1
|
Cicone F, Gnesin S, Santo G, Stokke C, Bartolomei M, Cascini GL, Minniti G, Paganelli G, Verger A, Cremonesi M. Do we need dosimetry for the optimization of theranostics in CNS tumors? Neuro Oncol 2024; 26:S242-S258. [PMID: 39351795 PMCID: PMC11631076 DOI: 10.1093/neuonc/noae200] [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] [Indexed: 12/12/2024] Open
Abstract
Radiopharmaceutical theranostic treatments have grown exponentially worldwide, and internal dosimetry has attracted attention and resources. Despite some similarities with chemotherapy, radiopharmaceutical treatments are essentially radiotherapy treatments, as the release of radiation into tissues is the determinant of the observed clinical effects. Therefore, absorbed dose calculations are key to explaining dose-effect correlations and individualizing radiopharmaceutical treatments. The present article introduces the basic principles of internal dosimetry and provides an overview of available loco-regional and systemic radiopharmaceutical treatments for central nervous system (CNS) tumors. The specific characteristics of dosimetry as applied to these treatments are highlighted, along with their limitations and most relevant results. Dosimetry is performed with higher precision and better reproducibility than in the past, and dosimetric data should be systematically collected, as treatment planning and verification may help exploit the full potential of theranostic of CNS tumors.
Collapse
Affiliation(s)
- Francesco Cicone
- Nuclear Medicine Unit, Department of Experimental and Clinical Medicine, “Magna Graecia” University of Catanzaro, Catanzaro, Italy
| | - Silvano Gnesin
- Institute of Radiation Physics, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Giulia Santo
- Nuclear Medicine Unit, Department of Experimental and Clinical Medicine, “Magna Graecia” University of Catanzaro, Catanzaro, Italy
| | - Caroline Stokke
- Department of Physics, University of Oslo, Oslo, Norway
- Department of Diagnostic Physics and Computational Radiology, Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Mirco Bartolomei
- Nuclear Medicine Unit, Department of Oncology and Haematology, Azienda Ospedaliero-Universitaria di Ferrara, Ferrara, Italy
| | - Giuseppe Lucio Cascini
- Nuclear Medicine Unit, Department of Experimental and Clinical Medicine, “Magna Graecia” University of Catanzaro, Catanzaro, Italy
| | - Giuseppe Minniti
- IRCCS Neuromed, Pozzilli (IS), Italy
- Radiation Oncology Unit, Department of Radiological Sciences, Oncology and Anatomical Pathology, “Sapienza” University of Rome, Rome, Italy
| | - Giovanni Paganelli
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori,”Meldola, Italy
| | - Antoine Verger
- Department of Nuclear Medicine and Nancyclotep Imaging Platform, CHRU-Nancy, IADI, INSERM, UMR 1254, Université de Lorraine, Nancy, France
| | - Marta Cremonesi
- Unit of Radiation Research, IEO, European Institute of Oncology IRCCS, Milan, Italy
| |
Collapse
|
2
|
Maroufpour S, Aryana K, Nasseri S, Fazeli Z, Arabi H, Momennezhad M. Validation of dosimetry programs (Olinda & IDAC) for evaluation of absorbed dose in 177LuPSMA therapy of metastatic castration-resistant prostate cancer (mCRPC) using Monte Carlo simulation. EJNMMI Phys 2024; 11:102. [PMID: 39623240 PMCID: PMC11612135 DOI: 10.1186/s40658-024-00691-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 10/10/2024] [Indexed: 12/06/2024] Open
Abstract
PURPOSE Clinical trials have yielded promising results for 177Lutetium Prostate Specific Membrane Antigen (177Lu-PSMA) therapy in metastatic castration resistant prostate cancer (mCRPC) patients. However, the development of precise methods for internal dosimetry and accurate dose estimation has been considered ongoing research. This study aimed to calculate the absorbed dose to the critical organs and metastasis regions using GATE 9.0 Monte Carlo simulation (MCS) as a gold standard to compare the OLINDA 1.1 and IDAC 2.1 software. MATERIAL AND METHODS This study investigated absorbed doses to different organs in 9 mCRPC patients during their first treatment cycle. Whole-body planar images were acquired at 1 ± 0.5, 4 ± 0.5, 24 ± 2, 48 ± 2, 72 ± 2, and 144 ± 2 h post-injection, with SPECT/CT images obtained at 24 ± 2 h. Absorbed doses were calculated for five organs and the entire metastasis regions using GATE, OLINDA, and IDAC platforms. The spherical method was used to determine and compare the absorbed doses in metastatic regions and undefined organs in OLINDA and IDAC Phantom. RESULTS The organ-absorbed dose calculations produced by GATE were consistent with those obtained from OLINDA and IDAC. The average percentage differences in absorbed dose for all organs between Monte Carlo calculations and the estimated from IDAC and OLINDA were -0.24 ± 2.14% and 5.16 ± 5.66%, respectively. There was a significant difference between GATE and both IDAC (17.55 ± 29.1%) and OLINDA (25.86 ± 18.04%) in determining absorbed doses to metastatic areas using the spherical model. CONCLUSION The absorbed dose of organs in the first treatment cycle remained below tolerable limits. However, cumulative absorbed doses should be considered for the administered activities in the next cycles of treatment. While Monte Carlo, IDAC, and OLINDA results were aligned for organ dose calculations, patient-specific dosimetry may be necessary due to anatomical and functional changes. Accurate dose estimation for undefined organs and metastatic regions using the spherical model is significantly influenced by tissue density, highlighting the value of CT imaging.
Collapse
Affiliation(s)
- Sirwan Maroufpour
- Medical Physics Group, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Kamran Aryana
- Nuclear Medicine Research Center, Faculty of Medicine, Imam Reza Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Shahrokh Nasseri
- Medical Physics Group, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Fazeli
- Nuclear Medicine Research Center, Faculty of Medicine, Imam Reza Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Arabi
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, Geneva, Switzerland
| | - Mehdi Momennezhad
- Medical Physics Group, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
3
|
Pandey S, Kaur G, Rana N, Chopra S, Rather I, Kumar R, Laroiya I, Chadha VD, Satz S, Stabin MG, Mittal BR, Shukla J. Advancing Cancer Theranostics Through Integrin αVβ3-Targeted Peptidomimetic IAC: From Bench to Bedside. Cancer Biother Radiopharm 2024; 39:632-643. [PMID: 38977419 DOI: 10.1089/cbr.2023.0140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/10/2024] Open
Abstract
Introduction: The expression of alpha-five beta-three (αVβ3) integrins is upregulated in various malignancies undergoing angiogenesis. The development of integrin antagonists as diagnostic probes makes the αVβ3 integrin a suitable candidate for targeting tumor angiogenesis. The goal of this study was to optimize the radiolabeling and evaluate the potential of conjugated integrin antagonist carbamate (IAC), a peptidomimetic, as a theranostic radiopharmaceutical for targeting tumor angiogenesis. Methodology: Radiolabeling of DOTAGA [2,2',2"-{10-(2,6-dioxotetrahydro-2H-pyran-3-yl)-1,4,7,10-tetraazacyclododecane-1,4,7-triyl} triacetic-acid]-IAC with [68Ga]Ga, [177Lu]Lu, and [225Ac]Ac was optimized. The binding affinity (Kd) of DOTAGA-IAC for the αVβ3 receptor and cancer cell lines was quantified. The biodistribution studies were conducted in healthy Wistar rats. Dosimetry analysis was performed on [177Lu]Lu-DOTAGA-IAC distribution data. A pilot study of [68Ga]Ga-DOTAGA-IAC and [18F]FDG Positron Emission Tomography (PET/CT) imaging was performed in five patients with histopathologically confirmed breast cancer. PET/CT findings were compared between [68Ga]Ga-DOTAGA-IAC and [18F]FDG in these patients. Results: Radiopharmaceuticals were prepared with high radiochemical purity (>99.9%). Kd and Bmax measurements were 15.02 nM and 417 fmol for αVβ3 receptor protein: 115.7 nM and 295.3 fmol for C6 glioma cells. Biodistribution studies in rats suggested the excretion via kidneys and partially through the hepatobiliary route. The effective dose of [177Lu]Lu-DOTAGA-IAC was found to be 0.17 mSv/MBq. The dynamic study in patients revealed the optimal imaging time to be 30-35 mins postadministration. Out of the cohort, [68Ga]Ga-DOTAGA-IAC detected the primary lesions in all five patients with a mean standard uptake value (SUVmax) of 3.94 ± 0.58 compared with [18F]FDG (SUVmax 13.8 ± 6.53). Conclusion: The study demonstrates that DOTAGA-IAC exhibits strong binding to αVβ3 integrin, positioning it as a promising PET agent for assessing primary and metastatic cancers. The outcomes from the pilot study suggest the potential of [68Ga]Ga-DOTAGA-IAC PET/CT in breast carcinoma diagnosis. While recognizing the theranostic potential of DOTAGA-IAC for αVβ3 integrin-expressing tumors, further clinical investigations are warranted to comprehensively assess therapeutic efficacy.
Collapse
Affiliation(s)
- Somit Pandey
- Department of Nuclear Medicine, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, India
| | - Gurvinder Kaur
- Department of Nuclear Medicine, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, India
| | - Nivedita Rana
- Department of Nuclear Medicine, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, India
| | - Sejal Chopra
- Department of Nuclear Medicine, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, India
| | - Imran Rather
- Department of Pharmacology, Post Graduate Institute of Medical Education & Research (P.G.I.M.E.R), Chandigarh, India
| | - Rajender Kumar
- Department of Nuclear Medicine, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, India
| | - Ishita Laroiya
- Department of Surgery, Post Graduate Institute of Medical Education & Research (P.G.I.M.E.R), Chandigarh, India
| | - Vijayta D Chadha
- Center for Nuclear Medicine, Panjab University, Chandigarh, India
| | - Stanley Satz
- Advanced Innovative Partners, Inc., Miami, Florida, USA
| | | | - Bhagwant Rai Mittal
- Department of Nuclear Medicine, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, India
| | - Jaya Shukla
- Department of Nuclear Medicine, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, India
| |
Collapse
|
4
|
Kuznetsov M, Adhikarla V, Caserta E, Wang X, Shively JE, Pichiorri F, Rockne RC. Mathematical Modeling Unveils Optimization Strategies for Targeted Radionuclide Therapy of Blood Cancers. CANCER RESEARCH COMMUNICATIONS 2024; 4:2955-2967. [PMID: 39466073 PMCID: PMC11562018 DOI: 10.1158/2767-9764.crc-24-0306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 08/17/2024] [Accepted: 10/23/2024] [Indexed: 10/29/2024]
Abstract
SIGNIFICANCE Mathematical modeling yields general principles for optimization of TRT in mouse models of multiple myeloma that can be extrapolated to other cancer models and clinical settings.
Collapse
Affiliation(s)
- Maxim Kuznetsov
- Department of Computational and Quantitative Medicine, Beckman Research Institute, City of Hope National Medical Center, Duarte, California
| | - Vikram Adhikarla
- Department of Computational and Quantitative Medicine, Beckman Research Institute, City of Hope National Medical Center, Duarte, California
| | - Enrico Caserta
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope National Medical Center, Duarte, California
| | - Xiuli Wang
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, California
| | - John E. Shively
- Department of Immunology and Theranostics, Beckman Research Institute, City of Hope National Medical Center, Duarte, California
| | - Flavia Pichiorri
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope National Medical Center, Duarte, California
| | - Russell C. Rockne
- Department of Computational and Quantitative Medicine, Beckman Research Institute, City of Hope National Medical Center, Duarte, California
| |
Collapse
|
5
|
Jia X, Carter BW, Duffton A, Harris E, Hobbs R, Li H. Advancing the Collaboration Between Imaging and Radiation Oncology. Semin Radiat Oncol 2024; 34:402-417. [PMID: 39271275 PMCID: PMC11407744 DOI: 10.1016/j.semradonc.2024.07.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
Abstract
The fusion of cutting-edge imaging technologies with radiation therapy (RT) has catalyzed transformative breakthroughs in cancer treatment in recent decades. It is critical for us to review our achievements and preview into the next phase for future synergy between imaging and RT. This paper serves as a review and preview for fostering collaboration between these two domains in the forthcoming decade. Firstly, it delineates ten prospective directions ranging from technological innovations to leveraging imaging data in RT planning, execution, and preclinical research. Secondly, it presents major directions for infrastructure and team development in facilitating interdisciplinary synergy and clinical translation. We envision a future where seamless integration of imaging technologies into RT will not only meet the demands of RT but also unlock novel functionalities, enhancing accuracy, efficiency, safety, and ultimately, the standard of care for patients worldwide.
