brachytherapy and radionuclide therapy, including the use of different repair models. The numerical method is shown to be accurate and versatile since it can be applied to any kind of absorbed dose rate function and allows for the incorporation of different repair models. Typical accuracies for clinically realistic examples are in the order of 10(-3)% to 10(-5)%. The method has thus the potential of being a useful tool for the calculation of BED, also in situations with complicated irradiation patterns or repair functions.
Collapse
Affiliation(s)
- Johan Gustafsson
- Department of Medical Radiation Physics, Lund University, Lund, Sweden.
| | | | | |
Collapse
|
36
|
Baechler S, Hobbs RF, Boubaker A, Buchegger F, He B, Frey EC, Sgouros G. Three-dimensional radiobiological dosimetry of kidneys for treatment planning in peptide receptor radionuclide therapy. Med Phys 2012; 39:6118-28. [PMID: 23039651 DOI: 10.1118/1.4752213] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
PURPOSE Peptide receptor radionuclide therapy (PRRT) delivers high absorbed doses to kidneys and may lead to permanent nephropathy. Reliable dosimetry of kidneys is thus critical for safe and effective PRRT. The aim of this work was to assess the feasibility of planning PRRT based on 3D radiobiological dosimetry (3D-RD) in order to optimize both the amount of activity to administer and the fractionation scheme, while limiting the absorbed dose and the biological effective dose (BED) to the renal cortex. METHODS Planar and SPECT data were available for a patient examined with (111)In-DTPA-octreotide at 0.5 (planar only), 4, 24, and 48 h post-injection. Absorbed dose and BED distributions were calculated for common therapeutic radionuclides, i.e., (111)In, (90)Y and (177)Lu, using the 3D-RD methodology. Dose-volume histograms were computed and mean absorbed doses to kidneys, renal cortices, and medullae were compared with results obtained using the MIRD schema (S-values) with the multiregion kidney dosimetry model. Two different treatment planning approaches based on (1) the fixed absorbed dose to the cortex and (2) the fixed BED to the cortex were then considered to optimize the activity to administer by varying the number of fractions. RESULTS Mean absorbed doses calculated with 3D-RD were in good agreement with those obtained with S-value-based SPECT dosimetry for (90)Y and (177)Lu. Nevertheless, for (111)In, differences of 14% and 22% were found for the whole kidneys and the cortex, respectively. Moreover, the authors found that planar-based dosimetry systematically underestimates the absorbed dose in comparison with SPECT-based methods, up to 32%. Regarding the 3D-RD-based treatment planning using a fixed BED constraint to the renal cortex, the optimal number of fractions was found to be 3 or 4, depending on the radionuclide administered and the value of the fixed BED. Cumulative activities obtained using the proposed simulated treatment planning are compatible with real activities administered to patients in PRRT. CONCLUSIONS The 3D-RD treatment planning approach based on the fixed BED was found to be the method of choice for clinical implementation in PRRT by providing realistic activity to administer and number of cycles. While dividing the activity in several cycles is important to reduce renal toxicity, the clinical outcome of fractionated PRRT should be investigated in the future.
Collapse
Affiliation(s)
- Sebastien Baechler
- Institute of Radiation Physics, Lausanne University Hospital, Lausanne, Switzerland.
| | | | | | | | | | | | | |
Collapse
|
37
|
Van Binnebeek S, Baete K, Terwinghe C, Vanbilloen B, Haustermans K, Mortelmans L, Borbath I, Van Cutsem E, Verslype C, Mottaghy FM, Verbruggen A, Deroose CM. Significant impact of transient deterioration of renal function on dosimetry in PRRT. Ann Nucl Med 2012; 27:74-7. [PMID: 22961123 DOI: 10.1007/s12149-012-0651-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Accepted: 08/13/2012] [Indexed: 10/27/2022]
Abstract
Peptide receptor radionuclide therapy (PRRT), with (90)Y-DOTATOC and (177)Lu-DOTATATE as most clinically used radiopeptides, is widely used in the management of metastatic neuroendocrine tumors. With respect to radiation dosimetry, the kidneys are the critical organ for (90)Y-DOTATOC. Renal irradiation is significant because of reabsorption of the radiopeptide from the proximal tubuli and the resulting retention in the interstitium, mainly in the inner cortical zone. The high energy and consequently wide range in tissue of the yttrium-90 beta particle result in high absorbed doses to the kidney cortex and medulla. Accurate renal dosimetry can help minimizing radiation nephropathy. We report a case of a 69-year-old candidate for PRRT with an acceptable kidney function at the time of screening. When performing (111)In-octreotide pretreatment dosimetry 3 weeks later, we observed a drastic deterioration in kidney function, caused by undisclosed non-steroidal anti-inflammatory drug intake. The calculated kidney biological effective dose (BED) was 153 Gy after four projected cycles. PRRT was canceled as our full-course BED limit is 37 Gy and the patient was switched to morphine analgesics. Renal function normalized after 3 months and repeated dosimetry yielded an acceptable kidney BED of 28 Gy after four projected cycles (7 Gy/cycle). This case emphasizes that acute kidney insufficiency can yield toxic kidney doses in a single therapy cycle, with an inherent risk of persistent renal insufficiency. All clinical factors which might influence kidney function should be verified at screening and before PRRT administration.
