1
|
Steiner YTS, Zizi V, Mangoni M, Nardini P, Bani D, Bencini A, Bianchi A, Savastano M, Romano GM. Fluorescent Deferoxamine Complexes of Cu(II) and Zr(IV): Insights in the Development of Dual Imaging Probes. Chemistry 2025:e202501203. [PMID: 40342180 DOI: 10.1002/chem.202501203] [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: 03/26/2025] [Revised: 05/07/2025] [Accepted: 05/08/2025] [Indexed: 05/11/2025]
Abstract
Deferoxamine (DFO) is widely regarded as benchmark chelator for 89Zr(IV), a commonly used PET (positron emission tomography) tracer. We have introduced a novel fluorescent coumarin derivative of DFO (DFOKC), characterized by chelating unit and fluorophore covalently linked via a lysine molecule. This design introduces a free primary amine group, which, in perspective, can be functionalized with biological vectors, potentially improving tumor tissue selectivity. Its acid-base and metal coordination properties toward Cu(II) and Zr(IV) ions were thoroughly characterized using UV-Vis and fluorescence emission spectroscopy. DFOKC strongly coordinates both metal ions, forming somewhat more stable complexes than DFO, while retaining fluorescence emission, thus enabling dual-mode optical and PET imaging. Biodistribution assays conducted on NIH-3T3 fibroblasts, and MDA-MB-231 mammary adenocarcinoma cell lines demonstrated that the presence of primary amine groups favors Zr-DFOKC complex cell internalization via pinocytosis compared to the parent molecule DFOC, in which the fluorophore is linked to the amine group of DFO. Furthermore, crystal violet and MTT assays revealed no cytotoxic effects or mitochondrial impairment, even at concentrations higher than those typically used for radio-diagnostic applications. These results strongly support the potential of DFOKC as a versatile and promising tool for dual imaging, offering significant advantages in molecular imaging.
Collapse
Affiliation(s)
| | - Virginia Zizi
- Imaging Platform, Department of Experimental & Clinical Medicine, University of Florence, Viale G.Pieraccini 6, 50139, Florence, Italy
| | - Monica Mangoni
- Radiotherapy Unit, Department of Experimental & Clinical Biomedical Sciences, University of Florence, Largo Brambilla 3, 50134, Florence, Italy
| | - Patrizia Nardini
- Imaging Platform, Department of Experimental & Clinical Medicine, University of Florence, Viale G.Pieraccini 6, 50139, Florence, Italy
| | - Daniele Bani
- Imaging Platform, Department of Experimental & Clinical Medicine, University of Florence, Viale G.Pieraccini 6, 50139, Florence, Italy
| | - Andrea Bencini
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3, 50019, Sesto Fiorentino, Italy
| | - Antonio Bianchi
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3, 50019, Sesto Fiorentino, Italy
| | - Matteo Savastano
- Department for the Promotion of Human Science and Quality of Life, University San Raffaele Roma, Via di Val Cannuta 247, 00166, Rome, Italy
| | - Giammarco Maria Romano
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3, 50019, Sesto Fiorentino, Italy
| |
Collapse
|
2
|
Yusufaly T, Roncali E, Brosch-Lenz J, Uribe C, Jha AK, Currie G, Dutta J, El-Fakhri G, McMeekin H, Pandit-Taskar N, Schwartz J, Shi K, Strigari L, Zaidi H, Saboury B, Rahmim A. Computational Nuclear Oncology Toward Precision Radiopharmaceutical Therapies: Current Tools, Techniques, and Uncharted Territories. J Nucl Med 2025; 66:509-515. [PMID: 39947910 PMCID: PMC11960611 DOI: 10.2967/jnumed.124.267927] [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: 04/22/2024] [Accepted: 01/27/2025] [Indexed: 04/03/2025] Open
Abstract
Radiopharmaceutical therapy (RPT), with its targeted delivery of cytotoxic ionizing radiation, demonstrates significant potential for treating a wide spectrum of malignancies, with particularly unique benefits for metastatic disease. There is an opportunity to optimize RPTs and enhance the precision of theranostics by moving beyond a one-size-fits-all approach and using patient-specific image-based dosimetry for personalized treatment planning. Such an approach, however, requires accurate methods and tools for the mathematic modeling and prediction of dose and clinical outcome. To this end, the SNMMI AI-Dosimetry Working Group is promoting the paradigm of computational nuclear oncology: mathematic models and computational tools describing the hierarchy of etiologic mechanisms involved in RPT dose response. This includes radiopharmacokinetics for image-based internal dosimetry and radiobiology for the mapping of dose response to clinical endpoints. The former area originates in pharmacotherapy, whereas the latter originates in radiotherapy. Accordingly, models and methods developed in these predecessor disciplines serve as a foundation on which to develop a repurposed set of tools more appropriate to RPT. Over the long term, this computational nuclear oncology framework also promises to facilitate widespread cross-fertilization of ideas between nuclear medicine and the greater mathematic and computational oncology communities.