Collapse
Affiliation(s)
- Xun Jia
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, Baltimore, MD..
| | - Brett W Carter
- Department of Thoracic Imaging, Division of Diagnostic Imaging, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Aileen Duffton
- Beatson West of Scotland Cancer Centre, Glasgow, UK.; Institute of Cancer Science, University of Glasgow, UK
| | - Emma Harris
- Division of Radiotherapy and Imaging, Institute of Cancer Research, London, UK
| | - Robert Hobbs
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, Baltimore, MD
| | - Heng Li
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, Baltimore, MD
| |
Collapse
|
6
|
Vaughn BA, Lee SG, Vargas DB, Seo S, Rinne SS, Xu H, Guo HF, Le Roux AB, Gajecki L, Krebs S, Yang G, Ouerfelli O, Zanzonico PB, Fung EK, St Jean S, Carrasco SE, Jungbluth A, Cheung NKV, Larson SM, Veach DR, Cheal SM. Theranostic GPA33-Pretargeted Radioimmunotherapy of Human Colorectal Carcinoma with a Bivalent 177Lu-Labeled Radiohapten. J Nucl Med 2024; 65:1611-1618. [PMID: 39168519 PMCID: PMC11448610 DOI: 10.2967/jnumed.124.267685] [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: 02/28/2024] [Accepted: 07/10/2024] [Indexed: 08/23/2024] Open
Abstract
Radiolabeled small-molecule DOTA-haptens can be combined with antitumor/anti-DOTA bispecific antibodies (BsAbs) for pretargeted radioimmunotherapy (PRIT). For optimized delivery of the theranostic γ- and β-emitting isotope 177Lu with DOTA-based PRIT (DOTA-PRIT), bivalent Gemini (DOTA-Bn-thiourea-PEG4-thiourea-Bn-DOTA, aka (3,6,9,12-tetraoxatetradecane-1,14-diyl)bis(DOTA-benzyl thiourea)) was developed. Methods: Gemini was synthesized by linking 2 S-2-(4-isothiocyanatobenzyl)-DOTA molecules together via a 1,14-diamino-PEG4 linker. [177Lu]Lu-Gemini was prepared with no-carrier-added 177LuCl3 to a molar-specific activity of 123 GBq/μmol and radiochemical purity of more than 99%. The specificity of BsAb-177Lu-Gemini was verified in vitro. Subsequently, we evaluated biodistribution and whole-body clearance for [177Lu]Lu-Gemini and, for comparison, our gold-standard monovalent [177Lu]Lu-S-2-(4-aminobenzyl)-DOTA ([177Lu]Lu-DOTA-Bn) in naïve (tumor-free) athymic nude mice. For our proof-of-concept system, a 3-step pretargeting approach was performed with an established DOTA-PRIT regimen (anti-GPA33/anti-DOTA IgG-scFv BsAb, a clearing agent, and [177Lu]Lu-Gemini) in mouse models. Results: Initial in vivo studies showed that [177Lu]Lu-Gemini behaved similarly to [177Lu]Lu-DOTA-Bn, with almost identical blood and whole-body clearance kinetics, as well as biodistribution and mouse kidney dosimetry. Pretargeting [177Lu]Lu-Gemini to GPA33-expressing SW1222 human colorectal xenografts was highly effective, leading to absorbed doses of [177Lu]Lu-Gemini for blood, tumor, liver, spleen, and kidneys of 3.99, 455, 6.93, 5.36, and 14.0 cGy/MBq, respectively. Tumor-to-normal tissue absorbed-dose ratios (i.e., therapeutic indices [TIs]) for the blood and kidneys were 114 and 33, respectively. In addition, we demonstrate that the use of bivalent [177Lu]Lu-Gemini in DOTA-PRIT leads to improved TIs and augmented [177Lu]Lu-Gemini tumor uptake and retention in comparison to monovalent [177Lu]Lu-DOTA-Bn. Finally, we established efficacy in SW1222 tumor-bearing mice, demonstrating that a single injection of anti-GPA33 DOTA-PRIT with 44 MBq (1.2 mCi) of [177Lu]Lu-Gemini (estimated tumor-absorbed dose, 200 Gy) induced complete responses in 5 of 5 animals and a histologic cure in 2 of 5 (40%) animals. Moreover, a significant increase in survival compared with nontreated controls was noted (maximum tolerated dose not reached). Conclusion: We have developed a bivalent DOTA-radiohapten, [177Lu]Lu-Gemini, that showed improved radiopharmacology for DOTA-PRIT application. The use of bivalent [177Lu]Lu-Gemini in DOTA-PRIT, as opposed to monovalent [177Lu]Lu-DOTA-Bn, allows curative treatments with considerably less administered 177Lu activity while still achieving high TIs for both the blood (>100) and the kidneys (>30).
Collapse
Affiliation(s)
- Brett A Vaughn
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, New York, New York
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sang-Gyu Lee
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Daniela Burnes Vargas
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, New York, New York
| | - Shin Seo
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sara S Rinne
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, New York, New York
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Hong Xu
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Hong-Fen Guo
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Alexandre B Le Roux
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Leah Gajecki
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Simone Krebs
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiology, Weill Cornell Medicine, New York, New York
| | - Guangbin Yang
- Organic Synthesis Core Facility, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ouathek Ouerfelli
- Organic Synthesis Core Facility, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Pat B Zanzonico
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Edward K Fung
- Department of Medical Physics, Weill Cornell Medicine, New York, New York
| | - Samantha St Jean
- Laboratory of Comparative Pathology, Memorial Sloan Kettering Cancer Center-Weill Cornell Medicine-Rockefeller University, New York, New York; and
| | - Sebastian E Carrasco
- Laboratory of Comparative Pathology, Memorial Sloan Kettering Cancer Center-Weill Cornell Medicine-Rockefeller University, New York, New York; and
| | - Achim Jungbluth
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nai Kong V Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Steven M Larson
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Darren R Veach
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiology, Weill Cornell Medicine, New York, New York
| | - Sarah M Cheal
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, New York, New York;
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
7
|
Quintana J, Carlson JCT, Scott E, Ng TSC, Miller MA, Weissleder R. Scission-Enhanced Molecular Imaging (SEMI). Bioconjug Chem 2024; 35. [PMID: 39255972 PMCID: PMC11488501 DOI: 10.1021/acs.bioconjchem.4c00337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 07/23/2024] [Accepted: 07/23/2024] [Indexed: 09/12/2024]
Abstract
Positron emission tomography (PET) imaging methods have advanced our understanding of human biology, while targeted radiotherapeutic drug treatments are now routinely used clinically. The field is expected to grow considerably based on an expanding repertoire of available affinity ligands, radionuclides, conjugation chemistries, and their FDA approvals. With this increasing use, strategies for dose reduction have become of high interest to protect patients from unnecessary and off-target toxicity. Here, we describe a simple and powerful method, scission-enhanced molecular imaging (SEMI). The technique allows for rapid corporeal elimination of radionuclides once imaging or theranostic treatment is completed and relies on "click-to-release" bioorthogonal linkers.
Collapse
Affiliation(s)
- Jeremy
M. Quintana
- Center
for Systems Biology, Massachusetts General
Hospital, 185 Cambridge Street, CPZN 5206, Boston, Massachusetts 02114, United States
- Department
of Radiology, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
| | - Jonathan C. T. Carlson
- Center
for Systems Biology, Massachusetts General
Hospital, 185 Cambridge Street, CPZN 5206, Boston, Massachusetts 02114, United States
- Cancer
Center, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
| | - Ella Scott
- Center
for Systems Biology, Massachusetts General
Hospital, 185 Cambridge Street, CPZN 5206, Boston, Massachusetts 02114, United States
| | - Thomas S. C. Ng
- Center
for Systems Biology, Massachusetts General
Hospital, 185 Cambridge Street, CPZN 5206, Boston, Massachusetts 02114, United States
- Department
of Radiology, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
| | - Miles A. Miller
- Center
for Systems Biology, Massachusetts General
Hospital, 185 Cambridge Street, CPZN 5206, Boston, Massachusetts 02114, United States
- Department
of Radiology, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
| | - Ralph Weissleder
- Center
for Systems Biology, Massachusetts General
Hospital, 185 Cambridge Street, CPZN 5206, Boston, Massachusetts 02114, United States
- Cancer
Center, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
- Department
of Radiology, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
- Department
of Systems Biology, Harvard Medical School, 200 Longwood Avenue, Boston, Massachusetts 02115, United States
| |
Collapse
|
8
|
Mukkamala R, Carlson DJ, Miller NK, Lindeman SD, Bowen ER, Tudi P, Schleinkofer T, Booth OC, Cox A, Srinivasarao M, Low PS. Design of a Fibroblast Activation Protein-Targeted Radiopharmaceutical Therapy with High Tumor-to-Healthy-Tissue Ratios. J Nucl Med 2024; 65:1257-1263. [PMID: 38871387 DOI: 10.2967/jnumed.124.267756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 05/13/2024] [Indexed: 06/15/2024] Open
Abstract
Because of upregulated expression on cancer-associated fibroblasts, fibroblast activation protein (FAP) has emerged as an attractive biomarker for the imaging and therapy of solid tumors. Although many FAP ligands have already been developed for radiopharmaceutical therapies (RPTs), most suffer from inadequate tumor uptake, insufficient tumor residence times, or off-target accumulation in healthy tissues, suggesting a need for further improvements. Methods: A new FAP-targeted RPT with a novel ligand (FAP8-PEG3-IP-DOTA) was designed by combining the desirable features of several previous ligand-targeted RPTs. Uptake and retention of [111In]In or [177Lu]Lu-FAP8-PEG3-IP-DOTA were assessed in KB, HT29, MDA-MB-231, and 4T1 murine tumor models by radioimaging or ex vivo biodistribution analyses. Radiotherapeutic potencies and gross toxicities were also investigated by monitoring tumor growth, body weight, and tissue damage in tumor-bearing mice. Results: FAP8-PEG3-IP-DOTA exhibited high affinity (half-maximal inhibitory concentration, 1.6 nM) and good selectivity for FAP relative to its closest homologs, prolyl oligopeptidase (half-maximal inhibitory concentration, ∼14.0 nM) and dipeptidyl peptidase-IV (half-maximal inhibitory concentration, ∼860 nM). SPECT/CT scans exhibited high retention in 2 different solid tumor models and minimal uptake in healthy tissues. Quantitative biodistribution analyses revealed tumor-to-healthy-tissue ratios of more than 5 times for all major organs, and live animal studies demonstrated 65%-93% suppression of tumor growth in all 4 models tested, with minimal or no evidence of systemic toxicity. Conclusion: We conclude that [177Lu]Lu-FAP8-PEG3-IP-DOTA constitutes a promising and safe RPT candidate for FAPα-targeted radionuclide therapy of solid tumors.
Collapse
Affiliation(s)
- Ramesh Mukkamala
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana; and
| | - Daniel J Carlson
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana; and
| | - Nicholas Kaine Miller
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana; and
| | - Spencer D Lindeman
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana; and
| | - Emily Renee Bowen
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana; and
| | - Pooja Tudi
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana; and
| | - Taylor Schleinkofer
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana; and
| | - Owen C Booth
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana; and
| | - Abigail Cox
- Department of Comparative Pathobiology, Purdue College of Veterinary Medicine, West Lafayette, Indiana
| | - Madduri Srinivasarao
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana; and
| | - Philip S Low
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana; and
| |
Collapse
|
9
|
Lindeman SD, Booth OC, Tudi P, Schleinkofer TC, Moss JN, Kearney NB, Mukkamala R, Thompson LK, Modany MA, Srinivasarao M, Low PS. FAP Radioligand Linker Optimization Improves Tumor Dose and Tumor-to-Healthy Organ Ratios in 4T1 Syngeneic Model. J Med Chem 2024; 67:11827-11840. [PMID: 39013156 DOI: 10.1021/acs.jmedchem.4c00448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
Fibroblast activation protein (FAP) has attracted considerable attention as a possible target for the radiotherapy of solid tumors. Unfortunately, initial efforts to treat solid tumors with FAP-targeted radionuclides have yielded only modest clinical responses, suggesting that further improvements in the molecular design of FAP-targeted radiopharmaceutical therapies (RPT) are warranted. In this study, we report several advances on the previously described FAP6 radioligand that increase tumor retention and accelerate healthy tissue clearance. Seven FAP6 derivatives with different linkers or albumin binders were synthesized, radiolabeled, and investigated for their effects on binding and cellular uptake. The radioligands were then characterized in 4T1 tumor-bearing Balb/c mice using both single-photon emission computed tomography (SPECT) and ex vivo biodistribution analyses to identify the conjugate with the best tumor retention and tumor-to-healthy organ ratios. The results reveal an optimized FAP6 radioligand that exhibits efficacy and safety properties that potentially justify its translation into the clinic.
Collapse
Affiliation(s)
- Spencer D Lindeman
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
- MorphImmune, Inc., 1281 Win Hentschel Blvd, West Lafayette, Indiana 47906, United States
| | - Owen C Booth
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Pooja Tudi
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Taylor C Schleinkofer
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Jackson N Moss
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Nicholas B Kearney
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Ramesh Mukkamala
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Lauren K Thompson
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Mollie A Modany
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Madduri Srinivasarao
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Philip S Low
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
- MorphImmune, Inc., 1281 Win Hentschel Blvd, West Lafayette, Indiana 47906, United States
| |
Collapse
|
10
|
Adam DP, Grudzinski JJ, Marsh IR, Hill PM, Cho SY, Bradshaw TJ, Longcor J, Burr A, Bruce JY, Harari PM, Bednarz BP. Voxel-Level Dosimetry for Combined Iodine 131 Radiopharmaceutical Therapy and External Beam Radiation Therapy Treatment Paradigms for Head and Neck Cancer. Int J Radiat Oncol Biol Phys 2024; 119:1275-1284. [PMID: 38367914 DOI: 10.1016/j.ijrobp.2024.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 12/20/2023] [Accepted: 02/08/2024] [Indexed: 02/19/2024]
Abstract
PURPOSE Targeted radiopharmaceutical therapy (RPT) in combination with external beam radiation therapy (EBRT) shows promise as a method to increase tumor control and mitigate potential high-grade toxicities associated with re-treatment for patients with recurrent head and neck cancer. This work establishes a patient-specific dosimetry framework that combines Monte Carlo-based dosimetry from the 2 radiation modalities at the voxel level using deformable image registration (DIR) and radiobiological constructs for patients enrolled in a phase 1 clinical trial combining EBRT and RPT. METHODS AND MATERIALS Serial single-photon emission computed tomography (SPECT)/computed tomography (CT) patient scans were performed at approximately 24, 48, 72, and 168 hours postinjection of 577.2 MBq/m2 (15.6 mCi/m2) CLR 131, an iodine 131-containing RPT agent. Using RayStation, clinical EBRT treatment plans were created with a treatment planning CT (TPCT). SPECT/CT images were deformably registered to the TPCT using the Elastix DIR module in 3D Slicer software and assessed by measuring mean activity concentrations and absorbed doses. Monte Carlo EBRT dosimetry was computed using EGSnrc. RPT dosimetry was conducted using RAPID, a GEANT4-based RPT dosimetry platform. Radiobiological metrics (biologically effective dose and equivalent dose in 2-Gy fractions) were used to combine the 2 radiation modalities. RESULTS The DIR method provided good agreement for the activity concentrations and calculated absorbed dose in the tumor volumes for the SPECT/CT and TPCT images, with a maximum mean absorbed dose difference of -11.2%. Based on the RPT absorbed dose calculations, 2 to 4 EBRT fractions were removed from patient EBRT treatments. For the combined treatment, the absorbed dose to target volumes ranged from 57.14 to 75.02 Gy. When partial volume corrections were included, the mean equivalent dose in 2-Gy fractions to the planning target volume from EBRT + RPT differed -3.11% to 1.40% compared with EBRT alone. CONCLUSIONS This work demonstrates the clinical feasibility of performing combined EBRT + RPT dosimetry on TPCT scans. Dosimetry guides treatment decisions for EBRT, and this work provides a bridge for the same paradigm to be implemented within the rapidly emerging clinical RPT space.
