1
|
Kręcisz P, Stefańska K, Studziński J, Pitucha M, Czylkowska A, Szymański P. Radiocopper in Radiopharmacy and Medical Use: Current Status and Perspective. J Med Chem 2025; 68:2356-2376. [PMID: 39895089 PMCID: PMC11831595 DOI: 10.1021/acs.jmedchem.4c02885] [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: 11/24/2024] [Revised: 01/21/2025] [Accepted: 01/27/2025] [Indexed: 02/04/2025]
Abstract
Of the 32 known copper isotopes, some have interesting properties for nuclear medicine, for example the short-lived 60Cu, 61Cu, 62Cu, the moderate long-lived 64Cu and the long-lived 67Cu. Due to their emission properties, copper isotopes are suitable for both imaging diagnostics (60Cu, 61Cu, 62Cu, 64Cu) and targeted radiotherapy (64Cu and 67Cu). As their chemical properties are virtually identical, a single radiopharmaceutical structure can be labeled with different isotopes, depending on the clinical application. This, combined with the ability to combine radioisotopes with different nuclear properties with the same ligand, makes them extremely versatile. The purpose of this review is to introduce the world of copper radiopharmaceuticals and to summarize recent advances in methods for producing copper radioisotopes and the preclinical research of radiopharmaceuticals labeled with copper radioisotopes.
Collapse
Affiliation(s)
- Paweł Kręcisz
- Department
of Pharmaceutical Chemistry, Drug Analyses and Radiopharmacy, Faculty
of Pharmacy, Medical University of Lodz, Muszyńskiego 1, 90-151 Lodz, Poland
| | - Katarzyna Stefańska
- Department
of Pharmaceutical Chemistry, Drug Analyses and Radiopharmacy, Faculty
of Pharmacy, Medical University of Lodz, Muszyńskiego 1, 90-151 Lodz, Poland
| | - Jakub Studziński
- Department
of Pharmaceutical Chemistry, Drug Analyses and Radiopharmacy, Faculty
of Pharmacy, Medical University of Lodz, Muszyńskiego 1, 90-151 Lodz, Poland
| | - Monika Pitucha
- Independent
Radiopharmacy Unit, Faculty of Pharmacy, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland
| | - Agnieszka Czylkowska
- Institute
of General and Ecological Chemistry, Faculty of Chemistry, Lodz University of Technology, Żeromskiego 116, 90-924 Lodz, Poland
| | - Paweł Szymański
- Department
of Pharmaceutical Chemistry, Drug Analyses and Radiopharmacy, Faculty
of Pharmacy, Medical University of Lodz, Muszyńskiego 1, 90-151 Lodz, Poland
- Department
of Radiobiology and Radiation Protection, Military Institute of Hygiene and Epidemiology, Kozielska 4, 01-163 Warsaw, Poland
| |
Collapse
|
2
|
Azadinejad H, Farhadi Rad M, Shariftabrizi A, Rahmim A, Abdollahi H. Optimizing Cancer Treatment: Exploring the Role of AI in Radioimmunotherapy. Diagnostics (Basel) 2025; 15:397. [PMID: 39941326 PMCID: PMC11816985 DOI: 10.3390/diagnostics15030397] [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: 12/10/2024] [Revised: 01/14/2025] [Accepted: 01/22/2025] [Indexed: 02/16/2025] Open
Abstract
Radioimmunotherapy (RIT) is a novel cancer treatment that combines radiotherapy and immunotherapy to precisely target tumor antigens using monoclonal antibodies conjugated with radioactive isotopes. This approach offers personalized, systemic, and durable treatment, making it effective in cancers resistant to conventional therapies. Advances in artificial intelligence (AI) present opportunities to enhance RIT by improving precision, efficiency, and personalization. AI plays a critical role in patient selection, treatment planning, dosimetry, and response assessment, while also contributing to drug design and tumor classification. This review explores the integration of AI into RIT, emphasizing its potential to optimize the entire treatment process and advance personalized cancer care.