Collapse
Affiliation(s)
- Tahir Yusufaly
- Division of Radiology and Radiological Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland;
| | - Emilie Roncali
- Department of Biomedical Engineering, University of California Davis, Davis, California
| | | | - Carlos Uribe
- Department of Radiology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Abhinav K Jha
- Department of Biomedical Engineering and Mallinckrodt Institute of Radiology, Washington University, St. Louis, Missouri
| | - Geoffrey Currie
- School of Dentistry and Medical Sciences, Charles Sturt University, Wagga Wagga, New South Wales, Australia
| | - Joyita Dutta
- Department of Biomedical Engineering, University of Massachusetts, Amherst, Massachusetts
| | - Georges El-Fakhri
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut
| | | | - Neeta Pandit-Taskar
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiology, Weill Cornell Medical College, New York, New York
| | - Jazmin Schwartz
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kuangyu Shi
- Department of Nuclear Medicine, University of Bern, Bern, Switzerland
| | - Lidia Strigari
- Department of Medical Physics, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Habib Zaidi
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, Geneva, Switzerland
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | | | - Arman Rahmim
- Department of Radiology, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Physics, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
3
|
Ailawadhi S, Pafundi D, Peterson J. Advances and future directions in radiopharmaceutical delivery for cancer treatment. Expert Rev Anticancer Ther 2025; 25:351-361. [PMID: 40019019 DOI: 10.1080/14737140.2025.2472859] [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: 09/09/2024] [Revised: 01/29/2025] [Accepted: 02/24/2025] [Indexed: 03/01/2025]
Abstract
INTRODUCTION Targeted radiopharmaceutical therapies (RPTs) have emerged as a promising approach for the precise treatment of various cancers. Delivering ionizing radiation directly to cancer cells while sparing surrounding healthy tissue, radiopharmaceuticals offer enhanced efficacy and reduced toxicity compared to conventional external beam radiation therapy (i.e. photons and electrons). AREAS COVERED In the current era of personalized cancer care, the appropriate choice of RPTs for a clinical condition and the specific patient's care needs to be better understood. Several available RPT agents with their respective clinical applicability along with rapidly ongoing research in this field have now given RPTs the ability to lend themselves to a personalized medicine focus. This review provides an overview of recent advancements in RPT, including nuclide selection and development, molecular targeting strategies, radiopharmaceutical development, and clinical applications. EXPERT OPINION We discuss the underlying principles, challenges, and opportunities for future development. Furthermore, we explore emerging technologies and future directions in the field, highlighting the potential impact on personalized cancer care.