Collapse
Affiliation(s)
- David P Adam
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Joseph J Grudzinski
- Department of Radiology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin
| | - Ian R Marsh
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Patrick M Hill
- Department of Human Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin
| | - Steve Y Cho
- Department of Radiology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin; University of Wisconsin Carbone Cancer Center, Madison, Wisconsin
| | - Tyler J Bradshaw
- Department of Radiology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin
| | | | - Adam Burr
- Department of Human Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin; University of Wisconsin Carbone Cancer Center, Madison, Wisconsin
| | - Justine Y Bruce
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin; Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin
| | - Paul M Harari
- Department of Human Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin; University of Wisconsin Carbone Cancer Center, Madison, Wisconsin
| | - Bryan P Bednarz
- Department of Medical Physics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin.
| |
Collapse
|
11
|
Song H, Leonio MI, Ferri V, Duan H, Aparici CM, Davidzon G, Franc BL, Moradi F, Shah J, Bergstrom CP, Fan AC, Shah S, Khaki AR, Srinivas S, Iagaru A. Same-day post-therapy imaging with a new generation whole-body digital SPECT/CT in assessing treatment response to [ 177Lu]Lu-PSMA-617 in metastatic castration-resistant prostate cancer. Eur J Nucl Med Mol Imaging 2024; 51:2784-2793. [PMID: 38635050 DOI: 10.1007/s00259-024-06718-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 04/15/2024] [Indexed: 04/19/2024]
Abstract
PURPOSE Lutetium-177 [177Lu]Lu-PSMA-617 radioligand therapy (RLT) represents a significant advancement for metastatic castration-resistant prostate cancer (mCRPC), demonstrating improvements in radiographic progression free survival (rPFS) and overall survival (OS) with a low rate of associated side effects. Currently, most post-therapy SPECT/CT is conducted at 24 h after infusion. This study examines the clinical utility of a next-generation multi-detector Cadmium-Zinc-Telluride (CZT) SPECT/CT system (StarGuide) in same-day post-infusion assessment and early treatment response to [177Lu]Lu-PSMA-617. METHODS In this retrospective study, 68 men with progressive mCRPC treated with [177Lu]Lu-PSMA-617 at our center from June 2022 to June 2023 were evaluated. Digital whole-body SPECT/CT imaging was performed after [177Lu]Lu-PSMA-617infusion (mean ± SD: 1.8 ± 0.6 h, range 1.1-4.9 h). Quantitative analysis of [177Lu]Lu-PSMA-617 positive lesions was performed in patients who underwent at least 2 post-therapy SPECT/CT, using liver parenchyma uptake as reference. Metrics including [177Lu]Lu-PSMA-617 positive total tumor volume (Lu-TTV), SUVmax and SUVmean were calculated. These quantitative metrics on post-infusion SPECT/CT images after cycles 1, 2 and 3 were correlated with overall survival (OS), prostate specific antigen-progression free survival (PSA-PFS) as defined by prostate cancer working group 3 (PCWG3), and PSA decrease over 50% (PSA50) response rates. RESULTS 56 patients (means age 76.2 ± 8.1 years, range: 60-93) who underwent at least 2 post-therapy SPECT/CT were included in the image analysis. The whole-body SPECT/CT scans (~ 12 min per scan) were well tolerated, with 221 same-day scans performed (89%). At a median of 10-months follow-up, 33 (58.9%) patients achieved PSA50 after [177Lu]Lu-PSMA-617 treatment and median PSA-PFS was 5.0 months (range: 1.0-15 months) while median OS was not reached. Quantitative analysis of SPECT/CT images showed that 37 patients (66%) had > 30% reduction in Lu-TTV, associated with significantly improved overall survival (median not reached vs. 6 months, P = 0.008) and PSA-PFS (median 6 months vs. 1 months, P < 0.001). However, changes in SUVmax or SUVmean did not correlate with PSA-PFS or OS. CONCLUSION We successfully implemented same-day post-therapy SPECT/CT after [177Lu]Lu-PSMA-617 infusions. Quantitation of 1-2 h post-therapy SPECT/CT images is a promising method for assessing treatment response. However, the approach is currently limited by its suboptimal detection of small tumor lesions and the necessity of incorporating a third-cycle SPECT/CT to mitigate the effects of any potential treatment-related flare-up. Further investigation in a larger patient cohort and prospective validation is essential to confirm these findings and to explore the role of SPECT/CT as a potential adjunct to PSMA PET/CT in managing mCRPC.
Collapse
Affiliation(s)
- Hong Song
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, 300 Pasteur Drive, H2200, Stanford, CA, 94305, USA
| | - Maria Isabel Leonio
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, 300 Pasteur Drive, H2200, Stanford, CA, 94305, USA
| | - Valentina Ferri
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, 300 Pasteur Drive, H2200, Stanford, CA, 94305, USA
| | - Heying Duan
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, 300 Pasteur Drive, H2200, Stanford, CA, 94305, USA
| | - Carina Mari Aparici
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, 300 Pasteur Drive, H2200, Stanford, CA, 94305, USA
| | - Guido Davidzon
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, 300 Pasteur Drive, H2200, Stanford, CA, 94305, USA
| | - Benjamin L Franc
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, 300 Pasteur Drive, H2200, Stanford, CA, 94305, USA
| | - Farshad Moradi
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, 300 Pasteur Drive, H2200, Stanford, CA, 94305, USA
| | - Jagruti Shah
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, 300 Pasteur Drive, H2200, Stanford, CA, 94305, USA
| | - Colin P Bergstrom
- Department of Medicine, Division of Oncology, Stanford University, Stanford, CA, 94305, USA
| | - Alice C Fan
- Department of Medicine, Division of Oncology, Stanford University, Stanford, CA, 94305, USA
| | - Sumit Shah
- Department of Medicine, Division of Oncology, Stanford University, Stanford, CA, 94305, USA
| | - Ali Raza Khaki
- Department of Medicine, Division of Oncology, Stanford University, Stanford, CA, 94305, USA
| | - Sandy Srinivas
- Department of Medicine, Division of Oncology, Stanford University, Stanford, CA, 94305, USA
| | - Andrei Iagaru
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, 300 Pasteur Drive, H2200, Stanford, CA, 94305, USA.
| |
Collapse
|
12
|
Yadav S, Jiang F, Kurkowska S, Saelee R, Morley A, Feng F, Aggarwal R, Lawhn-Heath C, Uribe C, Hope TA. Assessing Response to PSMA Radiopharmaceutical Therapies with Single SPECT Imaging at 24 Hours After Injection. J Nucl Med 2024; 65:1064-1069. [PMID: 38724282 DOI: 10.2967/jnumed.123.267208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 04/08/2024] [Indexed: 07/03/2024] Open
Abstract
Understanding the relationship between lesion-absorbed dose and tumor response in 177Lu-PSMA-617 radiopharmaceutical therapies (RPTs) remains complex. We aimed to investigate whether baseline lesion-absorbed dose can predict lesion-based responses and to explore the connection between lesion-absorbed dose and prostate-specific antigen (PSA) response. Methods: In this retrospective study, we evaluated 50 patients with 335 index lesions undergoing 177Lu-PSMA-617 RPT, who had dosimetry analysis performed on SPECT/CT at 24 h after cycles 1 and 2. First, we identified the index lesions for each patient and measured the lesion-based absorbed doses. Lesion-based response was calculated after cycle 2. Additionally, PSA50 response (a decline of 50% from baseline PSA) after cycle 2 was also calculated. The respective responses for mean and maximum absorbed doses and prostate-specific membrane antigen (PSMA) volumetric intensity product (VIP-PSMA) at cycles 1 and 2 were termed SPECTmean, SPECTmaximum, and SPECTVIP-PSMA, respectively. Results: Of the 50 patients reviewed, 46% achieved a PSA50 response after cycle 2. Of the 335 index lesions, 58% were osseous, 32% were lymph nodes, and 10% were soft-tissue metastatic lesions. The SPECT lesion-based responses were higher in PSA responders than in nonresponders (SPECTmean response of 46.8% ± 26.1% vs. 26.2% ± 24.5%, P = 0.007; SPECTmaximum response of 45% ± 25.1% vs. 19% ± 27.0%, P = 0.001; SPECTVIP-PSMA response of 49.2% ± 30.3% vs. 14% ± 34.7%, P = 0.0005). An association was observed between PSA response and SPECTVIP-PSMA response (R 2 = 0.40 and P < 0.0001). A limited relationship was found between baseline absorbed dose measured with a 24-h single time point and SPECT lesion-based response (R 2 = 0.05, P = 0.001, and R 2 = 0.03, P = 0.007, for mean and maximum absorbed doses, respectively). Conclusion: In this retrospective study, quantitative lesion-based response correlated with patient-level PSA response. We observed a limited relationship between baseline absorbed dose and lesion-based responses. Most of the variance in response remains unexplained solely by baseline absorbed dose. Establishment of a dose-response relationship in RPT with a single time point at 24 h presented some limitations.
Collapse
Affiliation(s)
- Surekha Yadav
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Fei Jiang
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California
| | - Sara Kurkowska
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, British Columbia, Canada
- Department of Nuclear Medicine, Pomeranian Medical University, Szczecin, Poland
| | - Rachelle Saelee
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Amanda Morley
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Felix Feng
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California
| | - Rahul Aggarwal
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, San Francisco, California
| | - Courtney Lawhn-Heath
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Carlos Uribe
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, British Columbia, Canada
- Department of Radiology, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Molecular Imaging and Therapy, BC Cancer, Vancouver, British Columbia, Canada
| | - Thomas A Hope
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California;
- Department of Radiology, San Francisco VA Medical Center, San Francisco, California; and
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| |
Collapse
|
13
|
Kuznetsov M, Adhikarla V, Caserta E, Wang X, Shively JE, Pichiorri F, Rockne RC. Mathematical Modeling Unveils Optimization Strategies for Targeted Radionuclide Therapy of Blood Cancers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.22.595377. [PMID: 38826403 PMCID: PMC11142146 DOI: 10.1101/2024.05.22.595377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Targeted radionuclide therapy is based on injections of cancer-specific molecules conjugated with radioactive nuclides. Despite the specificity of this treatment, it is not devoid of side-effects limiting its use and is especially harmful for rapidly proliferating organs well perfused by blood, like bone marrow. Optimization of radioconjugates administration accounting for toxicity constraints can increase treatment efficacy. Based on our experiments on disseminated multiple myeloma mouse model treated by 225Ac-DOTA-daratumumab, we developed a mathematical model which investigation highlighted the following principles for optimization of targeted radionuclide therapy. 1) Nuclide to antibody ratio importance. The density of radioconjugates on cancer cells determines the density of radiation energy deposited in them. Low labeling ratio as well as accumulation of unlabeled antibodies and antibodies attached to decay products in the bloodstream can mitigate cancer radiation damage due to excessive occupation of specific receptors by antibodies devoid of radioactive nuclides. 2) Cancer binding capacity-based dosing. The rate of binding of drug to cancer cells depends on the total number of their specific receptors, which therefore can be estimated from the pharmacokinetic curve of diagnostic radioconjugates. Injection of doses significantly exceeding cancer binding capacity should be avoided since radioconjugates remaining in the bloodstream have negligible efficacy to toxicity ratio. 3) Particle range-guided multi-dosing. The use of short-range particle emitters and high-affinity antibodies allows for robust treatment optimization via initial saturation of cancer binding capacity, enabling redistribution of further injected radioconjugates and deposited dose towards still viable cells that continue expressing specific receptors.
Collapse
Affiliation(s)
- Maxim Kuznetsov
- Department of Computational and Quantitative Medicine, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Vikram Adhikarla
- Department of Computational and Quantitative Medicine, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Enrico Caserta
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Xiuli Wang
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, California, United States
| | - John E Shively
- Department of Molecular Imaging & Therapy, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States
| | - Flavia Pichiorri
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Russell C Rockne
- Department of Computational and Quantitative Medicine, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| |
Collapse
|
14
|
Kristiansson A, Vilhelmsson Timmermand O, Altai M, Strand SE, Åkerström B, Örbom A. Hematological and renal toxicity in mice after three cycles of high activity [ 177Lu]Lu-PSMA-617 with or without human α 1-microglobulin. Sci Rep 2024; 14:10787. [PMID: 38734765 PMCID: PMC11088679 DOI: 10.1038/s41598-024-61370-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 05/06/2024] [Indexed: 05/13/2024] Open
Abstract
Radioligand therapy with [177Lu]Lu-PSMA-617 can be used to prolong life and reduce tumor burden in terminally ill castration resistant prostate cancer patients. Still, accumulation in healthy tissue limits the activity that can be administered. Therefore, fractionated therapy is used to lower toxicity. However, there might be a need to reduce toxicity even further with e.g. radioprotectors. The aim of this study was to (i). establish a preclinical mouse model with fractionated high activity therapy of three consecutive doses of 200 MBq [177Lu]Lu-PSMA-617 in which we aimed to (ii). achieve measurable hematotoxicity and nephrotoxicity and to (iii). analyze the potential protective effect of co-injecting recombinant α1-microglobulin (rA1M), a human antioxidant previously shown to have radioprotective effects. In both groups, three cycles resulted in increased albuminuria for each cycle, with large individual variation. Another marker of kidney injury, serum blood urea nitrogen (BUN), was only significantly increased compared to control animals after the third cycle. The number of white and red blood cells decreased significantly and did not reach the levels of control animals during the experiment. rA1M did reduce absorbed dose to kidney but did not show significant protection here, but future studies are warranted due to the recent clinical studies showing a significant renoprotective effect in patients.