Collapse
Affiliation(s)
- Hossein Azadinejad
- Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah 6714869914, Iran;
| | - Mohammad Farhadi Rad
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah 6715847141, Iran
| | - Ahmad Shariftabrizi
- Department of Radiology, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA;
| | - Arman Rahmim
- Department of Radiology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 0B4, Canada
- Department of Physics and Astronomy, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Hamid Abdollahi
- Department of Radiology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 0B4, Canada
| |
Collapse
|
3
|
Hong J, Bae S, Cavinato L, Seifert R, Ryhiner M, Rominger A, Erlandsson K, Wilks M, Normandin M, El-Fakhri G, Choi H, Shi K. Deciphering the effects of radiopharmaceutical therapy in the tumor microenvironment of prostate cancer: an in-silico exploration with spatial transcriptomics. Theranostics 2024; 14:7122-7139. [PMID: 39629134 PMCID: PMC11610134 DOI: 10.7150/thno.99516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 09/26/2024] [Indexed: 12/06/2024] Open
Abstract
Radiopharmaceutical therapy (RPT) is an emerging prostate cancer treatment that delivers radiation to specific molecules within the tumor microenvironment (TME), causing DNA damage and cell death. Given TME heterogeneity, it's crucial to explore RPT dosimetry and biological impacts at the cellular level. We integrated spatial transcriptomics (ST) with computational modeling to investigate the effects of RPT targeting prostate-specific membrane antigen (PSMA), fibroblast activation protein (FAP), and gastrin-releasing peptide receptor (GRPR) each labelled with beta-emitting lutetium-177 (177Lu) and alpha-emitting actinium-225 (225Ac). Methods: Three ST datasets from primary tissue samples of two prostate cancer patients were obtained. From these datasets, we extracted gene expressions, including FOLH1, GRPR, FAP, and Harris Hypoxia, and estimated the proportions of different cell types-epithelial, endothelial, and prostate cancer (PC) cells-in the corresponding ST spots. We computed the spatiotemporal distribution of each RPT targeting PSMA, FAP, and GRPR at each ST spot by solving the partial differential equation (PDE) using a convection-reaction-diffusion (CRD) model, assuming similar pharmacokinetic parameters across all ligands. A well-established physiologically based pharmacokinetic (PBPK) model was used to simulate the input function in the prostate, carefully calibrated to deliver 10 Gy to the prostate tumor over 20 days. Dosimetry was estimated using the Medical Internal Radiation Dose (MIRD) formalism, applying the dose point kernels (DVK) method. The survival probability was estimated using the linear quadratic model, applied to both beta-emitting RPT labeled with 177Lu and 225Ac. A modified linear quadratic model was used to estimate the bioeffect of the alpha-emitting RPT. Results: The results demonstrate distinct dose-response and efficacy patterns across ST samples, with FAP-targeted RPT exhibiting limited effectiveness in tumor cell-rich areas compared to PSMA- and GRPR-targeted therapies. GRPR-targeted RPT showed higher resistance in hypoxic regions relative to the other therapies. Additionally, 225Ac-labeled RPT was more effective overall than 177Lu-labeled RPT, especially in areas with low cancer-cell fraction or high hypoxia. The findings suggest that a combination of 225Ac-labeled FAP- and PSMA-targeted RPT offers the best therapeutic strategy. Conclusion: The proposed method, which combines ST and computational modeling to determine the dosimetry and cell survival probability of RPT in the TME, holds promise for identifying optimal personalized RPT strategies.
Collapse
Affiliation(s)
- Jimin Hong
- Department of Nuclear Medicine, Inselspital, University of Bern, Bern, Switzerland
- Gordon Center of Medical Imaging, Massachusetts General Hospital, Massachusetts, United States of America
| | - Sungwoo Bae
- Institute of Radiation Medicine, Medical Research Center, Seoul National University College of Medicine, Seoul, South Korea
- Portrai, Inc., 78-18, Dongsulla-gil, Jongno-gu, Seoul, South Korea
| | - Lara Cavinato
- Department of Mathematics, Politecnico de Milano, Milan, Italy
| | - Robert Seifert
- Department of Nuclear Medicine, Inselspital, University of Bern, Bern, Switzerland
| | - Marc Ryhiner
- Department of Nuclear Medicine, Inselspital, University of Bern, Bern, Switzerland
| | - Axel Rominger
- Department of Nuclear Medicine, Inselspital, University of Bern, Bern, Switzerland
| | - Kjell Erlandsson
- Institute of Nuclear Medicine, University College London, London, United Kingdom
| | - Moses Wilks
- PET Center at Yale School of Medicine, Yale University, New Havens, United States of America
| | - Marc Normandin
- PET Center at Yale School of Medicine, Yale University, New Havens, United States of America
| | - Georges El-Fakhri
- PET Center at Yale School of Medicine, Yale University, New Havens, United States of America
| | - Hongyoon Choi
- Institute of Radiation Medicine, Medical Research Center, Seoul National University College of Medicine, Seoul, South Korea
- Portrai, Inc., 78-18, Dongsulla-gil, Jongno-gu, Seoul, South Korea
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Kuangyu Shi
- Department of Nuclear Medicine, Inselspital, University of Bern, Bern, Switzerland
| |
Collapse
|
4
|
Liatsou I, Fu Y, Li Z, Hasan M, Guo X, Yu J, Piccolo J, Cartee A, Wang H, Du Y, Bryan J, Gabrielson K, Kraitchman DL, Sgouros G. Therapeutic efficacy of an alpha-particle emitter labeled anti-GD2 humanized antibody against osteosarcoma-a proof of concept study. Eur J Nucl Med Mol Imaging 2024; 51:1409-1420. [PMID: 38108831 DOI: 10.1007/s00259-023-06528-2] [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: 08/07/2023] [Accepted: 11/14/2023] [Indexed: 12/19/2023]
Abstract
PURPOSE Current treatments for osteosarcoma (OS) have a poor prognosis, particularly for patients with metastasis and recurrence, underscoring an urgent need for new targeted therapies to improve survival. Targeted alpha-particle therapy selectively delivers cytotoxic payloads to tumors with radiolabeled molecules that recognize tumor-associated antigens. We have recently demonstrated the potential of an FDA approved, humanized anti-GD2 antibody, hu3F8, as a targeted delivery vector for radiopharmaceutical imaging of OS. The current study aims to advance this system for alpha-particle therapy of OS. METHODS The hu3F8 antibody was radiolabeled with actinium-225, and the safety and therapeutic efficacy of the [225Ac]Ac-DOTA-hu3F8 were evaluated in both orthotopic murine xenografts of OS and spontaneously occurring OS in canines. RESULTS Significant antitumor activity was proven in both cases, leading to improved overall survival. In the murine xenograft's case, tumor growth was delayed by 16-18 days compared to the untreated cohort as demonstrated by bioluminescence imaging. The results were further validated with magnetic resonance imaging at 33 days after treatment, and microcomputed tomography and planar microradiography post-mortem. Histological evaluations revealed radiation-induced renal toxicity, manifested as epithelial cell karyomegaly and suggestive polyploidy in the kidneys, suggesting rapid recovery of renal function after radiation damage. Treatment of the two canine patients delayed the progression of metastatic spread, with an overall survival time of 211 and 437 days and survival beyond documented metastasis of 111 and 84 days, respectively. CONCLUSION This study highlights the potential of hu3F8-based alpha-particle therapy as a promising treatment strategy for OS.