Collapse
Affiliation(s)
| | - Deanna Pafundi
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL, USA
| | - Jennifer Peterson
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL, USA
| |
Collapse
|
4
|
Barata P, Oliveira A, Soares R, Fernandes A. Gut Microbiota Is Not Significantly Altered by Radioiodine Therapy. Nutrients 2025; 17:395. [PMID: 39940254 PMCID: PMC11819986 DOI: 10.3390/nu17030395] [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: 12/14/2024] [Revised: 01/13/2025] [Accepted: 01/17/2025] [Indexed: 02/14/2025] Open
Abstract
Purpose: Radiotherapy treatments are known to alter the gut microbiota. However, little is known regarding the effect of nuclear medicine treatments on gut microbiota, and it is established that nuclear medicine is inherently different from radiotherapy. To address this knowledge gap, we conducted a prospective study to identify changes in the gut microbiota of patients treated with [131I]NaI by comparing fecal samples before and after RAIT. Methods: Fecal samples of 64 patients (37 with thyroid cancer and 27 with hyperthyroidism) with indication for RAIT were collected 2 to 3 days before treatment and 8 to 10 days post-treatment. After DNA extraction, the gut microbiota's richness, diversity, and composition were analyzed by shotgun metagenomics. In addition, LEfSe was performed to compare compositional changes in specific bacteria. Results: Gut microbiome richness and diversity remained unchanged after RAIT, with few changes in its composition identified, especially in patients with hyperthyroidism. Conclusions: This study provides a conceptual and analytical basis for increasing our understanding of the effects of radiopharmaceuticals on gut microbiota. Our preliminary results indicate that RAIT, contrary to radiotherapy, does not cause major disruptions to the human gut microbiota.
Collapse
Affiliation(s)
- Pedro Barata
- RISE-Health, Faculdade de Ciências da Saúde, Universidade Fernando Pessoa, Fundação Ensino e Cultura Fernando Pessoa, Rua Carlos da Maia 296, 4200-150 Porto, Portugal
- Centro Hospitalar de Santo António, Unidade Local de Saúde de Santo António, Largo do Professor Abel Salazar, 4099-001 Porto, Portugal
| | - Ana Oliveira
- Department of Nuclear Medicine, Centro Hospitalar e Universitário de São João, E.P.E., 4200-319 Porto, Portugal
| | - Raquel Soares
- Department of Biomedicine, Faculdade de Medicina da Universidade do Porto, 4200-319 Porto, Portugal
| | - Ana Fernandes
- RISE-Health, Faculdade de Ciências da Saúde, Universidade Fernando Pessoa, Fundação Ensino e Cultura Fernando Pessoa, Rua Carlos da Maia 296, 4200-150 Porto, Portugal
- Department of Nuclear Medicine, Centro Hospitalar e Universitário de São João, E.P.E., 4200-319 Porto, Portugal
| |
Collapse
|
5
|
Serafini D, Zancopè N, Pavone AM, Benfante V, Arzenton A, Russo V, Ballan M, Morselli L, Cammarata FP, Comelli A, Russo G, Scopelliti F, Di Marco V, Mastrotto F, Asti M, Maniglio D, Sbarra C, Bortolussi S, Donzella A, Zenoni A, Gandini A, Villa V, Paderno D, Zangrando L, Corradetti S, Mariotti E, Salvini A, Torrisi F, Lunardon M, Andrighetto A. 111Ag phantom images with Cerenkov Luminescence Imaging and digital autoradiography within the ISOLPHARM project. Appl Radiat Isot 2025; 215:111562. [PMID: 39488936 DOI: 10.1016/j.apradiso.2024.111562] [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: 03/15/2024] [Revised: 10/08/2024] [Accepted: 10/21/2024] [Indexed: 11/05/2024]
Abstract
Targeted Radionuclide Therapy (TRT) is a medical technique exploiting radionuclides to combat cancer growth and spread. TRT requires a supply of radionuclides that are currently produced by either cyclotrons or nuclear research reactors. In this context, the ISOLPHARM project investigates the production of innovative radionuclides for medical applications. This production will be based on the forthcoming SPES facility at the Legnaro National Laboratories (LNL) of the National Institute for Nuclear Physics (INFN), an ISOL facility where high-purity radioactive beams will be used to produce carrier-free radiopharmaceuticals. Previous studies demonstrated that a significant amount of 111Ag, an innovative β/γ emitter suitable for TRT with theranostic applications, can be obtained at the SPES facility. The present work describes the first imaging study on phantoms with 111Ag performed by the ISOLPHARM collaboration. This is a fundamental step to pave the way for the upcoming in vivo studies on the 111Ag-based radiopharmaceutical currently being developed. The imaging potential of this radionuclide was investigated by acquiring phantom images with Cerenkov Luminescence Imaging (CLI) and digital autoradiography (ARG).