Collapse
Affiliation(s)
- Amanda Kristiansson
- Department of Clinical Sciences Lund, Section for Oncology, Lund University, Barngatan 4, 222 42, Lund, Sweden
- Department of Clinical Sciences Lund, Section for Pediatrics, Lund University, Lund, Sweden
- Department of Neonatology, Skåne University Hospital, Lund, Sweden
| | - Oskar Vilhelmsson Timmermand
- Department of Clinical Sciences Lund, Section for Oncology, Lund University, Barngatan 4, 222 42, Lund, Sweden
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Mohamed Altai
- Department of Clinical Sciences Lund, Section for Oncology, Lund University, Barngatan 4, 222 42, Lund, Sweden
| | - Sven-Erik Strand
- Department of Clinical Sciences Lund, Section for Oncology, Lund University, Barngatan 4, 222 42, Lund, Sweden
- Department of Clinical Sciences Lund, Section for Medical Radiation Physics, Lund University, Lund, Sweden
| | - Bo Åkerström
- Department of Clinical Sciences Lund, Section for Infection Medicine, Lund University, Lund, Sweden
| | - Anders Örbom
- Department of Clinical Sciences Lund, Section for Oncology, Lund University, Barngatan 4, 222 42, Lund, Sweden.
| |
Collapse
|
15
|
Reilly RM, Georgiou CJ, Brown MK, Cai Z. Radiation nanomedicines for cancer treatment: a scientific journey and view of the landscape. EJNMMI Radiopharm Chem 2024; 9:37. [PMID: 38703297 PMCID: PMC11069497 DOI: 10.1186/s41181-024-00266-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 04/22/2024] [Indexed: 05/06/2024] Open
Abstract
BACKGROUND Radiation nanomedicines are nanoparticles labeled with radionuclides that emit α- or β-particles or Auger electrons for cancer treatment. We describe here our 15 years scientific journey studying locally-administered radiation nanomedicines for cancer treatment. We further present a view of the radiation nanomedicine landscape by reviewing research reported by other groups. MAIN BODY Gold nanoparticles were studied initially for radiosensitization of breast cancer to X-radiation therapy. These nanoparticles were labeled with 111In to assess their biodistribution after intratumoural vs. intravenous injection. Intravenous injection was limited by high liver and spleen uptake and low tumour uptake, while intratumoural injection provided high tumour uptake but low normal tissue uptake. Further, [111In]In-labeled gold nanoparticles modified with trastuzumab and injected iintratumourally exhibited strong tumour growth inhibition in mice with subcutaneous HER2-positive human breast cancer xenografts. In subsequent studies, strong tumour growth inhibition in mice was achieved without normal tissue toxicity in mice with human breast cancer xenografts injected intratumourally with gold nanoparticles labeled with β-particle emitting 177Lu and modified with panitumumab or trastuzumab to specifically bind EGFR or HER2, respectively. A nanoparticle depot (nanodepot) was designed to incorporate and deliver radiolabeled gold nanoparticles to tumours using brachytherapy needle insertion techniques. Treatment of mice with s.c. 4T1 murine mammary carcinoma tumours with a nanodepot incorporating [90Y]Y-labeled gold nanoparticles inserted into one tumour arrested tumour growth and caused an abscopal growth-inhibitory effect on a distant second tumour. Convection-enhanced delivery of [177Lu]Lu-AuNPs to orthotopic human glioblastoma multiforme (GBM) tumours in mice arrested tumour growth without normal tissue toxicity. Other groups have explored radiation nanomedicines for cancer treatment in preclinical animal tumour xenograft models using gold nanoparticles, liposomes, block copolymer micelles, dendrimers, carbon nanotubes, cellulose nanocrystals or iron oxide nanoparticles. These nanoparticles were labeled with radionuclides emitting Auger electrons (111In, 99mTc, 125I, 103Pd, 193mPt, 195mPt), β-particles (177Lu, 186Re, 188Re, 90Y, 198Au, 131I) or α-particles (225Ac, 213Bi, 212Pb, 211At, 223Ra). These studies employed intravenous or intratumoural injection or convection enhanced delivery. Local administration of these radiation nanomedicines was most effective and minimized normal tissue toxicity. CONCLUSIONS Radiation nanomedicines have shown great promise for treating cancer in preclinical studies. Local intratumoural administration avoids sequestration by the liver and spleen and is most effective for treating tumours, while minimizing normal tissue toxicity.
Collapse
Affiliation(s)
- Raymond M Reilly
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada.
- Princess Margaret Cancer Centre, Toronto, ON, Canada.
- Department of Medical Imaging, University of Toronto, Toronto, ON, Canada.
- Joint Department of Medical Imaging, University Health Network, Toronto, ON, Canada.
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, M5S 3M2, Canada.
| | | | - Madeline K Brown
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada
| | - Zhongli Cai
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
16
|
Kiess AP, O'Donoghue J, Uribe C, Bodei L, Hobbs RF, Hesterman J, Kesner AL, Sgouros G. How Can Radiopharmaceutical Therapies Reach Their Full Potential? Improving Dose Reporting and Phase I Clinical Trial Design. J Clin Oncol 2024:JCO2301241. [PMID: 38484205 DOI: 10.1200/jco.23.01241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 11/02/2023] [Accepted: 12/12/2023] [Indexed: 03/22/2024] Open
Affiliation(s)
- Ana P Kiess
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Joseph O'Donoghue
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Carlos Uribe
- Functional Imaging, BC Cancer, Vancouver, BC, Canada
- Department of Radiology, University of British Columbia, Vancouver, BC, Canada
| | - Lisa Bodei
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Robert F Hobbs
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD
| | | | - Adam L Kesner
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - George Sgouros
- Department of Radiology, Johns Hopkins Medical Institutes, Baltimore, MD
| |
Collapse
|
17
|
Sarrett S, Rodriguez C, Delaney S, Hosny MM, Sebastiano J, Santos-Coquillat A, Keinänen OM, Carter LM, Lastwika KJ, Lampe PD, Zeglis BM. Evaluating CD133 as a Radiotheranostic Target in Small-Cell Lung Cancer. Mol Pharm 2024; 21:1402-1413. [PMID: 38331430 PMCID: PMC10915790 DOI: 10.1021/acs.molpharmaceut.3c01063] [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: 11/08/2023] [Revised: 01/19/2024] [Accepted: 01/19/2024] [Indexed: 02/10/2024]
Abstract
Despite decades of work, small-cell lung cancer (SCLC) remains a frustratingly recalcitrant disease. Both diagnosis and treatment are challenges: low-dose computed tomography (the approved method used for lung cancer screening) is unable to reliably detect early SCLC, and the malignancy's 5 year survival rate stands at a paltry 7%. Clearly, the development of novel diagnostic and therapeutic tools for SCLC is an urgent, unmet need. CD133 is a transmembrane protein that is expressed at low levels in normal tissue but is overexpressed by a variety of tumors, including SCLC. We previously explored CD133 as a biomarker for a novel autoantibody-to-immunopositron emission tomography (PET) strategy for the diagnosis of SCLC, work that first suggested the promise of the antigen as a radiotheranostic target in the disease. Herein, we report the in vivo validation of a pair of CD133-targeted radioimmunoconjugates for the PET imaging and radioimmunotherapy of SCLC. To this end, [89Zr]Zr-DFO-αCD133 was first interrogated in a trio of advanced murine models of SCLC─i.e., orthotopic, metastatic, and patient-derived xenografts─with the PET probe consistently producing high activity concentrations (>%ID/g) in tumor lesions combined with low uptake in healthy tissues. Subsequently, a variant of αCD133 labeled with the β-emitting radiometal 177Lu─[177Lu]Lu-DTPA-A″-CHX-αCD133─was synthesized and evaluated in a longitudinal therapy study in a subcutaneous xenograft model of SCLC, ultimately revealing that treatment with a dose of 9.6 MBq of the radioimmunoconjugate produced a significant increase in median survival compared to a control cohort. Taken together, these data establish CD133 as a viable target for the nuclear imaging and radiopharmaceutical therapy of SCLC.
Collapse
Affiliation(s)
- Samantha
M. Sarrett
- Department
of Chemistry, Hunter College, City University
of New York, New York, New York 10065, United States
- Ph.D.
Program in Biochemistry, The Graduate Center
of the City University of New York, New York, New York 10016, United States
- Department
of Radiology, Memorial Sloan Kettering Cancer
Center, New York, New York 10065, United States
| | - Cindy Rodriguez
- Department
of Chemistry, Hunter College, City University
of New York, New York, New York 10065, United States
- Department
of Radiology, Memorial Sloan Kettering Cancer
Center, New York, New York 10065, United States
- Ph.D.
Program in Chemistry, The Graduate Center
of the City University of New York, New York, New York 10016, United States
| | - Samantha Delaney
- Department
of Chemistry, Hunter College, City University
of New York, New York, New York 10065, United States
- Ph.D.
Program in Biochemistry, The Graduate Center
of the City University of New York, New York, New York 10016, United States
- Department
of Radiology, Memorial Sloan Kettering Cancer
Center, New York, New York 10065, United States
| | - Meena M. Hosny
- Department
of Chemistry, Hunter College, City University
of New York, New York, New York 10065, United States
| | - Joni Sebastiano
- Department
of Chemistry, Hunter College, City University
of New York, New York, New York 10065, United States
- Ph.D.
Program in Biochemistry, The Graduate Center
of the City University of New York, New York, New York 10016, United States
- Department
of Radiology, Memorial Sloan Kettering Cancer
Center, New York, New York 10065, United States
| | - Ana Santos-Coquillat
- Department
of Chemistry, CICECO—Aveiro Institute of Materials, University of Aveiro, Campus Universitario de Santiago, Aveiro 3810-193, Portugal
| | - Outi M. Keinänen
- Department
of Chemistry, Hunter College, City University
of New York, New York, New York 10065, United States
- Department
of Radiology, Memorial Sloan Kettering Cancer
Center, New York, New York 10065, United States
- Department
of Chemistry, University of Helsinki, Helsinki 00100, Finland
| | - Lukas M. Carter
- Department
of Radiology, Memorial Sloan Kettering Cancer
Center, New York, New York 10065, United States
| | - Kristin J. Lastwika
- Translational
Research Program, Public Health Sciences
Division, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, United States
- Translational
Science and Therapeutics Division, Fred
Hutchinson Cancer Research Center, Seattle, Washington 98109, United States
| | - Paul D. Lampe
- Translational
Research Program, Public Health Sciences
Division, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, United States
- Human
Biology Division, Fred Hutchinson Cancer
Research Center, Seattle, Washington 98109, United States
| | - Brian M. Zeglis
- Department
of Chemistry, Hunter College, City University
of New York, New York, New York 10065, United States
- Ph.D.
Program in Biochemistry, The Graduate Center
of the City University of New York, New York, New York 10016, United States
- Department
of Radiology, Memorial Sloan Kettering Cancer
Center, New York, New York 10065, United States
- Ph.D.
Program in Chemistry, The Graduate Center
of the City University of New York, New York, New York 10016, United States
| |
Collapse
|
18
|
Strosberg J, Hofman MS, Al-Toubah T, Hope TA. Rethinking Dosimetry: The Perils of Extrapolated External-Beam Radiotherapy Constraints to Radionuclide Therapy. J Nucl Med 2024; 65:362-364. [PMID: 38212065 DOI: 10.2967/jnumed.123.267167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 12/08/2023] [Accepted: 12/08/2023] [Indexed: 01/13/2024] Open
Affiliation(s)
- Jonathan Strosberg
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida;
| | - Michael S Hofman
- Prostate Cancer Theranostics and Imaging Centre of Excellence, Molecular Imaging and Therapeutic Nuclear Medicine, Cancer Imaging, Peter MacCallum Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia; and
| | - Taymeyah Al-Toubah
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Thomas A Hope
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| |
Collapse
|
19
|
George SC, Tolakanahalli R, Aguirre S, Kim TP, Samuel EJJ, Mishra V. A single-institution experience with 177Lu RPT workflow improvements and qualifying the SPECT/CT imaging for dosimetry. Front Oncol 2024; 14:1331266. [PMID: 38469241 PMCID: PMC10925616 DOI: 10.3389/fonc.2024.1331266] [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: 10/31/2023] [Accepted: 01/22/2024] [Indexed: 03/13/2024] Open
Abstract
Background and purpose Implementing any radiopharmaceutical therapy (RPT) program requires a comprehensive review of system readiness, appropriate workflows, and training to ensure safe and efficient treatment delivery. A quantitative assessment of the dose delivered to targets and organs at risk (OAR) using RPT is possible by correlating the absorbed doses with the delivered radioactivity. Integrating dosimetry into an established RPT program demands a thorough analysis of the necessary components and system fine-tuning. This study aims to report an optimized workflow for molecular radiation therapy using 177Lu with a primary focus on integrating patient-specific dosimetry into an established radiopharmaceutical program in a radiation oncology setting. Materials and methods We comprehensively reviewed using the Plan-Do-Check-Act (PDCA) cycle, including efficacy and accuracy of delivery and all aspects of radiation safety of the RPT program. The GE Discovery SPECT/CT 670DR™ system was calibrated per MIM protocol for dose calculation on MIM SurePlan™ MRT software. Jaszcak Phantom with 15-20 mCi of 177Lu DOTATATE with 2.5 µM EDTA solution was used, with the main energy window defined as 208 keV ±10% (187.6 to 229.2 keV); the upper scatter energy window was set to 240 keV ±5% (228 to 252 keV), while the lower scatter energy window was 177.8 keV ±5% (168.9 to 186.7 keV). Volumetric quality control tests and adjustments were performed to ensure the correct alignment of the table, NM, and CT gantry on SPECT/CT. A comprehensive end-to-end (E2E) test was performed to ensure workflow, functionality, and quantitative dose accuracy. Results Workflow improvements and checklists are presented after systematically analyzing over 400 administrations of 177Lu-based RPT. Injected activity to each sphere in the NEMA Phantom scan was quantified, and the MIM Sureplan MRT reconstruction images calculated activities within ±12% of the injected activity. Image alignment tests on the SPECT/CT showed a discrepancy of more than the maximum tolerance of 2.2 mm on any individual axis. As a result of servicing the machine and updating the VQC and COR corrections, the hybrid imaging system was adjusted to achieve an accuracy of <1 mm in all directions. Conclusion Workflows and checklists, after analysis of system readiness and adequate training for staff and patients, are presented. Hardware and software components for patient-specific dosimetry are presented with a focus on hybrid image registration and correcting any errors that affect dosimetric quantification calculation. Moreover, this manuscript briefly overviews the necessary quality assurance requirements for converting diagnostic images into dosimetry measurement tools and integrating dosimetry for RPT based on 177Lu.