Collapse
Affiliation(s)
- Ioanna Liatsou
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Yingli Fu
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zhi Li
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mahmud Hasan
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Xin Guo
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jing Yu
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Joseph Piccolo
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Allison Cartee
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hao Wang
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yong Du
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jeffrey Bryan
- Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, MO, USA
| | - Kathleen Gabrielson
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dara L Kraitchman
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - George Sgouros
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
5
|
Piranfar A, Souri M, Rahmim A, Soltani M. Localized radiotherapy of solid tumors using radiopharmaceutical loaded implantable system: insights from a mathematical model. Front Oncol 2024; 14:1320371. [PMID: 38559559 PMCID: PMC10979490 DOI: 10.3389/fonc.2024.1320371] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 02/06/2024] [Indexed: 04/04/2024] Open
Abstract
Introduction Computational models yield valuable insights into biological interactions not fully elucidated by experimental approaches. This study investigates an innovative spatiotemporal model for simulating the controlled release and dispersion of radiopharmaceutical therapy (RPT) using 177Lu-PSMA, a prostate-specific membrane antigen (PSMA) targeted radiopharmaceutical, within solid tumors via a dual-release implantable delivery system. Local delivery of anticancer agents presents a strategic approach to mitigate adverse effects while optimizing therapeutic outcomes. Methods This study evaluates various factors impacting RPT efficacy, including hypoxia region extension, binding affinity, and initial drug dosage, employing a novel 3-dimensional computational model. Analysis gauges the influence of these factors on radiopharmaceutical agent concentration within the tumor microenvironment. Furthermore, spatial and temporal radiopharmaceutical distribution within both the tumor and surrounding tissue is explored. Results Analysis indicates a significantly higher total concentration area under the curve within the tumor region compared to surrounding normal tissue. Moreover, drug distribution exhibits notably superior efficacy compared to the radiation source. Additionally, low microvascular density in extended hypoxia regions enhances drug availability, facilitating improved binding to PSMA receptors and enhancing therapeutic effectiveness. Reductions in the dissociation constant (KD) lead to heightened binding affinity and increased internalized drug concentration. Evaluation of initial radioactivities (7.1×107, 7.1×108, and 7.1×109 [Bq]) indicates that an activity of 7.1×108 [Bq] offers a favorable balance between tumor cell elimination and minimal impact on normal tissues. Discussion These findings underscore the potential of localized radiopharmaceutical delivery strategies and emphasize the crucial role of released drugs relative to the radiation source (implant) in effective tumor treatment. Decreasing the proximity of the drug to the microvascular network and enhancing its distribution within the tumor promote a more effective therapeutic outcome. The study furnishes valuable insights for future experimental investigations and clinical trials, aiming to refine medication protocols and minimize reliance on in vivo testing.
Collapse
Affiliation(s)
- Anahita Piranfar
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | - Mohammad Souri
- Department of NanoBiotechnology, Pasteur Institute of Iran, Tehran, Iran
| | - Arman Rahmim
- Departments of Radiology and Physics, University of British Columbia, Vancouver, BC, Canada
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Madjid Soltani
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
- Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, ON, Canada
- Centre for Biotechnology and Bioengineering (CBB), University of Waterloo, Waterloo, ON, Canada
- Centre for Sustainable Business, International Business University, Toronto, ON, Canada
| |
Collapse
|
6
|
Fele-Paranj A, Saboury B, Uribe C, Rahmim A. Physiologically based radiopharmacokinetic (PBRPK) modeling to simulate and analyze radiopharmaceutical therapies: studies of non-linearities, multi-bolus injections, and albumin binding. EJNMMI Radiopharm Chem 2024; 9:6. [PMID: 38252191 PMCID: PMC10803696 DOI: 10.1186/s41181-023-00236-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 12/22/2023] [Indexed: 01/23/2024] Open
Abstract
BACKGROUND We aimed to develop a publicly shared computational physiologically based pharmacokinetic (PBPK) model to reliably simulate and analyze radiopharmaceutical therapies (RPTs), including probing of hot-cold ligand competitions as well as alternative injection scenarios and drug designs, towards optimal therapies. RESULTS To handle the complexity of PBPK models (over 150 differential equations), a scalable modeling notation called the "reaction graph" is introduced, enabling easy inclusion of various interactions. We refer to this as physiologically based radiopharmacokinetic (PBRPK) modeling, fine-tuned specifically for radiopharmaceuticals. As three important applications, we used our PBRPK model to (1) study the effect of competition between hot and cold species on delivered doses to tumors and organs at risk. In addition, (2) we evaluated an alternative paradigm of utilizing multi-bolus injections in RPTs instead of prevalent single injections. Finally, (3) we used PBRPK modeling to study the impact of varying albumin-binding affinities by ligands, and the implications for RPTs. We found that competition between labeled and unlabeled ligands can lead to non-linear relations between injected activity and the delivered dose to a particular organ, in the sense that doubling the injected activity does not necessarily result in a doubled dose delivered to a particular organ (a false intuition from external beam radiotherapy). In addition, we observed that fractionating injections can lead to a higher payload of dose delivery to organs, though not a differential dose delivery to the tumor. By contrast, we found out that increased albumin-binding affinities of the injected ligands can lead to such a differential effect in delivering more doses to tumors, and this can be attributed to several factors that PBRPK modeling allows us to probe. CONCLUSIONS Advanced computational PBRPK modeling enables simulation and analysis of a variety of intervention and drug design scenarios, towards more optimal delivery of RPTs.