Collapse
Affiliation(s)
- Davide Serafini
- Legnaro National Laboratories, National Institute for Nuclear Physics, INFN-LNL, Viale dell'Università 2, 35020 Legnaro, Italy; Department of Physical Sciences, Earth and Environment, University of Siena, Via Roma 56, 53100 Siena, Italy.
| | - Nicola Zancopè
- Department of Physics and Astronomy, University of Padova, Via Marzolo 8, 35131 Padova, Italy.
| | - Anna Maria Pavone
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via Santa Sofia 97, 95123 Catania, Italy; Ri.MED Foundation, Via Bandiera 11, 90133 Palermo, Italy.
| | - Viviana Benfante
- Ri.MED Foundation, Via Bandiera 11, 90133 Palermo, Italy; Institute of Molecular Bioimaging and Physiology, National Research Council. IBFM-CNR, Contrada Pietrapollastra-Pisciotto, 90015 Cefalù, Italy.
| | - Alberto Arzenton
- Legnaro National Laboratories, National Institute for Nuclear Physics, INFN-LNL, Viale dell'Università 2, 35020 Legnaro, Italy; Department of Physical Sciences, Earth and Environment, University of Siena, Via Roma 56, 53100 Siena, Italy.
| | - Vincenzo Russo
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via Santa Sofia 97, 95123 Catania, Italy.
| | - Michele Ballan
- Legnaro National Laboratories, National Institute for Nuclear Physics, INFN-LNL, Viale dell'Università 2, 35020 Legnaro, Italy.
| | - Luca Morselli
- Legnaro National Laboratories, National Institute for Nuclear Physics, INFN-LNL, Viale dell'Università 2, 35020 Legnaro, Italy; Department of Physics and Earth Science, University of Ferrara, Via G. Saragat 1, 44121 Ferrara, Italy.
| | - Francesco Paolo Cammarata
- Institute of Molecular Bioimaging and Physiology, National Research Council. IBFM-CNR, Contrada Pietrapollastra-Pisciotto, 90015 Cefalù, Italy; Laboratori Nazionali del Sud, National Institute for Nuclear Physics, INFN-LNS, Via Santa Sofia 62, 95123 Catania, Italy.
| | - Albert Comelli
- Ri.MED Foundation, Via Bandiera 11, 90133 Palermo, Italy.
| | - Giorgio Russo
- Institute of Molecular Bioimaging and Physiology, National Research Council. IBFM-CNR, Contrada Pietrapollastra-Pisciotto, 90015 Cefalù, Italy; Laboratori Nazionali del Sud, National Institute for Nuclear Physics, INFN-LNS, Via Santa Sofia 62, 95123 Catania, Italy.
| | - Fabrizio Scopelliti
- Nuclear Medicine Department of Cannizzaro Hospital, Via Messina 829, 95126 Catania, Italy.
| | - Valerio Di Marco
- Department of Chemical Sciences, University of Padova, Via Marzolo 1, 35131 Padova, Italy.
| | - Francesca Mastrotto
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via Marzolo 5, 35131 Padova, Italy.
| | - Mattia Asti
- Radiopharmaceutical Chemistry Section, Nuclear Medicine Unit, AUSL-IRCCS di Reggio Emilia, Viale Risorgimento 80, 42122 Reggio Emilia, Italy.
| | - Devid Maniglio
- Department of Industrial Engineering, BIOtech Research Center, University of Trento, Via delle Regole 101, 38123 Mattarello, Italy.
| | - Carla Sbarra
- Bologna Division, National Institute for Nuclear Physics, Viale C. Berti Pichat 6/2, 40127 Bologna, Italy.