Collapse
Affiliation(s)
- Siju C. George
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health, Miami, FL, United States
- Department of Physics, School of Advanced Sciences, Vellore Institute of Technology, Vellore, India
| | - Ranjini Tolakanahalli
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health, Miami, FL, United States
| | - Santiago Aguirre
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health, Miami, FL, United States
| | - Taehyung Peter Kim
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health, Miami, FL, United States
| | | | - Vivek Mishra
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health, Miami, FL, United States
| |
Collapse
|
20
|
Hofman MS, Tran B, Feldman DR, Pokorska-Bocci A, Pichereau S, Wessen J, Haskali MB, Sparks RB, Vlasyuk O, Galetic I. First-in-Human Safety, Imaging, and Dosimetry of a Carbonic Anhydrase IX-Targeting Peptide, [ 68Ga]Ga-DPI-4452, in Patients with Clear Cell Renal Cell Carcinoma. J Nucl Med 2024; 65:jnumed.123.267175. [PMID: 38388517 PMCID: PMC11064824 DOI: 10.2967/jnumed.123.267175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 01/23/2024] [Accepted: 01/23/2024] [Indexed: 02/24/2024] Open
Abstract
[68Ga]Ga-DPI-4452, a first-in-class carbonic anhydrase IX-binding radiolabeled peptide, is the imaging agent of a theranostic pair with [177Lu]Lu-DPI-4452, developed for selecting and treating patients with carbonic anhydrase IX-expressing tumors. Here, [68Ga]Ga-DPI-4452 imaging characteristics, dosimetry, pharmacokinetics, and safety were assessed in 3 patients with clear cell renal cell carcinoma. Methods: After [68Ga]Ga-DPI-4452 administration, patients underwent serial full-body PET/CT imaging. Blood and urine were sampled. Safety was monitored for 7 d after injection. Results: Tumor uptake was observed at all time points (15 min to 4 h). Across 36 lesions, the SUVmax at 1 h after administration ranged from 6.8 to 211.6 (mean, 64.6 [SD, 54.8]). The kidneys, liver, and bone marrow demonstrated low activity. [68Ga]Ga-DPI-4452 was rapidly eliminated from blood and urine. No clinically significant toxicity was observed. Conclusion: [68Ga]Ga-DPI-4452 showed exceptional tumor uptake in patients with clear cell renal cell carcinoma, with very high tumor-to-background ratios and no significant adverse events, suggesting potential diagnostic and patient selection applications.
Collapse
Affiliation(s)
- Michael S Hofman
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia;
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Ben Tran
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Darren R Feldman
- Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, New York
| | | | | | | | - Mohammad B Haskali
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | | | - Olena Vlasyuk
- Debiopharm International SA, Lausanne, Switzerland; and
| | - Ivana Galetic
- Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, New York
| |
Collapse
|
21
|
de Roode KE, Joosten L, Behe M. Towards the Magic Radioactive Bullet: Improving Targeted Radionuclide Therapy by Reducing the Renal Retention of Radioligands. Pharmaceuticals (Basel) 2024; 17:256. [PMID: 38399470 PMCID: PMC10892921 DOI: 10.3390/ph17020256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 02/07/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
Targeted radionuclide therapy (TRT) is an emerging field and has the potential to become a major pillar in effective cancer treatment. Several pharmaceuticals are already in routine use for treating cancer, and there is still a high potential for new compounds for this application. But, a major issue for many radiolabeled low-to-moderate-molecular-weight molecules is their clearance via the kidneys and their subsequent reuptake. High renal accumulation of radioactive compounds may lead to nephrotoxicity, and therefore, the kidneys are often the dose-limiting organs in TRT with these radioligands. Over the years, different strategies have been developed aiming for reduced kidney retention and enhanced therapeutic efficacy of radioligands. In this review, we will give an overview of the efforts and achievements of the used strategies, with focus on the therapeutic potential of low-to-moderate-molecular-weight molecules. Among the strategies discussed here is coadministration of compounds that compete for binding to the endocytic receptors in the proximal tubuli. In addition, the influence of altering the molecular design of radiolabeled ligands on pharmacokinetics is discussed, which includes changes in their physicochemical properties and implementation of cleavable linkers or albumin-binding moieties. Furthermore, we discuss the influence of chelator and radionuclide choice on reabsorption of radioligands by the kidneys.
Collapse
Affiliation(s)
- Kim E. de Roode
- Department of Medical Imaging, Nuclear Medicine, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands;
- Tagworks Pharmaceuticals, Toernooiveld 1, 6525 ED Nijmegen, The Netherlands
| | - Lieke Joosten
- Department of Medical Imaging, Nuclear Medicine, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands;
| | - Martin Behe
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institut, 5232 Villigen, Switzerland
| |
Collapse
|
22
|
Carter LM, Zanzonico PB. MIB Guides: Preclinical Radiopharmaceutical Dosimetry. Mol Imaging Biol 2024; 26:17-28. [PMID: 37964036 DOI: 10.1007/s11307-023-01868-9] [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: 08/03/2023] [Revised: 09/26/2023] [Accepted: 10/20/2023] [Indexed: 11/16/2023]
Abstract
Preclinical dosimetry is essential for guiding the design of animal radiopharmaceutical biodistribution, imaging, and therapy experiments, evaluating efficacy and/or toxicities in such experiments, ensuring compliance with ethical standards for animal research, and, perhaps most importantly, providing reasonable initial estimates of normal-organ doses in humans, required for clinical translation of new radiopharmaceuticals. This MIB Guide provides a basic protocol for obtaining preclinical dosimetry estimates with organ-level dosimetry software.
Collapse
Affiliation(s)
- Lukas M Carter
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Pat B Zanzonico
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
23
|
Ha S, O JH, Park C, Boo SH, Yoo IR, Moon HW, Chi DY, Lee JY. Dosimetric Analysis of a Phase I Study of PSMA-Targeting Radiopharmaceutical Therapy With [ 177Lu]Ludotadipep in Patients With Metastatic Castration-Resistant Prostate Cancer. Korean J Radiol 2024; 25:179-188. [PMID: 38288897 PMCID: PMC10831299 DOI: 10.3348/kjr.2023.0656] [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: 07/14/2023] [Revised: 10/30/2023] [Accepted: 11/17/2023] [Indexed: 02/01/2024] Open
Abstract
OBJECTIVE 177Lutetium [Lu] Ludotadipep is a novel prostate-specific membrane antigen targeting therapeutic agent with an albumin motif added to increase uptake in the tumors. We assessed the biodistribution and dosimetry of [177Lu]Ludotadipep in patients with metastatic castration-resistant prostate cancer (mCRPC). MATERIALS AND METHODS Data from 25 patients (median age, 73 years; range, 60-90) with mCRPC from a phase I study with activity escalation design of single administration of [177Lu]Ludotadipep (1.85, 2.78, 3.70, 4.63, and 5.55 GBq) were assessed. Activity in the salivary glands, lungs, liver, kidneys, and spleen was estimated from whole-body scan and abdominal SPECT/CT images acquired at 2, 24, 48, 72, and 168 h after administration of [177Lu]Ludotadipep. Red marrow activity was calculated from blood samples obtained at 3, 10, 30, 60, and 180 min, and at 24, 48, and 72 h after administration. Organ- and tumor-based absorbed dose calculations were performed using IDAC-Dose 2.1. RESULTS Absorbed dose coefficient (mean ± standard deviation) of normal organs was 1.17 ± 0.81 Gy/GBq for salivary glands, 0.05 ± 0.02 Gy/GBq for lungs, 0.14 ± 0.06 Gy/GBq for liver, 0.77 ± 0.28 Gy/GBq for kidneys, 0.12 ± 0.06 Gy/GBq for spleen, and 0.07 ± 0.02 Gy/GBq for red marrow. The absorbed dose coefficient of the tumors was 10.43 ± 7.77 Gy/GBq. CONCLUSION [177Lu]Ludotadipep is expected to be safe at the dose of 3.7 GBq times 6 cycles planned for a phase II clinical trial with kidneys and bone marrow being the critical organs, and shows a high tumor absorbed dose.
Collapse
Affiliation(s)
- Seunggyun Ha
- Division of Nuclear Medicine, Department of Radiology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Joo Hyun O
- Division of Nuclear Medicine, Department of Radiology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| | - Chansoo Park
- Research Institute of Labeling, FutureChem Co., Ltd., Seoul, Republic of Korea
| | - Sun Ha Boo
- Division of Nuclear Medicine, Department of Radiology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Ie Ryung Yoo
- Division of Nuclear Medicine, Department of Radiology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hyong Woo Moon
- Department of Urology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Dae Yoon Chi
- Research Institute of Labeling, FutureChem Co., Ltd., Seoul, Republic of Korea
| | - Ji Youl Lee
- Department of Urology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| |
Collapse
|
24
|
Kayal G, Barbosa N, Marín CC, Ferrer L, Fragoso-Negrín JA, Grosev D, Gupta SK, Hidayati NR, Moalosi TCG, Poli GL, Thakral P, Tsapaki V, Vauclin S, Vergara-Gil A, Knoll P, Hobbs RF, Bardiès M. Quality Assurance Considerations in Radiopharmaceutical Therapy Dosimetry Using PLANETDose: An International Atomic Energy Agency Study. J Nucl Med 2024; 65:125-131. [PMID: 37884334 PMCID: PMC10755524 DOI: 10.2967/jnumed.122.265340] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 07/25/2023] [Indexed: 10/28/2023] Open
Abstract
Implementation of radiopharmaceutical therapy dosimetry varies depending on the clinical application, dosimetry protocol, software, and ultimately the operator. Assessing clinical dosimetry accuracy and precision is therefore a challenging task. This work emphasizes some pitfalls encountered during a structured analysis, performed on a single-patient dataset consisting of SPECT/CT images by various participants using a standard protocol and clinically approved commercial software. Methods: The clinical dataset consisted of the dosimetric study of a patient administered with [177Lu]Lu-DOTATATE at Tygerberg Hospital, South Africa, as a part of International Atomic Energy Agency-coordinated research project E23005. SPECT/CT images were acquired at 5 time points postinjection. Patient and calibration images were reconstructed on a workstation, and a calibration factor of 122.6 Bq/count was derived independently and provided to the participants. A standard dosimetric protocol was defined, and PLANETDose (version 3.1.1) software was installed at 9 centers to perform the dosimetry of 3 treatment cycles. The protocol included rigid image registration, segmentation (semimanual for organs, activity threshold for tumors), and dose voxel kernel convolution of activity followed by absorbed dose (AD) rate integration to obtain the ADs. Iterations of the protocol were performed by participants individually and within collective training, the results of which were analyzed for dosimetric variability, as well as for quality assurance and error analysis. Intermediary checkpoints were developed to understand possible sources of variation and to differentiate user error from legitimate user variability. Results: Initial dosimetric results for organs (liver and kidneys) and lesions showed considerable interoperator variability. Not only was the generation of intermediate checkpoints such as total counts, volumes, and activity required, but also activity-to-count ratio, activity concentration, and AD rate-to-activity concentration ratio to determine the source of variability. Conclusion: When the same patient dataset was analyzed using the same dosimetry procedure and software, significant disparities were observed in the results despite multiple sessions of training and feedback. Variations due to human error could be minimized or avoided by performing intensive training sessions, establishing intermediate checkpoints, conducting sanity checks, and cross-validating results across physicists or with standardized datasets. This finding promotes the development of quality assurance in clinical dosimetry.
Collapse
Affiliation(s)
- Gunjan Kayal
- CRCT, UMR 1037, INSERM, Université Toulouse III Paul Sabatier, Toulouse, France
- SCK CEN, Belgian Nuclear Research Centre, Mol, Belgium
| | | | | | - Ludovic Ferrer
- Medical Physics Department, ICO René Gauducheau, Nantes, France
- CRCINA, UMR 1232, INSERM, France
| | - José-Alejandro Fragoso-Negrín
- DOSIsoft SA, Cachan, France
- IRCM, UMR 1194 INSERM, Universite de Montpellier and Institut Regional du Cancer de Montpellier, Montpellier, France
| | - Darko Grosev
- Department of Nuclear Medicine and Radiation Protection, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Santosh Kumar Gupta
- Department of Nuclear Medicine and PET, Mahamana Pandit Madanmohan Malviya Cancer Centre and Homi Bhabha Cancer Centre, Varanasi, India
| | - Nur Rahmah Hidayati
- Research Center and Technology for Radiation Safety and Metrology-National Research and Innovation Agency, Jakarta, Indonesia
| | - Tumelo C G Moalosi
- Department of Medical Imaging and Clinical Oncology, Medical Physics, Nuclear Medicine Division, Faculty of Medicine and Health Science, Stellenbosch University, Tygerberg Hospital, Cape Town, South Africa
| | - Gian Luca Poli
- Department of Medical Physics, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Parul Thakral
- Department of Nuclear Medicine, Fortis Memorial Research Institute, Gurugram, India
| | - Virginia Tsapaki
- Dosimetry and Medical Radiation Physics, International Atomic Energy Agency, Vienna, Austria
| | | | - Alex Vergara-Gil
- CRCT, UMR 1037, INSERM, Université Toulouse III Paul Sabatier, Toulouse, France
| | - Peter Knoll
- Dosimetry and Medical Radiation Physics, International Atomic Energy Agency, Vienna, Austria
| | - Robert F Hobbs
- Johns Hopkins Medical Institute, Baltimore, Maryland; and
| | - Manuel Bardiès
- IRCM, UMR 1194 INSERM, Universite de Montpellier and Institut Regional du Cancer de Montpellier, Montpellier, France;
- Département de Médecine Nucléaire, Institut Régional du Cancer de Montpellier, Montpellier, France
| |
Collapse
|
25
|
Sedlack AJH, Meyer C, Mench A, Winters C, Barbon D, Obrzut S, Mallak N. Essentials of Theranostics: A Guide for Physicians and Medical Physicists. Radiographics 2024; 44:e230097. [PMID: 38060426 DOI: 10.1148/rg.230097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
Radiopharmaceutical therapies (RPTs) are gaining increased interest with the recent emergence of novel safe and effective theranostic agents, improving outcomes for thousands of patients. The term theranostics refers to the use of diagnostic and therapeutic agents that share the same molecular target; a major step toward precision medicine, especially for oncologic applications. The authors dissect the fundamentals of theranostics in nuclear medicine. First, they explain the radioactive decay schemes and the characteristics of emitted electromagnetic radiation used for imaging, as well as particles used for therapeutic purposes, followed by the interaction of the different types of radiation with tissue. These concepts directly apply to clinical RPTs and play a major role in the efficacy and toxicity profile of different radiopharmaceutical agents. Personalized dosimetry is a powerful tool that can help estimate patient-specific absorbed doses, in tumors as well as normal organs. Dosimetry in RPT is an area of active investigation, as most of what we know about the relationship between delivered dose and tissue damage is extrapolated from external-beam radiation therapy; more research is needed to understand this relationship as it pertains to RPTs. Tumor heterogeneity is increasingly recognized as an important prognostic factor. Novel molecular imaging agents, often in combination with fluorine 18-fluorodeoxyglucose, are crucial for assessment of target expression in the tumor and potential hypermetabolic disease that may lack the molecular target expression. ©RSNA, 2023 Test Your Knowledge questions are available in the supplemental material.