Collapse
Affiliation(s)
- Ali Fele-Paranj
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Babak Saboury
- Department of Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland, US
| | - Carlos Uribe
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
- Department of Functional Imaging, BC Cancer, Vancouver, BC, Canada
- Department of Radiology, University of British Columbia, Vancouver, BC, Canada
| | - Arman Rahmim
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada.
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada.
- Department of Radiology, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
7
|
Musket A, Davern S, Elam BM, Musich PR, Moorman JP, Jiang Y. The application of radionuclide therapy for breast cancer. FRONTIERS IN NUCLEAR MEDICINE (LAUSANNE, SWITZERLAND) 2024; 3:1323514. [PMID: 39355029 PMCID: PMC11440853 DOI: 10.3389/fnume.2023.1323514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 12/27/2023] [Indexed: 10/03/2024]
Abstract
Radionuclide-mediated diagnosis and therapy have emerged as effective and low-risk approaches to treating breast cancer. Compared to traditional anatomic imaging techniques, diagnostic radionuclide-based molecular imaging systems exhibit much greater sensitivity and ability to precisely illustrate the biodistribution and metabolic processes from a functional perspective in breast cancer; this transitions diagnosis from an invasive visualization to a noninvasive visualization, potentially ensuring earlier diagnosis and on-time treatment. Radionuclide therapy is a newly developed modality for the treatment of breast cancer in which radionuclides are delivered to tumors and/or tumor-associated targets either directly or using delivery vehicles. Radionuclide therapy has been proven to be eminently effective and to exhibit low toxicity when eliminating both primary tumors and metastases and even undetected tumors. In addition, the specific interaction between the surface modules of the delivery vehicles and the targets on the surface of tumor cells enables radionuclide targeting therapy, and this represents an exceptional potential for this treatment in breast cancer. This article reviews the development of radionuclide molecular imaging techniques that are currently employed for early breast cancer diagnosis and both the progress and challenges of radionuclide therapy employed in breast cancer treatment.
Collapse
Affiliation(s)
- Anna Musket
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Sandra Davern
- Oak Ridge National Laboratory, Oak Ridge, TN, United States
| | - Brianna M Elam
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Philip R Musich
- Department of Biomedical Science, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Jonathan P Moorman
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Yong Jiang
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| |
Collapse
|
8
|
Vernekar S, Budha RR, Alavala RR. Radiopharmaceuticals: A New Vista for Diagnosis and Treatment of Thyroid Cancer. Curr Radiopharm 2024; 17:148-162. [PMID: 38213166 DOI: 10.2174/0118744710277275231112081003] [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: 08/14/2023] [Revised: 09/26/2023] [Accepted: 10/02/2023] [Indexed: 01/13/2024]
Abstract
Radiopharmaceuticals are in the diagnosis and treatment of cancerous and noncancerous diseases, and a hope for optimistic effort in the field of nuclear medicine. They play a crucial role in clinical nuclear medicine by providing a tool to comprehend human disease and create efficient treatments. A detailed analysis is provided regarding the crux of molecular imaging including PET and SPECT overview for the detection of cancers. For a specified understanding of radiation therapy, topics include ranging from the selection of radionuclide to its development and manufacture, and dosage requirements to establishing the importance of I- 131 Radiotherapy in thyroid cancer. In this review, we also discussed the current state of the art of nuclear medicine in thyroid cancer, including the role of radioiodine (RAI) therapeutic scans in the diagnosis of differentiated thyroid cancer. In addition, we established a brief outlook into the current status of the research in thyroid cancer and discussed the future directions in this field.