| | - Silva Bortolussi
- Department of Physics, University of Pavia, Via Bassi 6, 27100 Pavia, Italy.
| | - Antonietta Donzella
- Department of Mechanical and Industrial Engineering, University of Brescia, Via Branze 38, 25123 Brescia, Italy; Pavia Division, National Institute for Nuclear Physics, Via Bassi 6, 27100 Pavia, Italy.
| | - Aldo Zenoni
- Department of Mechanical and Industrial Engineering, University of Brescia, Via Branze 38, 25123 Brescia, Italy; Pavia Division, National Institute for Nuclear Physics, Via Bassi 6, 27100 Pavia, Italy.
| | - Andrea Gandini
- Applied Nuclear Energy Laboratory, LENA, Via G. Aselli 41, 27100 Pavia, Italy.
| | - Valerio Villa
- Department of Mechanical and Industrial Engineering, University of Brescia, Via Branze 38, 25123 Brescia, Italy; Pavia Division, National Institute for Nuclear Physics, Via Bassi 6, 27100 Pavia, Italy.
| | - Diego Paderno
- Department of Mechanical and Industrial Engineering, University of Brescia, Via Branze 38, 25123 Brescia, Italy; Pavia Division, National Institute for Nuclear Physics, Via Bassi 6, 27100 Pavia, Italy.
| | - Lisa Zangrando
- Padova Division, National Institute for Nuclear Physics, Via Marzolo 8, 35131 Padova, Italy.
| | - Stefano Corradetti
- Legnaro National Laboratories, National Institute for Nuclear Physics, INFN-LNL, Viale dell'Università 2, 35020 Legnaro, Italy.
| | - Emilio Mariotti
- Department of Physical Sciences, Earth and Environment, University of Siena, Via Roma 56, 53100 Siena, Italy; Pisa Division, National Institute for Nuclear Physics, Largo Bruno Pontecorvo 3, 56127 Pisa, Italy.
| | - Andrea Salvini
- Applied Nuclear Energy Laboratory, LENA, Via G. Aselli 41, 27100 Pavia, Italy.
| | - Filippo Torrisi
- Department of Medicine and Surgery, University of Enna "Kore", Via Marzolo 8, 35131 Enna, Italy.
| | - Marcello Lunardon
- Department of Physics and Astronomy, University of Padova, Via Marzolo 8, 35131 Padova, Italy; Padova Division, National Institute for Nuclear Physics, Via Marzolo 8, 35131 Padova, Italy.
| | - Alberto Andrighetto
- Legnaro National Laboratories, National Institute for Nuclear Physics, INFN-LNL, Viale dell'Università 2, 35020 Legnaro, Italy.
| |
Collapse
|
6
|
Bunjes D. Radioimmunotherapy of acute myeloid leukemia: a critical assessment of its prospects and limitations. Expert Rev Hematol 2025; 18:81-89. [PMID: 39754536 DOI: 10.1080/17474086.2025.2449863] [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: 08/27/2024] [Revised: 12/08/2024] [Accepted: 01/02/2025] [Indexed: 01/06/2025]
Abstract
INTRODUCTION Leukemic stem cells (LSC) are the source of relapse in acute myeloid leukemia (AML). Thus, eliminating LSC is one of the overarching goals of AML research. Radioimmunotherapy is an immunotherapeutic approach which utilizes radioactive isotopes as effector molecules based on the proven ability of ionizing radiation (IR) to kill LSC.It has the potential to eliminate target-antigen negative LSC. AREAS COVERED LSC biology, radiobiological principles of RIT, an overview of published and unpublished clinical results of RIT in AML. Issues of practical implementation of RIT in clinical trials. EXPERT OPINION RIT for AML isat a critical juncture. Its ability to target antigen negative LSC gives it an advantage compared with other forms of immunotherapy. In order to compete with other forms of targeted therapy the procedure has to be simplified.