Collapse
Affiliation(s)
- Andrew J H Sedlack
- From the Medical Scientist Training Program, Feinberg School of Medicine, Northwestern University, Chicago, Ill (A.J.H.S.); and Department of Diagnostic Radiology, Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, L340, Portland, OR 97239-3098 (C.M., A.M., C.W., D.B., S.O., N.M.)
| | - Catherine Meyer
- From the Medical Scientist Training Program, Feinberg School of Medicine, Northwestern University, Chicago, Ill (A.J.H.S.); and Department of Diagnostic Radiology, Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, L340, Portland, OR 97239-3098 (C.M., A.M., C.W., D.B., S.O., N.M.)
| | - Anna Mench
- From the Medical Scientist Training Program, Feinberg School of Medicine, Northwestern University, Chicago, Ill (A.J.H.S.); and Department of Diagnostic Radiology, Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, L340, Portland, OR 97239-3098 (C.M., A.M., C.W., D.B., S.O., N.M.)
| | - Celeste Winters
- From the Medical Scientist Training Program, Feinberg School of Medicine, Northwestern University, Chicago, Ill (A.J.H.S.); and Department of Diagnostic Radiology, Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, L340, Portland, OR 97239-3098 (C.M., A.M., C.W., D.B., S.O., N.M.)
| | - Dennis Barbon
- From the Medical Scientist Training Program, Feinberg School of Medicine, Northwestern University, Chicago, Ill (A.J.H.S.); and Department of Diagnostic Radiology, Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, L340, Portland, OR 97239-3098 (C.M., A.M., C.W., D.B., S.O., N.M.)
| | - Sebastian Obrzut
- From the Medical Scientist Training Program, Feinberg School of Medicine, Northwestern University, Chicago, Ill (A.J.H.S.); and Department of Diagnostic Radiology, Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, L340, Portland, OR 97239-3098 (C.M., A.M., C.W., D.B., S.O., N.M.)
| | - Nadine Mallak
- From the Medical Scientist Training Program, Feinberg School of Medicine, Northwestern University, Chicago, Ill (A.J.H.S.); and Department of Diagnostic Radiology, Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, L340, Portland, OR 97239-3098 (C.M., A.M., C.W., D.B., S.O., N.M.)
| |
Collapse
|
26
|
Meyer C, Szidonya L, Winters C, Mench A, Mallak N, Mittra E. Quantitative imaging for 177Lu-PSMA treatment response monitoring and dosimetry. FRONTIERS IN NUCLEAR MEDICINE (LAUSANNE, SWITZERLAND) 2023; 3:1291253. [PMID: 39355047 PMCID: PMC11440845 DOI: 10.3389/fnume.2023.1291253] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 11/20/2023] [Indexed: 10/03/2024]
Abstract
PSMA-targeted radiopharmaceutical therapy is an established treatment option for metastatic castration-resistant prostate cancer (mCRPC). However, response rates and duration using 177Lu-PSMA-617 vary considerably between patients. Quantitative 177Lu SPECT imaging is one approach that may be leveraged to more closely monitor inter-cycle response, as well as patient-specific absorbed doses. In this work, we describe our experience implementing quantitative imaging throughout the course of 177Lu-PSMA treatment, including serial SPECT imaging to monitor response and for individualized dosimetry. We also describe our imaging protocols and dose calculation workflows for 3D voxelized patient-specific organ and tumor dosimetry, including a review of the current landscape and efforts towards harmonized dosimetry.
Collapse
Affiliation(s)
| | | | | | | | | | - Erik Mittra
- Department of Diagnostic Radiology, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
27
|
Fu H, Huang J, Zhao T, Wang H, Chen Y, Xu W, Pang Y, Guo W, Sun L, Wu H, Xu P, Su B, Zhang J, Chen X, Chen H. Fibroblast Activation Protein-Targeted Radioligand Therapy with 177Lu-EB-FAPI for Metastatic Radioiodine-Refractory Thyroid Cancer: First-in-Human, Dose-Escalation Study. Clin Cancer Res 2023; 29:4740-4750. [PMID: 37801296 PMCID: PMC10690094 DOI: 10.1158/1078-0432.ccr-23-1983] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 08/20/2023] [Accepted: 10/02/2023] [Indexed: 10/07/2023]
Abstract
PURPOSE Fibroblast activation protein (FAP) is a promising target for tumor treatment. In this study, we aimed to investigate the safety and efficacy of the albumin binder-conjugated FAP-targeted radiopharmaceutical, 177Lu-EB-FAPI (177Lu-LNC1004), in patients with metastatic radioiodine-refractory thyroid cancer (mRAIR-TC). PATIENTS AND METHODS This open-label, non-randomized, first-in-human, dose-escalation, investigator-initiated trial had a 3+3 design and involved a 6-week 177Lu-LNC1004 treatment cycle in patients with mRAIR-TC at 2.22 GBq initially, with subsequent cohorts receiving an incremental 50% dose increase until dose-limiting toxicity (DLT) was observed. RESULTS 177Lu-LNC1004 administration was well tolerated, with no life-threatening adverse events observed. No patients experienced DLT in Group A (2.22 GBq/cycle). One patient experienced grade 4 thrombocytopenia in Group B (3.33 GBq/cycle); hence, another three patients were enrolled, none of whom experienced DLT. Two patients experienced grade 3 and 4 hematotoxicity in Group C (4.99 GBq/cycle). The mean whole-body effective dose was 0.17 ± 0.04 mSv/MBq. Intense 177Lu-LNC1004 uptake and prolonged tumor retention resulted in high mean absorbed tumor doses (8.50 ± 12.36 Gy/GBq). The mean effective half-lives for the whole-body and tumor lesions were 90.20 ± 7.68 and 92.46 ± 9.66 hours, respectively. According to RECIST, partial response, stable disease, and progressive disease were observed in 3 (25%), 7 (58%), and 2 (17%) patients, respectively. The objective response and disease control rates were 25% and 83%, respectively. CONCLUSIONS FAP-targeted radioligand therapy with 177Lu-LNC1004 at 3.33 GBq/cycle was well tolerated in patients with advanced mRAIR-TC, with high radiation dose delivery to the tumor lesions, encouraging therapeutic efficacy, and acceptable side effects.
Collapse
Affiliation(s)
- Hao Fu
- Department of Nuclear Medicine and Minnan PET Center, Xiamen Key Laboratory of Radiopharmaceuticals, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jingxiong Huang
- Department of Nuclear Medicine and Minnan PET Center, Xiamen Key Laboratory of Radiopharmaceuticals, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Tianzhi Zhao
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore
- Department of Chemical and Biomolecular Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore
- Department of Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Hongjian Wang
- School of Clinical Medicine, Fujian Medical University, Fuzhou, China
| | - Yuhang Chen
- School of Clinical Medicine, Fujian Medical University, Fuzhou, China
| | - Weizhi Xu
- Department of Nuclear Medicine and Minnan PET Center, Xiamen Key Laboratory of Radiopharmaceuticals, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Yizhen Pang
- Department of Nuclear Medicine and Minnan PET Center, Xiamen Key Laboratory of Radiopharmaceuticals, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Wei Guo
- Department of Nuclear Medicine and Minnan PET Center, Xiamen Key Laboratory of Radiopharmaceuticals, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Long Sun
- Department of Nuclear Medicine and Minnan PET Center, Xiamen Key Laboratory of Radiopharmaceuticals, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Hua Wu
- Department of Nuclear Medicine and Minnan PET Center, Xiamen Key Laboratory of Radiopharmaceuticals, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Pengfei Xu
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore
- Department of Chemical and Biomolecular Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore
- Department of Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Bishan Su
- Department of Nuclear Medicine and Minnan PET Center, Xiamen Key Laboratory of Radiopharmaceuticals, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jingjing Zhang
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore
- Department of Chemical and Biomolecular Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore
- Department of Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Xiaoyuan Chen
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore
- Department of Chemical and Biomolecular Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore
- Department of Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Haojun Chen
- Department of Nuclear Medicine and Minnan PET Center, Xiamen Key Laboratory of Radiopharmaceuticals, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
28
|
Han G, Hwang E, Lin F, Clift R, Kim D, Guest M, Bischoff E, Moran S, Li G. RYZ101 (Ac-225 DOTATATE) Opportunity beyond Gastroenteropancreatic Neuroendocrine Tumors: Preclinical Efficacy in Small-Cell Lung Cancer. Mol Cancer Ther 2023; 22:1434-1443. [PMID: 37616528 DOI: 10.1158/1535-7163.mct-23-0029] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 04/28/2023] [Accepted: 08/21/2023] [Indexed: 08/26/2023]
Abstract
Overexpression of somatostatin receptors (SSTR), particularly SSTR2, is found in gastroenteropancreatic neuroendocrine tumors (GEP-NET), and subsets of other solid tumors such as small-cell lung cancer (SCLC). SCLC accounts for approximately 13% to 15% of lung cancer and lacks effective therapeutic options. IHC analysis indicates that up to 50% of SCLC tumors are SSTR2-positive, with a substantial subset showing high and homogenous expression. Peptide receptor radionuclide therapy with radiolabeled somatostatin analogue, Lu-177 DOTATATE, has been approved for GEP-NETs. Different strategies aimed at improving outcomes, such as the use of alpha-emitting radioisotopes, are currently being investigated. RYZ101 (Ac-225 DOTATATE) is comprised of the alpha-emitting radioisotope actinium-225, chemical chelator DOTA, and octreotate (TATE), a somatostatin analogue. In the cell-based competitive radioligand binding assay, RAYZ-10001-La (lanthanum surrogate for RYZ101) showed high binding affinity (Ki = 0.057 nmol/L) to human SSTR2 and >600-fold selectivity against other SSTR subtypes. RAYZ-10001-La exhibited efficient internalization to SSTR2-positive cells. In multiple SSTR2-expressing SCLC xenograft models, single-dose intravenous RYZ101 3 μCi (0.111 MBq) or 4 μCi (0.148 MBq) significantly inhibited tumor growth, with deeper responses, including sustained regression, observed in the models with higher SSTR2 levels. The antitumor effect was further enhanced when RYZ101 was combined with carboplatin and etoposide at clinically relevant doses. In summary, RYZ101 is a highly potent, alpha-emitting radiopharmaceutical agent, and preclinical data demonstrate the potential of RYZ101 for the treatment of patients with SSTR-positive cancers.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Gary Li
- RayzeBio, Inc., San Diego, California
| |
Collapse
|
29
|
Barbosa N, Niño F, Vallejo-Ortega MT, Naranjo M, Granados CE. Comparison of Whole-Body Dosimetry Measurements in 131I Therapy Using Ceiling-Mounted Geiger-Müller Detectors and γ-Camera Scans: An Agreement Analysis. Cancer Biother Radiopharm 2023; 38:445-449. [PMID: 37358596 DOI: 10.1089/cbr.2022.0094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2023] Open
Abstract
Background: In 131I therapies internal dosimetry is crucial for determining the mean absorbed dose to organs at risk, particularly the bone marrow, which has a dose constraint of 2 Gy. Traditionally, multicompartmental models have been used for bone marrow dosimetry, necessitating whole-body absorbed-dose assessments. However, noninvasive techniques, such as γ-camera scans or ceiling-mounted Geiger-Müller (GM) counters, can estimate the aforementioned. This study was aimed to evaluate the agreement between whole-body mean absorbed dose using γ-camera scans and ceiling-mounted GM in patients with thyroid carcinoma undergoing 131I therapy. Methods: This study included 31 patients with thyroid cancer who were treated with 131I. The whole-body time-integrated activity (TIA) and mean absorbed dose were estimated using the elimination curves obtained with γ-camera scans and ceiling-mounted GM. In addition, statistical analysis was performed on the data to determine the Coefficient Correlation Coefficient and the Bland-Altman limits of agreement for both parameters, as well as for the elimination curves' effective half-life. Results: The study revealed correlations of 0.562 and 0.586 between whole-body TIA and mean absorbed dose, respectively. The Bland-Altman limits of agreement were found to be below -3.75% and within 12.75% of the bone marrow dose constraint of 2 Gy. The nonparametric evaluation revealed that whole-body TIA and mean absorbed dose medians from GM were lower than those from γ-camera scans (p < 0.001). Effective half-life estimation mean was significantly lower in the GM than in the γ-camera of 13 and 23 h. Conclusions: Although GM calculates the whole-body absorbed dose with margins of error within clinical acceptance, underestimation of the effective half-life makes it an unacceptable substitute method for γ-cameras in clinical practice. Further research should be conducted to evaluate single-point GM measurement substitutions in time-activity curves.
Collapse
Affiliation(s)
- Nathaly Barbosa
- Department of Nuclear Medicine, Instituto Nacional de Cancerologia E.S.E., Bogotá, Colombia
| | - Franklin Niño
- Department of Physics, Universidad Nacional de Colombia, Bogotá, Colombia
| | | | - Mónica Naranjo
- Department of Nuclear Medicine, Instituto Nacional de Cancerologia E.S.E., Bogotá, Colombia
| | | |
Collapse
|
30
|
George SC, Samuel EJJ. Developments in 177Lu-based radiopharmaceutical therapy and dosimetry. Front Chem 2023; 11:1218670. [PMID: 37583569 PMCID: PMC10424930 DOI: 10.3389/fchem.2023.1218670] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 06/27/2023] [Indexed: 08/17/2023] Open
Abstract
177Lu is a radioisotope that has become increasingly popular as a therapeutic agent for treating various conditions, including neuroendocrine tumors and metastatic prostate cancer. 177Lu-tagged radioligands are molecules precisely designed to target and bind to specific receptors or proteins characteristic of targeted cancer. This review paper will present an overview of the available 177Lu-labelled radioligands currently used to treat patients. Based on recurring, active, and completed clinical trials and other available literature, we evaluate current status, interests, and developments in assessing patient-specific dosimetry, which will define the future of this particular treatment modality. In addition, we will discuss the challenges and opportunities of the existing dosimetry standards to measure and calculate the radiation dose delivered to patients, which is essential for ensuring treatments' safety and efficacy. Finally, this article intends to provide an overview of the current state of 177Lu- tagged radioligand therapy and highlight the areas where further research can improve patient treatment outcomes.