Collapse
Affiliation(s)
- Siddhi Vernekar
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai, 400056, India
| | - Roja Rani Budha
- Amity Institute of Pharmacy, Amity University, Panvel, Mumbai, Maharashtra, 410206, India
| | - Rajasekhar Reddy Alavala
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai, 400056, India
| |
Collapse
|
9
|
Liatsou I, Josefsson A, Yu J, Li Z, Davis K, Brayton C, Wang H, Hobbs RF, Sgouros G. Early Normal Tissue Effects and Bone Marrow Relative Biological Effectiveness for an Actinium 225-Labeled HER2/neu-Targeting Antibody. Int J Radiat Oncol Biol Phys 2023; 117:1028-1037. [PMID: 37331568 DOI: 10.1016/j.ijrobp.2023.06.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 05/16/2023] [Accepted: 06/11/2023] [Indexed: 06/20/2023]
Abstract
PURPOSE In this study we determined the dose-independent relative biological effectiveness (RBE2) of bone marrow for an anti-HER2/neu antibody labeled with the alpha-particle emitter actinium 225 (225Ac). Hematologic toxicity is often a consequence of radiopharmaceutical therapy (RPT) administration, and dosimetric guidance to the bone marrow is required to limit toxicity. METHODS AND MATERIALS Female neu/N transgenic mice (MMTV-neu) were intravenously injected with 0 to 16.65 kBq of the alpha-particle emitter labeled antibody, 225Ac-DOTA-7.16.4, and euthanized at 1 to 9 days after treatment. Complete blood counts were performed. Femurs and tibias were collected, and bone marrow was isolated from 1 femur and tibia and counted for radioactivity. Contralateral intact femurs were fixed, decalcified, and assessed by histology. Marrow cellularity was the biologic endpoint selected for RBE2 determination. For the reference radiation, both femurs of the mice were photon irradiated with 0 to 5 Gy using a small animal radiation research platform. RESULTS Response as measured by cellularity for the alpha-particle emitter RPT (αRPT) RPT and the external beam radiation therapy were linear and linear quadratic, respectively, as a function of absorbed dose. The resulting dose-independent RBE2 for bone marrow was 6. CONCLUSIONS As αRPT gains prominence, preclinical studies evaluating RBE in vivo will be important in relating to human experience with beta-particle emitter RPT. Such normal tissue RBE evaluations will help mitigate unexpected toxicity in αRPT.
Collapse
Affiliation(s)
- Ioanna Liatsou
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Anders Josefsson
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jing Yu
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Zhi Li
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Kaori Davis
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Cory Brayton
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Hao Wang
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Robert F Hobbs
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - George Sgouros
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
10
|
Benfante V, Stefano A, Ali M, Laudicella R, Arancio W, Cucchiara A, Caruso F, Cammarata FP, Coronnello C, Russo G, Miele M, Vieni A, Tuttolomondo A, Yezzi A, Comelli A. An Overview of In Vitro Assays of 64Cu-, 68Ga-, 125I-, and 99mTc-Labelled Radiopharmaceuticals Using Radiometric Counters in the Era of Radiotheranostics. Diagnostics (Basel) 2023; 13:diagnostics13071210. [PMID: 37046428 PMCID: PMC10093267 DOI: 10.3390/diagnostics13071210] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 03/11/2023] [Accepted: 03/17/2023] [Indexed: 04/14/2023] Open
Abstract
Radionuclides are unstable isotopes that mainly emit alpha (α), beta (β) or gamma (γ) radiation through radiation decay. Therefore, they are used in the biomedical field to label biomolecules or drugs for diagnostic imaging applications, such as positron emission tomography (PET) and/or single-photon emission computed tomography (SPECT). A growing field of research is the development of new radiopharmaceuticals for use in cancer treatments. Preclinical studies are the gold standard for translational research. Specifically, in vitro radiopharmaceutical studies are based on the use of radiopharmaceuticals directly on cells. To date, radiometric β- and γ-counters are the only tools able to assess a preclinical in vitro assay with the aim of estimating uptake, retention, and release parameters, including time- and dose-dependent cytotoxicity and kinetic parameters. This review has been designed for researchers, such as biologists and biotechnologists, who would like to approach the radiobiology field and conduct in vitro assays for cellular radioactivity evaluations using radiometric counters. To demonstrate the importance of in vitro radiopharmaceutical assays using radiometric counters with a view to radiogenomics, many studies based on 64Cu-, 68Ga-, 125I-, and 99mTc-labeled radiopharmaceuticals have been revised and summarized in this manuscript.
Collapse
Affiliation(s)
- Viviana Benfante
- Ri.MED Foundation, Via Bandiera 11, 90133 Palermo, Italy
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Molecular and Clinical Medicine, University of Palermo, 90127 Palermo, Italy
- Institute of Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), 90015 Cefalù, Italy
| | - Alessandro Stefano
- Institute of Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), 90015 Cefalù, Italy
| | - Muhammad Ali
- Ri.MED Foundation, Via Bandiera 11, 90133 Palermo, Italy
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Molecular and Clinical Medicine, University of Palermo, 90127 Palermo, Italy
| | | | - Walter Arancio
- Ri.MED Foundation, Via Bandiera 11, 90133 Palermo, Italy
| | - Antonino Cucchiara
- Department of Diagnostic and Therapeutic Services, IRCCS-ISMETT (Mediterranean Institute for Transplantation and Advanced Specialized Therapies), Via Tricomi 5, 90127 Palermo, Italy
| | - Fabio Caruso
- Department of Diagnostic and Therapeutic Services, IRCCS-ISMETT (Mediterranean Institute for Transplantation and Advanced Specialized Therapies), Via Tricomi 5, 90127 Palermo, Italy
| | - Francesco Paolo Cammarata
- Institute of Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), 90015 Cefalù, Italy
| | - Claudia Coronnello
- Ri.MED Foundation, Via Bandiera 11, 90133 Palermo, Italy
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
| | - Giorgio Russo
- Institute of Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), 90015 Cefalù, Italy
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
| | - Monica Miele
- Ri.MED Foundation, Via Bandiera 11, 90133 Palermo, Italy
| | - Alessandra Vieni
- Department of Diagnostic and Therapeutic Services, IRCCS-ISMETT (Mediterranean Institute for Transplantation and Advanced Specialized Therapies), Via Tricomi 5, 90127 Palermo, Italy
| | - Antonino Tuttolomondo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Molecular and Clinical Medicine, University of Palermo, 90127 Palermo, Italy
| | - Anthony Yezzi
- Department of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Albert Comelli
- Ri.MED Foundation, Via Bandiera 11, 90133 Palermo, Italy
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
| |
Collapse
|
11
|
Khabirova S, Aleshin G, Anokhin E, Shchukina A, Zubenko A, Fedorova O, Averin A, Trusov L, Kalmykov S. Novel candidate theranostic radiopharmaceutical based on strontium hexaferrite nanoparticles conjugated with azacrown ligand. Dalton Trans 2023; 52:1731-1741. [PMID: 36655497 DOI: 10.1039/d2dt03548k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
In this article, we report to the best of our knowledge the first modification of NPs with ligands for combined radiopharmaceuticals. Nanoparticles with suitable magnetic properties can be used both for diagnostics as a contrast for MRI and for therapy, including the insufficiently studied magneto-mechanical therapy. Strontium hexaferrite is one of the few hard-magnetic materials for which stable biocompatible colloidal solutions can be obtained. Strontium hexaferrite nanoparticles coated with silicon dioxide (SHF@SiO2) were modified with an amino silane coupling agent (3-aminopropyl)triethoxysilane and azacrown ether derivatives with six heteroatoms in rings were covalently linked to the amine group through the carboxyl group. The hard magnetic nanoparticles were then radiolabeled with 207Bi with a labelling yield of up to 99.8%. In vitro experiments showed that the complex SHF@SiO2-APTES-L2-207Bi is stable enough to be a potential theranostic radiopharmaceutical.