Collapse
MESH Headings
- Humans
- Radioimmunotherapy/methods
- Radioimmunotherapy/adverse effects
- Leukemia, Myeloid, Acute/radiotherapy
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/immunology
- Neoplastic Stem Cells/radiation effects
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Neoplastic Stem Cells/immunology
- Animals
Collapse
Affiliation(s)
- Donald Bunjes
- Department of Internal Medicine III, Ulm University Hospital, Ulm, Germany
| |
Collapse
|
7
|
Tomiyoshi K, Wilson LJ, Mourtada F, Mourtada JS, Namiki Y, Kamata W, Yang DJ, Inoue T. Optimization Processes of Clinical Chelation-Based Radiopharmaceuticals for Pathway-Directed Targeted Radionuclide Therapy in Oncology. Pharmaceutics 2024; 16:1458. [PMID: 39598580 PMCID: PMC11597032 DOI: 10.3390/pharmaceutics16111458] [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: 10/23/2024] [Revised: 11/09/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024] Open
Abstract
Targeted radionuclide therapy (TRT) for internal pathway-directed treatment is a game changer for precision medicine. TRT improves tumor control while minimizing damage to healthy tissue and extends the survival for patients with cancer. The application of theranostic-paired TRT along with cellular phenotype and genotype correlative analysis has the potential for malignant disease management. Chelation chemistry is essential for the development of theranostic-paired radiopharmaceuticals for TRT. Among image-guided TRT, 68Ga and 99mTc are the current standards for diagnostic radionuclides, while 177Lu and 225Ac have shown great promise for β- and α-TRT, respectively. Their long half-lives, potent radiobiology, favorable decay schemes, and ability to form stable chelation conjugates make them ideal for both manufacturing and clinical use. The current challenges include optimizing radionuclide production processes, coordinating chelation chemistry stability of theranostic-paired isotopes to reduce free daughters [this pertains to 225Ac daughters 221Fr and 213Bi]-induced tissue toxicity, and improving the modeling of micro dosimetry to refine dose-response evaluation. The empirical approach to TRT delivery is based on standard radionuclide administered activity levels, although clinical trials have revealed inconsistent outcomes and normal-tissue toxicities despite equivalent administered activities. This review presents the latest optimization methods for chelation-based theranostic radiopharmaceuticals, advancements in micro-dosimetry, and SPECT/CT technologies for quantifying whole-body uptake and monitoring therapeutic response as well as cytogenetic correlative analyses.
Collapse
Affiliation(s)
- Katsumi Tomiyoshi
- Shonan Research Institute of Innovative Medicine, Shonan Kamakura General Hospital, Kamakura 247-8533, Japan
| | - Lydia J. Wilson
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA 19107, USA; (L.J.W.); (F.M.)
| | - Firas Mourtada
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA 19107, USA; (L.J.W.); (F.M.)
| | | | - Yuta Namiki
- Advanced Medical Center, Shonan Kamakura General Hospital, Kamakura 247-8533, Japan; (Y.N.); (W.K.); (D.J.Y.)
| | - Wataru Kamata
- Advanced Medical Center, Shonan Kamakura General Hospital, Kamakura 247-8533, Japan; (Y.N.); (W.K.); (D.J.Y.)
| | - David J. Yang
- Advanced Medical Center, Shonan Kamakura General Hospital, Kamakura 247-8533, Japan; (Y.N.); (W.K.); (D.J.Y.)
| | - Tomio Inoue
- Advanced Medical Center, Shonan Kamakura General Hospital, Kamakura 247-8533, Japan; (Y.N.); (W.K.); (D.J.Y.)