Collapse
Affiliation(s)
- Siju C. George
- Radiation Oncology Department, Miami Cancer Institute, Baptist Health, Miami, FL, United States
- Department of Physics, School of Advanced Sciences, Vellore Institute of Technology, Vellore, India
| | | |
Collapse
|
31
|
Hoffman SLV, Mixdorf JC, Kwon O, Johnson TR, Makvandi M, Lee H, Aluicio-Sarduy E, Barnhart TE, Jeffery JJ, Patankar MS, Engle JW, Bednarz BP, Ellison PA. Preclinical studies of a PARP targeted, Meitner-Auger emitting, theranostic radiopharmaceutical for metastatic ovarian cancer. Nucl Med Biol 2023; 122-123:108368. [PMID: 37490805 PMCID: PMC10529069 DOI: 10.1016/j.nucmedbio.2023.108368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 07/11/2023] [Accepted: 07/17/2023] [Indexed: 07/27/2023]
Abstract
Advanced ovarian cancer currently has few therapeutic options. Poly(ADP-ribose) polymerase (PARP) inhibitors bind to nuclear PARP and trap the protein-inhibitor complex to DNA. This work investigates a theranostic PARP inhibitor for targeted radiopharmaceutical therapy of ovarian cancer in vitro and PET imaging of healthy mice in vivo. METHODS [77Br]RD1 was synthesized and assessed for pharmacokinetics and cytotoxicity in human and murine ovarian cancer cell lines. [76Br]RD1 biodistribution and organ uptake in healthy mice were quantified through longitudinal PET/CT imaging and ex vivo radioactivity measurements. Organ-level dosimetry following [76/77Br]RD1 administration was calculated using RAPID, an in-house platform for absorbed dose in mice, and OLINDA for equivalent and effective dose in human. RESULTS The maximum specific binding (Bmax), equilibrium dissociation constant (Kd), and nonspecific binding slope (NS) were calculated for each cell line. These values were used to calculate the cell specific activity uptake for cell viability studies. The half maximal effective concentration (EC50) was measured as 0.17 (95 % CI: 0.13-0.24) nM and 0.46 (0.13-0.24) nM for PARP(+) and PARP(-) expressing cell lines, respectively. The EC50 was 0.27 (0.21-0.36) nM and 0.30 (0.22-0.41) nM for BRCA1(-) and BRCA1(+) expressing cell lines, respectively. When measuring the EC50 as a function of cellular activity uptake and nuclear dose, the EC50 ranges from 0.020 to 0.039 Bq/cell and 3.3-9.2 Gy, respectively. Excretion through the hepatobiliary and renal pathways were observed in mice, with liver uptake of 2.3 ± 0.4 %ID/g after 48 h, contributing to estimated absorbed dose values in mice of 19.3 ± 0.3 mGy/MBq and 290 ± 10 mGy/MBq for [77Br]RD1 and [76Br]RD1, respectively. CONCLUSION [77Br]RD1 cytotoxicity was dependent on PARP expression and independent of BRCA1 status. The in vitro results suggest that [77Br]RD1 cytotoxicity is driven by the targeted Meitner-Auger electron (MAe) radiotherapeutic effect of the agent. Further studies investigating the theranostic potential, organ dose, and tumor uptake of [76/77Br]RD1 are warranted.
Collapse
Affiliation(s)
- S L V Hoffman
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - J C Mixdorf
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - O Kwon
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - T R Johnson
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - M Makvandi
- Department of Radiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - H Lee
- Department of Radiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - E Aluicio-Sarduy
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - T E Barnhart
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - J J Jeffery
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - M S Patankar
- Department of Obstetrics and Gynecology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - J W Engle
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - B P Bednarz
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - P A Ellison
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
| |
Collapse
|
32
|
Gnesin S, Chouin N, Cherel M, Dunn SM, Schaefer N, Faivre-Chauvet A, Prior JO, Delage JA. From bench to bedside: 64Cu/ 177Lu 1C1m-Fc anti TEM-1: mice-to-human dosimetry extrapolations for future theranostic applications. EJNMMI Res 2023; 13:59. [PMID: 37314509 DOI: 10.1186/s13550-023-01010-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 06/06/2023] [Indexed: 06/15/2023] Open
Abstract
The development of diagnostic and therapeutic radiopharmaceuticals is an hot topic in nuclear medicine. Several radiolabeled antibodies are under development necessitating both biokinetic and dosimetry extrapolations for effective human translation. The validation of different animal-to-human dosimetry extrapolation methods still is an open issue. This study reports the mice-to-human dosimetry extrapolation of 64Cu/177Lu 1C1m-Fc anti-TEM-1 for theranostic application in soft-tissue sarcomas. We adopt four methods; direct mice-to-human extrapolation (M1); dosimetry extrapolation considering a relative mass scaling factor (M2), application of a metabolic scaling factor (M3) and combination of M2 and M3 (M4). Predicted in-human dosimetry for the [64Cu]Cu-1C1m-Fc resulted in an effective dose of 0.05 mSv/MBq. Absorbed dose (AD) extrapolation for the [177Lu]Lu-1C1m-Fc indicated that the AD of 2 Gy and 4 Gy to the red-marrow and total-body can be reached with 5-10 GBq and 25-30 GBq of therapeutic activity administration respectively depending on applied dosimetry method. Dosimetry extrapolation methods provided significantly different absorbed doses in organs. Dosimetry properties for the [64Cu]Cu-1C1m-Fc are suitable for a diagnostic in-human use. The therapeutic application of [177Lu]Lu-1C1m-Fc presents challenges and would benefit from further assessments in animals' models such as dogs before moving into the clinic.
Collapse
Affiliation(s)
- Silvano Gnesin
- Institute of Radiation Physics, Lausanne University Hospital and University of Lausanne, 1011, Lausanne, Switzerland
| | - Nicolas Chouin
- Inserm, CNRS, University of Angers, Oniris, CRCI2NA, University of Nantes, Nantes, France
| | - Michel Cherel
- CHU Nantes, CNRS, Inserm, CRCINA, University of Nantes, 44000, Nantes, France
| | - Steven Mark Dunn
- LAbCore, Ludwig Institute for Cancer Research, Lausanne University Hospital and University of Lausanne, 1066, Epalinges, Switzerland
| | - Niklaus Schaefer
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital and University of Lausanne, Rue du Bugnon 46, 1011, Lausanne, Switzerland
| | | | - John O Prior
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital and University of Lausanne, Rue du Bugnon 46, 1011, Lausanne, Switzerland.
| | - Judith Anna Delage
- Radiopharmacy Unit, Department of Pharmacy, Lausanne University Hospital and University of Lausanne, 1011, Lausanne, Switzerland
| |
Collapse
|
33
|
Development of a voxel S-value database for patient internal radiation dosimetry. Phys Med 2023; 106:102519. [PMID: 36641901 DOI: 10.1016/j.ejmp.2022.102519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 12/13/2022] [Accepted: 12/27/2022] [Indexed: 01/15/2023] Open
Abstract
PURPOSE Personalized dosimetry with high accuracy drew great attention in clinical practices. Voxel S-value (VSV) convolution has been proposed to speed up absorbed dose calculations. However, the VSV method is efficient for personalized internal radiation dosimetry only when there are pre-calculated VSVs of the radioisotope. In this work, we propose a new method for VSV calculation based on the developed mono-energetic particle VSV database of γ, β, α, and X-ray for any radioisotopes. METHODS Mono-energetic VSV database for γ, β, α, and X-ray was calculated using Monte Carlo methods. Radiation dose was first calculated based on mono-energetic VSVs for [F-18]-FDG in 10 patients. The estimated doses were compared with the values obtained from direct Monte Carlo simulation for validation of the proposed method. The number of VSVs used in calculation was optimized based on the estimated dose accuracy and computation time. RESULTS The generated VSVs showed a great consistency with the results calculated using direct Monte Carlo simulation. For [F-18]-FDG, the proposed VSV method with number of VSV of 9 shows the best relative average organ absorbed dose uncertainty of 3.25% while the calculation time was reduced by 99% and 97% compared to the Monte Carlo simulation and traditional multiple VSV methods, respectively. CONCLUSIONS In this work, we provided a method to generate the VSV kernels for any radioisotope based on the pre-calculated mono-energetic VSV database and significantly reduced the time cost for the multiple VSVs dosimetry approach. A software was developed to generate VSV kernels for any radioisotope in 19 mediums.
Collapse
|
34
|
Parihar AS, Hofman MS, Iravani A. 177Lu-Prostate-specific Membrane Antigen Radioligand Therapy in Patients with Metastatic Castration-resistant Prostate Cancer. Radiology 2023; 306:e220859. [PMID: 36125377 DOI: 10.1148/radiol.220859] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
A 76-year-old man with metastatic castration-resistant prostate carcinoma progressing with antiandrogen and taxane therapy was treated with lutetium 177 prostate-specific membrane antigen (PSMA)-617 and showed marked biochemical and imaging response, with improvement in clinical status and osseous pain. A summary of nuclear medicine theranostics with emphasis on PSMA targeting agents is presented.
Collapse
Affiliation(s)
- Ashwin Singh Parihar
- From the Mallinckrodt Institute of Radiology, Washington University School of Medicine, 4525 Scott Ave, Suite 3433, MIR East Building, St Louis, MO 63110 (A.S.P., A.I.); and Department of Molecular Imaging and Therapeutic Nuclear Medicine, Peter MacCallum Cancer Centre, Melbourne, Australia (M.S.H.)
| | - Michael S Hofman
- From the Mallinckrodt Institute of Radiology, Washington University School of Medicine, 4525 Scott Ave, Suite 3433, MIR East Building, St Louis, MO 63110 (A.S.P., A.I.); and Department of Molecular Imaging and Therapeutic Nuclear Medicine, Peter MacCallum Cancer Centre, Melbourne, Australia (M.S.H.)
| | - Amir Iravani
- From the Mallinckrodt Institute of Radiology, Washington University School of Medicine, 4525 Scott Ave, Suite 3433, MIR East Building, St Louis, MO 63110 (A.S.P., A.I.); and Department of Molecular Imaging and Therapeutic Nuclear Medicine, Peter MacCallum Cancer Centre, Melbourne, Australia (M.S.H.)
| |
Collapse
|
35
|
Pirozzi Palmese V, D'Ambrosio L, Di Gennaro F, Maisto C, de Marino R, Morisco A, Coluccia S, Di Gennaro P, De Lauro F, Raddi M, Gaballo P, Tafuto S, Celentano E, Lastoria S. A comparison of simplified protocols of personalized dosimetry in NEN patients treated by radioligand therapy (RLT) with [ 177Lu]Lu-DOTATATE to favor its use in clinical practice. Eur J Nucl Med Mol Imaging 2023; 50:1753-1764. [PMID: 36688980 PMCID: PMC10119237 DOI: 10.1007/s00259-023-06112-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 01/05/2023] [Indexed: 01/24/2023]
Abstract
The role of internal dosimetry is usually proposed for investigational purposes in patients treated by RLT, even if its application is not yet the standard method in clinical practice. This limited use is partially justified by several concomitant factors that make calculations a complex process. Therefore, simplified dosimetry protocols are required. METHODS In our study, dosimetric evaluations were performed in thirty patients with NENs who underwent RLT with [177Lu]Lu-DOTATATE. The reference method (M0) calculated the cumulative absorbed dose performing dosimetry after each of the four cycles. Obtained data were employed to assess the feasibility of simplified protocols: defining the dosimetry only after the first cycle (M1) and after the first and last one (M2). RESULTS The mean differences of the cumulative absorbed doses between M1 and M0 were - 10% for kidney, - 5% for spleen, + 34% for liver, + 13% for red marrow, and + 37% for tumor lesions. Conversely, differences lower than ± 10% were measured between M2 and M0. CONCLUSION Cumulative absorbed doses obtained with the M2 protocol resembled the doses calculated by M0, while the M1 protocol overestimated the absorbed doses in all organs at risk, except for the spleen.
Collapse
Affiliation(s)
| | - Laura D'Ambrosio
- S.C. Medicina Nucleare E Terapia Radiometabolica, INT IRCCS Fondazione G. Pascale, Naples, Italy
| | - Francesca Di Gennaro
- S.C. Medicina Nucleare E Terapia Radiometabolica, INT IRCCS Fondazione G. Pascale, Naples, Italy
| | - Costantina Maisto
- S.C. Medicina Nucleare E Terapia Radiometabolica, INT IRCCS Fondazione G. Pascale, Naples, Italy
| | - Roberta de Marino
- S.C. Medicina Nucleare E Terapia Radiometabolica, INT IRCCS Fondazione G. Pascale, Naples, Italy
| | - Anna Morisco
- S.C. Medicina Nucleare E Terapia Radiometabolica, INT IRCCS Fondazione G. Pascale, Naples, Italy
| | - Sergio Coluccia
- S.C. Epidemiologia E Biostatistica, INT IRCCS Fondazione G. Pascale, Naples, Italy
| | | | - Francesco De Lauro
- S.C. Medicina Nucleare E Terapia Radiometabolica, INT IRCCS Fondazione G. Pascale, Naples, Italy
| | - Marco Raddi
- S.C. Medicina Nucleare E Terapia Radiometabolica, INT IRCCS Fondazione G. Pascale, Naples, Italy
| | - Paolo Gaballo
- S.C. Medicina Nucleare E Terapia Radiometabolica, INT IRCCS Fondazione G. Pascale, Naples, Italy
| | - Salvatore Tafuto
- S.C. Sarcomi E Tumori Rari, INT IRCCS Fondazione G. Pascale, Naples, Italy
| | - Egidio Celentano
- S.C. Epidemiologia E Biostatistica, INT IRCCS Fondazione G. Pascale, Naples, Italy
| | - Secondo Lastoria
- S.C. Medicina Nucleare E Terapia Radiometabolica, INT IRCCS Fondazione G. Pascale, Naples, Italy.
| |
Collapse
|
36
|
Akter A, Lyons O, Mehra V, Isenman H, Abbate V. Radiometal chelators for infection diagnostics. FRONTIERS IN NUCLEAR MEDICINE (LAUSANNE, SWITZERLAND) 2023; 2:1058388. [PMID: 37388440 PMCID: PMC7614707 DOI: 10.3389/fnume.2022.1058388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/01/2023]
Abstract
Infection of native tissues or implanted devices is common, but clinical diagnosis is frequently difficult and currently available noninvasive tests perform poorly. Immunocompromised individuals (for example transplant recipients, or those with cancer) are at increased risk. No imaging test in clinical use can specifically identify infection, or accurately differentiate bacterial from fungal infections. Commonly used [18F]fluorodeoxyglucose (18FDG) positron emission computed tomography (PET/CT) is sensitive for infection, but limited by poor specificity because increased glucose uptake may also indicate inflammation or malignancy. Furthermore, this tracer provides no indication of the type of infective agent (bacterial, fungal, or parasitic). Imaging tools that directly and specifically target microbial pathogens are highly desirable to improve noninvasive infection diagnosis and localization. A growing field of research is exploring the utility of radiometals and their chelators (siderophores), which are small molecules that bind radiometals and form a stable complex allowing sequestration by microbes. This radiometal-chelator complex can be directed to a specific microbial target in vivo, facilitating anatomical localization by PET or single photon emission computed tomography. Additionally, bifunctional chelators can further conjugate therapeutic molecules (e.g., peptides, antibiotics, antibodies) while still bound to desired radiometals, combining specific imaging with highly targeted antimicrobial therapy. These novel therapeutics may prove a useful complement to the armamentarium in the global fight against antimicrobial resistance. This review will highlight current state of infection imaging diagnostics and their limitations, strategies to develop infection-specific diagnostics, recent advances in radiometal-based chelators for microbial infection imaging, challenges, and future directions to improve targeted diagnostics and/or therapeutics.