Collapse
Affiliation(s)
- Sofia Khabirova
- Lomonosov Moscow State University, 119991 Leninskie Gory, 1/3, Moscow, Russian Federation.
| | - Gleb Aleshin
- Lomonosov Moscow State University, 119991 Leninskie Gory, 1/3, Moscow, Russian Federation.
| | - Evgeny Anokhin
- Lomonosov Moscow State University, 119991 Leninskie Gory, 1/3, Moscow, Russian Federation.
| | - Anna Shchukina
- A. N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences, 119991 Vavilova, 28, GSP-1, Moscow, Russian Federation
| | - Anastasia Zubenko
- A. N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences, 119991 Vavilova, 28, GSP-1, Moscow, Russian Federation
| | - Olga Fedorova
- A. N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences, 119991 Vavilova, 28, GSP-1, Moscow, Russian Federation
| | - Aleksey Averin
- Frumkin Institute of Physical chemistry and Electrochemistry Russian Academy of Sciences, Leninskiy ave. 31b4, Moscow, Russia
| | - Lev Trusov
- Lomonosov Moscow State University, 119991 Leninskie Gory, 1/3, Moscow, Russian Federation.
| | - Stepan Kalmykov
- Lomonosov Moscow State University, 119991 Leninskie Gory, 1/3, Moscow, Russian Federation.
| |
Collapse
|
12
|
Chang MC, Chiang PF, Kuo YJ, Peng CL, Chen IC, Huang CY, Chen CA, Chiang YC. Develop companion radiopharmaceutical YKL40 antibodies as potential theranostic agents for epithelial ovarian cancer. Biomed Pharmacother 2022; 155:113668. [PMID: 36088858 DOI: 10.1016/j.biopha.2022.113668] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 08/31/2022] [Accepted: 09/05/2022] [Indexed: 11/29/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is usually diagnosed at an advanced stage and has poor prognosis. Theranostic agents are the current trend in drug development, but are lacking in EOC. YKL40 is predominantly expressed and involved in tumorigenesis in EOC. In this study, we developed a companion theranostic agent targeting YKL40. We measured YKL40 expression levels in ascites using ELISA and correlated them with the clinical outcomes of patients with EOC. We developed radionuclide labeled In-111/Lu-177-DTPA-YKL40 neutralizing antibodies and investigated their radiochemical purity, SPECT/CT imaging, bio-distribution, and therapeutic responses in ovarian cancer xenograft mice. We demonstrated that YKL40 expression levels in ascites were significantly higher in EOC patients with serous histological type, high tumor grade, advanced stage, tumor recurrence, chemoresistance, and tumor-related death. The radiochemical purity of In-111/Lu-177-DTPA-YKL40 neutralizing antibodies reached more than 90% after 24 h of labeling. SPECT/CT imaging showed significant accumulation of In-111-DTPA-YKL40 and Lu-177-DTPA-YKL40 antibodies at the tumor site of ovarian cancer xenograft mice 24 h after administration. Lu-177-DTPA-YKL40 antibodies significantly inhibited tumor growth in ovarian cancer xenograft mice. Our study indicated that In-111/Lu-177-DTPA-YKL40 neutralizing antibodies could be potential companion theranostic agents for patients with EOC.