Collapse
|
38
|
Sgouros G, Hobbs RF. Patient-Specific Dosimetry, Radiobiology, and the Previously-Treated Patient. ACTA ACUST UNITED AC 2012. [DOI: 10.1007/174_2012_684] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
|
39
|
Wierts R, de Pont CD, Brans B, Mottaghy FM, Kemerink GJ. Dosimetry in molecular nuclear therapy. Methods 2011; 55:196-202. [DOI: 10.1016/j.ymeth.2011.09.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Revised: 09/11/2011] [Accepted: 09/13/2011] [Indexed: 01/06/2023] Open
|
40
|
Soundararajan A, Bao A, Phillips WT, McManus LM, Goins BA. Chemoradionuclide therapy with 186re-labeled liposomal doxorubicin: toxicity, dosimetry, and therapeutic response. Cancer Biother Radiopharm 2011; 26:603-14. [PMID: 21834653 DOI: 10.1089/cbr.2010.0948] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
This study was performed to determine the maximum tolerated dose (MTD) and therapeutic effects of rhenium-186 ((186)Re)-labeled liposomal doxorubicin (Doxil), investigate associated toxicities, and calculate radiation absorbed dose in head and neck tumor xenografts and normal organs. Doxil and control polyethylene glycol (PEG)-liposomes were labeled using (186)Re-N,N-bis(2-mercaptoethyl)-N',N'-diethylethylenediamine (BMEDA) method. Tumor-bearing rats received either no therapy (n=6), intravenous Doxil (n=4), or escalating radioactivity of (186)Re-Doxil (185-925 MBq/kg) or (186)Re-PEG-liposomes (1110-1665 MBq/kg) and were monitored for 28 days. Based on body weight loss and systemic toxicity, MTD for (186)Re-Doxil and (186)Re-PEG-liposomes were established at injected radioactivity/body weight of 740 and 1480 MBq/kg, respectively. (186)Re-injected radioactivity/body weight for therapy studies was determined to be 555 MBq/kg for (186)Re-Doxil and 1295 MBq/kg for (186)Re-PEG-liposomes. All groups recovered from their body weight loss, leucopenia, and thrombocytopenia by 28 days postinjection. Normalized radiation absorbed dose to tumor was significantly higher for (186)Re-Doxil (0.299±0.109 Gy/MBq) compared with (186)Re-PEG-liposomes (0.096±0.120 Gy/MBq) (p<0.05). In a separate therapy study, tumor volumes were significantly smaller for (186)Re-Doxil (555 MBq/kg) compared with (186)Re-PEG-liposomes (1295 MBq/kg) (p<0.01) at 42 days postinjection. In conclusion, combination chemoradionuclide therapy with (186)Re-Doxil has promising potential, because good tumor control was achieved with limited associated toxicity.
Collapse
Affiliation(s)
- Anuradha Soundararajan
- Department of Radiology, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229-3900, USA
| | | | | | | | | |
Collapse
|
41
|
Sgouros G, Hobbs RF, Atkins FB, Van Nostrand D, Ladenson PW, Wahl RL. Three-dimensional radiobiological dosimetry (3D-RD) with 124I PET for 131I therapy of thyroid cancer. Eur J Nucl Med Mol Imaging 2011; 38 Suppl 1:S41-7. [PMID: 21484384 DOI: 10.1007/s00259-011-1769-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2011] [Accepted: 02/22/2011] [Indexed: 01/08/2023]
Abstract
Radioiodine therapy of thyroid cancer was the first and remains among the most successful radiopharmaceutical (RPT) treatments of cancer although its clinical use is based on imprecise dosimetry. The positron emitting radioiodine, (124)I, in combination with positron emission tomography (PET)/CT has made it possible to measure the spatial distribution of radioiodine in tumors and normal organs at high resolution and sensitivity. The CT component of PET/CT has made it simpler to match the activity distribution to the corresponding anatomy. These developments have facilitated patient-specific dosimetry (PSD), utilizing software packages such as three-dimensional radiobiological dosimetry (3D-RD), which can account for individual patient differences in pharmacokinetics and anatomy. We highlight specific examples of such calculations and discuss the potential impact of (124)I PET/CT on thyroid cancer therapy.