Collapse
Affiliation(s)
- Asma Akter
- Department of Analytical, Environmental and Forensic Sciences, King’s College London, London, United Kingdom
| | - Oliver Lyons
- Vascular Endovascular and Transplant Surgery, Christchurch Public Hospital, Christchurch, New Zealand
- Department of Surgery, University of Otago, Christchurch, New Zealand
| | - Varun Mehra
- Department of Hematology, King’s College Hospital NHS Foundation Trust, London, United Kingdom
| | - Heather Isenman
- Department of Infectious Diseases, General Medicine, Christchurch Hospital, Christchurch, New Zealand
| | - Vincenzo Abbate
- Department of Analytical, Environmental and Forensic Sciences, King’s College London, London, United Kingdom
| |
Collapse
|
37
|
Salerno KE, Roy S, Ribaudo C, Fisher T, Patel RB, Mena E, Escorcia FE. A Primer on Radiopharmaceutical Therapy. Int J Radiat Oncol Biol Phys 2023; 115:48-59. [PMID: 35970373 PMCID: PMC9772089 DOI: 10.1016/j.ijrobp.2022.08.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/25/2022] [Accepted: 08/03/2022] [Indexed: 12/24/2022]
Abstract
The goal of this article is to serve as a primer for the United States-based radiation oncologist who may be interested in learning more about radiopharmaceutical therapy (RPT). Specifically, we define RPT, review the data behind its current and anticipated indications, and discuss important regulatory considerations for incorporating it into clinical practice. RPT represents an opportunity for radiation oncologists to leverage 2 key areas of expertise, namely therapeutic radiation therapy and oncology, and apply them in a distinct context in collaboration with nuclear medicine and medical oncology colleagues. Although not every radiation oncologist will incorporate RPT into their day-to-day practice, it is important to understand the role for this modality and how it can be appropriately used in select patients.
Collapse
Affiliation(s)
- Kilian E Salerno
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Soumyajit Roy
- Radiation Oncology Department, Rush Medical Center, Chicago, Illinois
| | - Cathy Ribaudo
- Division of Radiation Safety, National Institutes of Health, Bethesda, Maryland
| | - Teresa Fisher
- Division of Radiation Safety, National Institutes of Health, Bethesda, Maryland
| | - Ravi B Patel
- Radiation Oncology Department, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Esther Mena
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Freddy E Escorcia
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland; Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
38
|
Kerr CP, Grudzinski JJ, Nguyen TP, Hernandez R, Weichert JP, Morris ZS. Developments in Combining Targeted Radionuclide Therapies and Immunotherapies for Cancer Treatment. Pharmaceutics 2022; 15:128. [PMID: 36678756 PMCID: PMC9865370 DOI: 10.3390/pharmaceutics15010128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 12/26/2022] [Accepted: 12/27/2022] [Indexed: 01/01/2023] Open
Abstract
Targeted radionuclide therapy (TRT) and immunotherapy are rapidly growing classes of cancer treatments. Basic, translational, and clinical research are now investigating therapeutic combinations of these agents. In comparison to external beam radiation therapy (EBRT), TRT has the unique advantage of treating all disease sites following intravenous injection and selective tumor uptake and retention-a particularly beneficial property in metastatic disease settings. The therapeutic value of combining radiation therapy with immune checkpoint blockade to treat metastases has been demonstrated in preclinical studies, whereas results of clinical studies have been mixed. Several clinical trials combining TRT and immune checkpoint blockade have been initiated based on preclinical studies combining these with EBRT and/or TRT. Despite the interest in translation of TRT and immunotherapy combinations, many questions remain surrounding the mechanisms of interaction and the optimal approach to clinical implementation of these combinations. This review highlights the mechanisms of interaction between anti-tumor immunity and radiation therapy and the status of basic and translational research and clinical trials investigating combinations of TRT and immunotherapies.
Collapse
Affiliation(s)
- Caroline P. Kerr
- Department of Radiology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Human Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Joseph J. Grudzinski
- Department of Radiology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Thanh Phuong Nguyen
- Department of Human Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Reinier Hernandez
- Department of Medical Physics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Jamey P. Weichert
- Department of Radiology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Zachary S. Morris
- Department of Human Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
39
|
Li WB, Bouvier-Capely C, Saldarriaga Vargas C, Andersson M, Madas B. Heterogeneity of dose distribution in normal tissues in case of radiopharmaceutical therapy with alpha-emitting radionuclides. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2022; 61:579-596. [PMID: 36239799 PMCID: PMC9630198 DOI: 10.1007/s00411-022-01000-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 10/06/2022] [Indexed: 05/10/2023]
Abstract
Heterogeneity of dose distribution has been shown at different spatial scales in diagnostic nuclear medicine. In cancer treatment using new radiopharmaceuticals with alpha-particle emitters, it has shown an extensive degree of dose heterogeneity affecting both tumour control and toxicity of organs at risk. This review aims to provide an overview of generalized internal dosimetry in nuclear medicine and highlight the need of consideration of the dose heterogeneity within organs at risk. The current methods used for patient dosimetry in radiopharmaceutical therapy are summarized. Bio-distribution and dose heterogeneities of alpha-particle emitting pharmaceutical 223Ra (Xofigo) within bone tissues are presented as an example. In line with the strategical research agendas of the Multidisciplinary European Low Dose Initiative (MELODI) and the European Radiation Dosimetry Group (EURADOS), future research direction of pharmacokinetic modelling and dosimetry in patient radiopharmaceutical therapy are recommended.
Collapse
Affiliation(s)
- Wei Bo Li
- Helmholtz Zentrum München-German Research Center for Environmental Health (GmbH), Institute of Radiation Medicine, Neuherberg, Germany.
| | - Céline Bouvier-Capely
- Institut de Radioprotection et Sûreté Nucléaire (IRSN), PSE-SANTE/SESANE/LRSI, Fontenay-aux-Roses, France
| | - Clarita Saldarriaga Vargas
- Radiation Protection Dosimetry and Calibrations, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
- In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, Brussels, Belgium
| | - Michelle Andersson
- Radiation Protection Dosimetry and Calibrations, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
- Medical Physics Department, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium
| | - Balázs Madas
- Environmental Physics Department, Centre for Energy Research, Budapest, Hungary
| |
Collapse
|
40
|
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: 15] [Impact Index Per Article: 5.0] [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
|
41
|
Soulek DK, Mastascusa NJ, Martin ME, Graves SA. Practical Considerations for Implementation of 177Lu-DOTATATE Neuroendocrine Tumor Treatment Programs. J Nucl Med Technol 2022; 50:jnmt.122.263813. [PMID: 35701215 DOI: 10.2967/jnmt.122.263813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 06/08/2022] [Indexed: 11/16/2022] Open
Abstract
The 2018 FDA approval of 177Lu-DOTATATE for the treatment of somatostatin receptor-positive (SSTR) neuroendocrine tumors (NETs) represents a paradigm shifting approach to cancer treatments around the globe. Gastroenteropancreatic (GEP) NETs overexpress the somatostatin subtype receptor 2, which is now exploited for receptor-based imaging and therapy, thus generating significant progress in the diagnosis and treatment of this orphan disease. The recent FDA approval of receptor-based PET radiopharmaceuticals and a new peptide receptor radiopharmaceutical therapy (PRRT), 177Lu-DOTATATE, has dramatically impacted NET patient management. The focus of this paper is to review clinical considerations associated with implementing a 177Lu-DOTATATE program. We review receptor-based NET radiopharmaceuticals, 177Lu-DOTATATE patient selection criteria, administration methods, clinical, regulatory, and radiation safety considerations, technical factors, tissue dosimetry, and reimbursement guidelines.
Collapse
|
42
|
Yusufaly TI. Extending the relative seriality formalism for interpretable deep learning of normal tissue complication probability models. MACHINE LEARNING: SCIENCE AND TECHNOLOGY 2022. [DOI: 10.1088/2632-2153/ac6932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Abstract
We formally demonstrate that the relative seriality (RS) model of normal tissue complication probability (NTCP) can be recast as a simple neural network with one convolutional and one pooling layer. This approach enables us to systematically construct deep relative seriality networks (DRSNs), a new class of mechanistic generalizations of the RS model with radiobiologically interpretable parameters amenable to deep learning. To demonstrate the utility of this formulation, we analyze a simplified example of xerostomia due to irradiation of the parotid gland during alpha radiopharmaceutical therapy. Using a combination of analytical calculations and numerical simulations, we show for both the RS and DRSN cases that the ability of the neural network to generalize without overfitting is tied to ‘stiff’ and ‘sloppy’ directions in the parameter space of the mechanistic model. These results serve as proof-of-concept for radiobiologically interpretable deep learning of NTCP, while simultaneously yielding insight into how such techniques can robustly generalize beyond the training set despite uncertainty in individual parameters.
Collapse
|
43
|
Park EA, Graves SA, Menda Y. The Impact of Radiopharmaceutical Therapy on Renal Function. Semin Nucl Med 2022; 52:467-474. [DOI: 10.1053/j.semnuclmed.2022.02.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 02/13/2022] [Accepted: 02/20/2022] [Indexed: 11/11/2022]
|
44
|
Danieli R, Milano A, Gallo S, Veronese I, Lascialfari A, Indovina L, Botta F, Ferrari M, Cicchetti A, Raspanti D, Cremonesi M. Personalized Dosimetry in Targeted Radiation Therapy: A Look to Methods, Tools and Critical Aspects. J Pers Med 2022; 12:205. [PMID: 35207693 PMCID: PMC8874397 DOI: 10.3390/jpm12020205] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/13/2022] [Accepted: 01/14/2022] [Indexed: 12/10/2022] Open
Abstract
Targeted radiation therapy (TRT) is a strategy increasingly adopted for the treatment of different types of cancer. The urge for optimization, as stated by the European Council Directive (2013/59/EURATOM), requires the implementation of a personalized dosimetric approach, similar to what already happens in external beam radiation therapy (EBRT). The purpose of this paper is to provide a thorough introduction to the field of personalized dosimetry in TRT, explaining its rationale in the context of optimization and describing the currently available methodologies. After listing the main therapies currently employed, the clinical workflow for the absorbed dose calculation is described, based on works of the most experienced authors in the literature and recent guidelines. Moreover, the widespread software packages for internal dosimetry are presented and critical aspects discussed. Overall, a selection of the most important and recent articles about this topic is provided.
Collapse
Affiliation(s)
- Rachele Danieli
- Dipartimento di Fisica, Università degli Studi di Pavia, Via Bassi 6, 27100 Pavia, Italy;
| | - Alessia Milano
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo F. Vito 1, 00168 Roma, Italy;
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Roma, Italy
| | - Salvatore Gallo
- Dipartimento di Fisica “Aldo Pontremoli”, Università degli Studi di Milano, Via Celoria 16, 20133 Milano, Italy; (S.G.); (I.V.)
- INFN Sezione di Milano, Via Celoria 16, 20133 Milano, Italy
| | - Ivan Veronese
- Dipartimento di Fisica “Aldo Pontremoli”, Università degli Studi di Milano, Via Celoria 16, 20133 Milano, Italy; (S.G.); (I.V.)
- INFN Sezione di Milano, Via Celoria 16, 20133 Milano, Italy
| | - Alessandro Lascialfari
- INFN-Pavia Unit, Department of Physics, University of Pavia, Via Bassi 6, 27100 Pavia, Italy;
| | - Luca Indovina
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo F. Vito 1, 00168 Roma, Italy;
| | - Francesca Botta
- Medical Physics Unit, European Institute of Oncology IRCCS, Via Giuseppe Ripamonti 435, 20141 Milano, Italy; (F.B.); (M.F.)
| | - Mahila Ferrari
- Medical Physics Unit, European Institute of Oncology IRCCS, Via Giuseppe Ripamonti 435, 20141 Milano, Italy; (F.B.); (M.F.)
| | - Alessandro Cicchetti
- Prostate Cancer Program, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian, 1, 20133 Milano, Italy;
| | - Davide Raspanti
- Temasinergie S.p.A., Via Marcello Malpighi 120, 48018 Faenza, Italy;
| | - Marta Cremonesi
- Radiation Research Unit, European Institute of Oncology IRCCS, Via Giuseppe Ripamonti 435, 20141 Milano, Italy;
| |
Collapse
|
45
|
Wahl RL, Sunderland J. Radiopharmaceutical Dosimetry for Cancer Therapy: From Theory to Practice. J Nucl Med 2021; 62:1S-2S. [PMID: 34857618 DOI: 10.2967/jnumed.121.263273] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Indexed: 11/16/2022] Open
Affiliation(s)
- Richard L Wahl
- Mallinckrodt Institute of Radiology, St. Louis, Missouri; and
| | | |
Collapse
|