Collapse
Affiliation(s)
- Ming-Cheng Chang
- Isotope Application Division, Institute of Nuclear Energy Research, P.O. Box 3-27, Longtan, Taoyuan 325, Taiwan
| | - Ping-Fang Chiang
- Isotope Application Division, Institute of Nuclear Energy Research, P.O. Box 3-27, Longtan, Taoyuan 325, Taiwan
| | - Yu-Jen Kuo
- Isotope Application Division, Institute of Nuclear Energy Research, P.O. Box 3-27, Longtan, Taoyuan 325, Taiwan
| | - Cheng-Liang Peng
- Isotope Application Division, Institute of Nuclear Energy Research, P.O. Box 3-27, Longtan, Taoyuan 325, Taiwan
| | - I-Chun Chen
- Department of Medical Oncology, National Taiwan University Cancer Center, Taipei, Taiwan; Graduate Institute of Oncology, National Taiwan University, Taipei, Taiwan; Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - Chia-Yen Huang
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan; Gynecologic Cancer Center, Department of Obstetrics and Gynecology, Cathay General Hospital, Taipei, Taiwan; School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Chi-An Chen
- Department of Obstetrics and Gynecology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ying-Cheng Chiang
- Department of Obstetrics and Gynecology, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
13
|
Pareri AU, Koijam AS, Kumar C. Breaking the Silence of Tumor Response: Future Prospects of Targeted Radionuclide Therapy. Anticancer Agents Med Chem 2021; 22:1845-1858. [PMID: 34477531 DOI: 10.2174/1871520621666210903152354] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 07/12/2021] [Accepted: 07/19/2021] [Indexed: 01/10/2023]
Abstract
Therapy-induced tumor resistance has always been a paramount hurdle in the clinical triumph of cancer therapy. Resistance acquired by tumor through interventions of chemotherapeutic drugs, ionizing radiation, and immunotherapy in the patientsis a severe drawback and major cause of recurrence of tumor and failure of therapeutic responses. To counter acquired resistance in tumor cells, several strategies are practiced such as chemotherapy regimens, immunotherapy, and immunoconjugates, but the outcome is very disappointing for the patients as well as clinicians. Radionuclide therapy using alpha or beta-emitting radionuclide as payload became state-of-the-art for cancer therapy. With the improvement in dosimetric studies, development of high-affinity target molecules, and design of several novel chelating agents which provide thermodynamically stable complexes in vivo, the scope of radionuclide therapy has increased by leaps and bounds. Additionally, radionuclide therapy along with the combination of chemotherapy is gaining importance in pre-clinics, which is quite encouraging. Thus, it opens an avenue for newer cancer therapy modalities where chemotherapy, radiation therapy, and immunotherapy are unable to break the silence of tumor response. This article describes, in brief, the causes of tumor resistance and discusses the potential of radionuclide therapy to enhance tumor response.
Collapse
Affiliation(s)
| | | | - Chandan Kumar
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre Mumbai-400085, India
| |
Collapse
|
14
|
Xuan S, de Barros AODS, Nunes RC, Ricci-Junior E, da Silva AX, Sahid M, Alencar LMR, Dos Santos CC, Morandi V, Alexis F, Iram SH, Santos-Oliveira R. Radioactive gold nanocluster (198-AuNCs) showed inhibitory effects on cancer cells lines. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2021; 48:1214-1221. [PMID: 32940067 DOI: 10.1080/21691401.2020.1821698] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cancer is a global epidemic disease responsible for over ten millions death worldwide. The early diagnosis and the precise treatment with reduced adverse reactions are the main goal worldwide. In this study, we produced, characterized and evaluated (in vitro) in three different cancer cell lines (protaste, breast and melanoma) a radioactive gold nanocluster (R-AuNC) (198Au25(Capt)18). The pharmacokinetics as the influence in the ABC transporter (MRP1 Efflux Transporter Protein) was also evaluated. The results showed that R-AuNC (198Au25(Capt)18) are capable to kill the cancer cells lines of protaste, breast and melanoma. The pharmacokinetics showed a fast clearance and great volume of distribution, confirming the use of R-AuNC as nanomedicine for cancer treatment. Finally, the ABC transporter assay corroborated that the R-AuNC (198Au25(Capt)18) has no risk of being pumped out of cells by this efflux transporter. The results validate the use of gold nanoparticles as therapeutic nanomedicine for cancer treatment.
Collapse
Affiliation(s)
- Shijin Xuan
- Department of Mammary and Thyroid Surgery, Jinan Central Hospital Affiliated to Shandong University, Jinan, P. R. China
| | | | | | - Eduardo Ricci-Junior
- Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ademir Xavier da Silva
- Programa de Engenharia Nuclear - COPPE (Universidade Federal do Rio de Janeiro - Ilha do fundão, Rio de Janeiro, Brazil
| | - Muhammad Sahid
- Brazilian Nuclear Energy Commission, Nuclear Engineering Institute, Rio de Janeiro, Brazil.,Pakistan Nuclear Regulatory Authority, National Institutes of Safety and Security, Islamabad, Pakistan
| | | | | | - Veronica Morandi
- Department of Cell Biology, Laboratory of Biology of Endothelial Cells and Angiogenesis (LabAngio), IBRAG, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Frank Alexis
- Department of Bioengineering, Clemson University, Clemson, SC, USA.,School of Biological Sciences and Engineering, Yachay Tech, San Miguel de Urcuquí, Ecuador
| | - Surtaj H Iram
- Department of Chemistry & Biochemistry, College of Natural Sciences, South Dakota State University, Brookings, SD, USA
| | - Ralph Santos-Oliveira
- Brazilian Nuclear Energy Commission, Nuclear Engineering Institute, Rio de Janeiro, Brazil.,Laboratory of Radiopharmacy and Nanoradiopharmaceuticals, Zona Oeste State University, Rio de Janeiro, Brazil
| |
Collapse
|
15
|
Altıparmak Güleç B, Yurt F. Treatment with Radiopharmaceuticals and Radionuclides in Breast Cancer: Current Options. Eur J Breast Health 2021; 17:214-219. [PMID: 34263148 DOI: 10.4274/ejbh.galenos.2021.2021-3-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 03/20/2021] [Indexed: 12/22/2022]
Abstract
Radiopharmaceutical therapy (RPT) is an effective and safe treatment for many types of cancer. RPT acts by binding radioactive atoms to tumor-associated antigens, monoclonal antibodies, nanoparticles, peptides, and small molecules. These treatments ensure that a concentrated dose is delivered to the targeted tumor tissue while preserving the normal tissues surrounding the tumor. Given these features, RPT is superior to traditional methods. This review article aimed to performa comprehensive review and evaluation of the potential of radionuclides and radiopharmaceuticals used in breast cancer treatment in preclinical studies conducted in the last five years.