Collapse
Affiliation(s)
- George Sgouros
- The Russell H. Morgan Department of Radiology, Division of Nuclear Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, USA.
| | | | | | | | | | | |
Collapse
|
42
|
Hobbs RF, McNutt T, Baechler S, He B, Esaias CE, Frey EC, Loeb DM, Wahl RL, Shokek O, Sgouros G. A treatment planning method for sequentially combining radiopharmaceutical therapy and external radiation therapy. Int J Radiat Oncol Biol Phys 2010; 80:1256-62. [PMID: 20950958 DOI: 10.1016/j.ijrobp.2010.08.022] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2009] [Revised: 08/10/2010] [Accepted: 08/23/2010] [Indexed: 11/18/2022]
Abstract
PURPOSE Effective cancer treatment generally requires combination therapy. The combination of external beam therapy (XRT) with radiopharmaceutical therapy (RPT) requires accurate three-dimensional dose calculations to avoid toxicity and evaluate efficacy. We have developed and tested a treatment planning method, using the patient-specific three-dimensional dosimetry package 3D-RD, for sequentially combined RPT/XRT therapy designed to limit toxicity to organs at risk. METHODS AND MATERIALS The biologic effective dose (BED) was used to translate voxelized RPT absorbed dose (D(RPT)) values into a normalized total dose (or equivalent 2-Gy-fraction XRT absorbed dose), NTD(RPT) map. The BED was calculated numerically using an algorithmic approach, which enabled a more accurate calculation of BED and NTD(RPT). A treatment plan from the combined Samarium-153 and external beam was designed that would deliver a tumoricidal dose while delivering no more than 50 Gy of NTD(sum) to the spinal cord of a patient with a paraspinal tumor. RESULTS The average voxel NTD(RPT) to tumor from RPT was 22.6 Gy (range, 1-85 Gy); the maximum spinal cord voxel NTD(RPT) from RPT was 6.8 Gy. The combined therapy NTD(sum) to tumor was 71.5 Gy (range, 40-135 Gy) for a maximum voxel spinal cord NTD(sum) equal to the maximum tolerated dose of 50 Gy. CONCLUSIONS A method that enables real-time treatment planning of combined RPT-XRT has been developed. By implementing a more generalized conversion between the dose values from the two modalities and an activity-based treatment of partial volume effects, the reliability of combination therapy treatment planning has been expanded.
Collapse
|
43
|
Siegel JA, Stabin MG, Sharkey RM. Renal Dosimetry in Peptide Radionuclide Receptor Therapy. Cancer Biother Radiopharm 2010; 25:581-8. [DOI: 10.1089/cbr.2010.0805] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
| | - Michael G. Stabin
- Department of Radiology and Radiological Sciences, Vanderbilt University, Nashville, Tennessee
| | - Robert M. Sharkey
- Center for Molecular Medicine and Immunology, Garden State Cancer Center, Belleville, New Jersey
| |
Collapse
|
44
|
Bouchat V, Nuttens VE, Michiels C, Masereel B, Feron O, Gallez B, Vander Borght T, Lucas S. Radioimmunotherapy with radioactive nanoparticles: biological doses and treatment efficiency for vascularized tumors with or without a central hypoxic area. Med Phys 2010; 37:1826-39. [PMID: 20443505 DOI: 10.1118/1.3368599] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
PURPOSE Radioactive atoms attached to monoclonal antibodies are used in radioimmunotherapy to treat cancer while limiting radiation to healthy tissues. One limitation of this method is that only one radioactive atom is linked to each antibody and the deposited dose is often insufficient to eradicate solid and radioresistant tumors. In a previous study, simulations with the Monte Carlo N-Particle eXtended code showed that physical doses up to 50 Gy can be delivered inside tumors by replacing the single radionuclide by a radioactive nanoparticle of 5 nm diameter containing hundreds of radioactive atoms. However, tumoral and normal tissues are not equally sensitive to radiation, and previous works did not take account the biological effects such as cellular repair processes or the presence of less radiosensitive cells such as hypoxic cells. METHODS The idea is to adapt the linear-quadratic expression to the tumor model and to determine biological effective doses (BEDs) delivered through and around a tumor. This BED is then incorporated into a Poisson formula to determine the shell control probability (SCP) which predicts the cell cluster-killing efficiency at different distances "r" from the center of the tumor. BED and SCP models are used to analyze the advantages of injecting radioactive nanoparticles instead of a single radionuclide per vector in radioimmunotherapy. RESULTS Calculations of BED and SCP for different distances r from the center of a solid tumor, using the non-small-cell lung cancer as an example, were investigated for 90Y2O3 nanoparticles. With a total activity of about 3.5 and 20 MBq for tumor radii of 0.5 and 1.0 cm, respectively, results show that a very high BED is deposited in the well oxygenated part of the spherical carcinoma. CONCLUSIONS For either small or large solid tumors, BED and SCP calculations highlight the important benefit in replacing the single beta-emitter 90Y attached to each antibody by a 90Y2O3 nanoparticle.
Collapse
Affiliation(s)
- V Bouchat
- Research Center in Physics of Matter and Radiation, Laboratoire d'Analyses par Réactions Nucléaires, University of Namur, Rue de Bruxelles 61, B-5000 Namur, Belgium.
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Hobbs RF, Baechler S, Wahl RL, He B, Song H, Esaias CE, Frey EC, Jacene H, Sgouros G. Arterial wall dosimetry for non-Hodgkin lymphoma patients treated with radioimmunotherapy. J Nucl Med 2010; 51:368-75. [PMID: 20150265 DOI: 10.2967/jnumed.109.069575] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED Tumors in non-Hodgkin lymphoma (NHL) patients are often proximal to the major blood vessels in the abdomen or neck. In external-beam radiotherapy, these tumors present a challenge because imaging resolution prevents the beam from being targeted to the tumor lesion without also irradiating the artery wall. This problem has led to potentially life-threatening delayed toxicity. Because radioimmunotherapy has resulted in long-term survival of NHL patients, we investigated whether the absorbed dose (AD) to the artery wall in radioimmunotherapy of NHL is of potential concern for delayed toxicity. SPECT resolution is not sufficient to enable dosimetric analysis of anatomic features of the thickness of the aortic wall. Therefore, we present a model of aortic wall toxicity based on data from 4 patients treated with (131)I-tositumomab. METHODS Four NHL patients with periaortic tumors were administered pretherapeutic (131)I-tositumomab. Abdominal SPECT and whole-body planar images were obtained at 48, 72, and 144 h after tracer administration. Blood-pool activity concentrations were obtained from regions of interest drawn on the heart on the planar images. Tumor and blood activity concentrations, scaled to therapeutic administered activities-both standard and myeloablative-were input into a geometry and tracking model (GEANT, version 4) of the aorta. The simulated energy deposited in the arterial walls was collected and fitted, and the AD and biologic effective dose values to the aortic wall and tumors were obtained for standard therapeutic and hypothetical myeloablative administered activities. RESULTS Arterial wall ADs from standard therapy were lower (0.6-3.7 Gy) than those typical from external-beam therapy, as were the tumor ADs (1.4-10.5 Gy). The ratios of tumor AD to arterial wall AD were greater for radioimmunotherapy by a factor of 1.9-4.0. For myeloablative therapy, artery wall ADs were in general less than those typical for external-beam therapy (9.4-11.4 Gy for 3 of 4 patients) but comparable for 1 patient (32.6 Gy). CONCLUSION Blood vessel radiation dose can be estimated using the software package 3D-RD combined with GEANT modeling. The dosimetry analysis suggested that arterial wall toxicity is highly unlikely in standard dose radioimmunotherapy but should be considered a potential concern and limiting factor in myeloablative therapy.