Collapse
Affiliation(s)
- Burcu Altıparmak Güleç
- Department of Nuclear Applications, Institute of Nuclear Science, Ege University, İzmir, Turkey (Graduated)
| | - Fatma Yurt
- Department of Nuclear Applications, Institute of Nuclear Science, Ege University, İzmir, Turkey
| |
Collapse
|
16
|
Atomic Nanogenerators in Targeted Alpha Therapies: Curie's Legacy in Modern Cancer Management. Pharmaceuticals (Basel) 2020; 13:ph13040076. [PMID: 32340103 PMCID: PMC7243103 DOI: 10.3390/ph13040076] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 04/17/2020] [Accepted: 04/21/2020] [Indexed: 12/12/2022] Open
Abstract
Atomic in vivo nanogenerators such as actinium-225, thorium-227, and radium-223 are of increasing interest and importance in the treatment of patients with metastatic cancer diseases. This is due to their peculiar physical, chemical, and biological characteristics, leading to astonishing responses in otherwise resistant patients. Nevertheless, there are still a few obstacles and hurdles to be overcome that hamper the broader utilization in the clinical setting. Next to the limited supply and relatively high costs, the in vivo complex stability and the fate of the recoiling daughter radionuclides are substantial problems that need to be solved. In radiobiology, the mechanisms underlying treatment efficiency, possible resistance mechanisms, and late side effect occurrence are still far from being understood and need to be unraveled. In this review, the current knowledge on the scientific and clinical background of targeted alpha therapies is summarized. Furthermore, open issues and novel approaches with a focus on the future perspective are discussed. Once these are unraveled, targeted alpha therapies with atomic in vivo nanogenerators can be tailored to suit the needs of each patient when applying careful risk stratification and combination therapies. They have the potential to become one of the major treatment pillars in modern cancer management.
Collapse
|
17
|
Hofmann W, Li WB, Friedland W, Miller BW, Madas B, Bardiès M, Balásházy I. Internal microdosimetry of alpha-emitting radionuclides. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2020; 59:29-62. [PMID: 31863162 PMCID: PMC7012986 DOI: 10.1007/s00411-019-00826-w] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 12/08/2019] [Indexed: 05/27/2023]
Abstract
At the tissue level, energy deposition in cells is determined by the microdistribution of alpha-emitting radionuclides in relation to sensitive target cells. Furthermore, the highly localized energy deposition of alpha particle tracks and the limited range of alpha particles in tissue produce a highly inhomogeneous energy deposition in traversed cell nuclei. Thus, energy deposition in cell nuclei in a given tissue is characterized by the probability of alpha particle hits and, in the case of a hit, by the energy deposited there. In classical microdosimetry, the randomness of energy deposition in cellular sites is described by a stochastic quantity, the specific energy, which approximates the macroscopic dose for a sufficiently large number of energy deposition events. Typical examples of the alpha-emitting radionuclides in internal microdosimetry are radon progeny and plutonium in the lungs, plutonium and americium in bones, and radium in targeted radionuclide therapy. Several microdosimetric approaches have been proposed to relate specific energy distributions to radiobiological effects, such as hit-related concepts, LET and track length-based models, effect-specific interpretations of specific energy distributions, such as the dual radiation action theory or the hit-size effectiveness function, and finally track structure models. Since microdosimetry characterizes only the initial step of energy deposition, microdosimetric concepts are most successful in exposure situations where biological effects are dominated by energy deposition, but not by subsequently operating biological mechanisms. Indeed, the simulation of the combined action of physical and biological factors may eventually require the application of track structure models at the nanometer scale.
Collapse
Affiliation(s)
- Werner Hofmann
- Biological Physics, Department of Chemistry and Physics of Materials, University of Salzburg, Hellbrunner Str. 34, 5020, Salzburg, Austria.
| | - Wei Bo Li
- Institute of Radiation Medicine, Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany.
| | - Werner Friedland
- Institute of Radiation Medicine, Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Brian W Miller
- Department of Radiation Oncology, School of Medicine, University of Colorado, Aurora, CO, 80045, USA
- College of Optical Sciences, University of Arizona, Tucson, AZ, 85721, USA
| | - Balázs Madas
- Environmental Physics Department, MTA Centre for Energy Research, Budapest, Hungary
| | - Manuel Bardiès
- Centre de Recherches en Cancérologie de Toulouse, UMR 1037, INSERM Université Paul Sabatier, Toulouse, France
| | - Imre Balásházy
- Environmental Physics Department, MTA Centre for Energy Research, Budapest, Hungary
| |
Collapse
|