Collapse
|
46
|
A theoretical dose-escalation study based on biological effective dose in radioimmunotherapy with (90)Y-ibritumomab tiuxetan (Zevalin). Eur J Nucl Med Mol Imaging 2010; 37:862-73. [PMID: 20069297 DOI: 10.1007/s00259-009-1333-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2009] [Accepted: 11/10/2009] [Indexed: 12/25/2022]
Abstract
AIM To investigate the variation in biological effective dose (BED) produced by the uncertainty in absorbed dose and radiobiological parameters in Zevalin radioimmunotherapy. METHODS Eight patients scheduled for treatment with standard administration of (90)Y-ibritumomab tiuxetan (Zevalin) were studied. Patient-specific pretherapy dosimetry was performed by injection of (111)In-ibritumomab tiuxetan. Absorbed doses and BEDs were calculated for critical organs (COs) and tumours, assuming a 30% dose uncertainty and varying the radiobiological parameters in a reasonable range. In an activity-escalation study, BEDs for the COs were compared with the BED limits of external beam radiotherapy (EBRT) and BEDs for the tumour with the EBRT dose prescriptions. RESULTS At standard activities, the absorbed doses per unit activity for the COs were in agreement with those in the literature. Absorbed doses to lesions were rather variable, ranging from 1.47 to 16.7 Gy/GBq. Median tumour absorbed dose to lesions in the range 80-110 g was 9.6 Gy/GBq (range 9.2-16.7 Gy/GBq), yielding a mean BED of about 12 Gy for administration of 15 MBq/kg. For the administration of the myeloablative activity of 45 MBq/kg, risk of liver toxicity in one patient would have been foreseen by the model. Considering also the dose uncertainty, the potential risk of liver toxicity in one more patient, lung toxicity in one patient, and kidney toxicity in one patient would have been suggested. The absorbed dose uncertainty was found to be the main source of uncertainty in the BED. As for radiobiological parameters, at myeloablative activities, the increase in the repair half-time for sublethally damaged tissue (T(mu)) from 0.5 h to 5 h induced more consistent increases in mean BED/BED(limit) than alpha/beta variation from 2 Gy to 5 Gy: at 53 MBq/kg, 38% for the liver, and 34% for the lungs and kidneys (about threefold higher than that obtained for the increase alpha/beta). CONCLUSION At standard activities, absorbed doses to lesions appear to be effective, even though lower than prescribed by EBRT. At myeloablative dosages, the uncertainty associated with the absorbed doses and radiobiological parameters considerably affect BED evaluation and may account for possible "second-organ" toxicities.
Collapse
|
47
|
Hobbs RF, Sgouros G. Calculation of the biological effective dose for piecewise defined dose-rate fits. Med Phys 2009; 36:904-7. [PMID: 19378750 DOI: 10.1118/1.3070587] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
An algorithmic solution to the biological effective dose (BED) calculation from the Lea-Catcheside formula for a piecewise defined function is presented. Data from patients treated for metastatic thyroid cancer were used to illustrate the solution. The Lea-Catcheside formula for the G-factor of the BED is integrated numerically using a large number of small trapezoidal fits to each integral. The algorithmically calculated BED is compatible with an analytic calculation for a similarly valued exponentially fitted dose-rate plot and is the only resolution for piecewise defined dose-rate functions.
Collapse
Affiliation(s)
- Robert F Hobbs
- Department of Radiology, Johns Hopkins University, Baltimore, Maryland 21231, USA.
| | | |
Collapse
|
48
|
Abstract
The development of effective immunotherapy strategies for glioma requires adequate understanding of the unique immunological microenvironment in the central nervous system (CNS) and CNS tumors. Although the CNS is often considered to be an immunologically privileged site and poses unique challenges for the delivery of effector cells and molecules, recent advances in technology and discoveries in CNS immunology suggest novel mechanisms that may significantly improve the efficacy of immunotherapy against gliomas. In this review, we first summarize recent advances in the CNS and CNS tumor immunology. We address factors that may promote immune escape of gliomas. We also review advances in passive and active immunotherapy strategies for glioma, with an emphasis on lessons learned from recent early-phase clinical trials. We also discuss novel immunotherapy strategies that have been recently tested in non-CNS tumors and show great potential for application to gliomas. Finally, we discuss how each of these promising strategies can be combined to achieve clinical benefit for patients with gliomas.
Collapse
Affiliation(s)
- Hideho Okada
- Brain Tumor Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213, USA.
| | | | | | | | | | | | | |
Collapse
|
49
|
Oehme L, Kotzerke J. Radiobiological considerations for radioembolization with 188Re-microspheres. Eur J Nucl Med Mol Imaging 2008; 36:322-5. [PMID: 19018527 DOI: 10.1007/s00259-008-0991-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2008] [Accepted: 10/09/2008] [Indexed: 01/17/2023]
|
50
|
Wessels BW, Konijnenberg MW, Dale RG, Breitz HB, Cremonesi M, Meredith RF, Green AJ, Bouchet LG, Brill AB, Bolch WE, Sgouros G, Thomas SR. MIRD Pamphlet No. 20: The Effect of Model Assumptions on Kidney Dosimetry and Response—Implications for Radionuclide Therapy. J Nucl Med 2008; 49:1884-99. [PMID: 18927342 DOI: 10.2967/jnumed.108.053173] [Citation(s) in RCA: 121] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Affiliation(s)
- Barry W Wessels
- Department of Radiation Oncology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|