| |
Collapse
|
8
|
Abdollahi H, Fele-Paranj A, Rahmim A. Model-Informed Radiopharmaceutical Therapy Optimization: A Study on the Impact of PBPK Model Parameters on Physical, Biological, and Statistical Measures in 177Lu-PSMA Therapy. Cancers (Basel) 2024; 16:3120. [PMID: 39335092 PMCID: PMC11430653 DOI: 10.3390/cancers16183120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 09/04/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
Purpose: To investigate the impact of physiologically based pharmacokinetic (PBPK) parameters on physical, biological, and statistical measures in lutetium-177-labeled radiopharmaceutical therapies (RPTs) targeting the prostate-specific membrane antigen (PSMA). Methods: Using a clinically validated PBPK model, realistic time-activity curves (TACs) for tumors, salivary glands, and kidneys were generated based on various model parameters. These TACs were used to calculate the area-under-the-TAC (AUC), dose, biologically effective dose (BED), and figure-of-merit BED (fBED). The effects of these parameters on radiobiological, pharmacokinetic, time, and statistical features were assessed. Results: Manipulating PBPK parameters significantly influenced AUC, dose, BED, and fBED outcomes across four different BED models. Higher association rates increased AUC, dose, and BED values for tumors, with minimal impact on non-target organs. Increased internalization rates reduced AUC and dose for tumors and kidneys. Higher serum protein-binding rates decreased AUC and dose for all tissues. Elevated tumor receptor density and ligand amounts enhanced uptake and effectiveness in tumors. Larger tumor volumes required dosimetry adjustments to maintain efficacy. Setting the tumor release rate to zero intensified the impact of association and internalization rates, enhancing tumor targeting while minimizing the effects on salivary glands and kidneys. Conclusions: Optimizing PBPK parameters can enhance the efficacy of lutetium-177-labeled RPTs targeting PSMA, providing insights for personalized and effective treatment regimens to minimize toxicity and improve therapeutic outcomes.
Collapse
Affiliation(s)
- Hamid Abdollahi
- Department of Radiology, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada;
| | - Ali Fele-Paranj
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada;
- Department of Mathematics, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Arman Rahmim
- Department of Radiology, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada;
- Department of Physics & Astronomy, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| |
Collapse
|
9
|
Lu G, Gao D, Liu Y, Yu X, Jiang W, Lv Z. Early and long-term responses of intestinal microbiota and metabolites to 131I treatment in differentiated thyroid cancer patients. BMC Med 2024; 22:300. [PMID: 39020393 PMCID: PMC11256643 DOI: 10.1186/s12916-024-03528-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 07/10/2024] [Indexed: 07/19/2024] Open
Abstract
BACKGROUND Multiple high doses of 131I therapy in patients with differentiated thyroid cancer (DTC) might disrupt the balance of gut microbiota and metabolites. This study aimed to investigate the alterations of intestinal bacteria and metabolism over two courses of 131I therapy, explore the interactions, and construct diagnostic models reflecting enteric microecology based on 131I therapy. METHODS A total of 81 patients were recruited for the first 131I therapy (131I-1st), among whom 16 received a second course (131I-2nd) after half a year. Fecal samples were collected 1 day before (Pre-131I-1st/2nd) and 3 days after (Post-131I-1st/2nd) 131I therapy for microbiome (16S rRNA gene sequencing) and metabolomic (LC-MS/MS) analyses. RESULTS A total of six microbial genera and 11 fecal metabolites enriched in three pathways were identified to show significant differences between Pre-131I-1st and other groups throughout the two courses of 131I treatment. In the Post-131I-1st group, the beneficial bacteria Bifidobacterium, Lachnoclostridium, uncultured_bacterium_f_Lachnospiraceae, and Lachnospiraceae_UCG004 were abundant and the radiation-sensitive pathways of linoleic acid (LA), arachidonic acid, and tryptophan metabolism were inhibited compared with the Pre-131I-1st group. Compared with the Pre-131I-1st group, the Pre-131I-2nd group exhibited a reduced diversity of flora and differentially expressed metabolites, with a low abundance of beneficial bacteria and dysregulated radiation-sensitive pathways. However, less significant differences in microbiota and metabolites were found between the Pre/Post-131I-2nd groups compared with those between the Pre/Post-131I-1st groups. A complex co-occurrence was observed between 6 genera and 11 metabolites, with Lachnoclostridium, Lachnospiraceae_UCG004, Escherichia-Shigella, and LA-related metabolites contributing the most. Furthermore, combined diagnostic models of charactered bacteria and metabolites answered well in the early, long-term, and dose-dependent responses for 131I therapy. CONCLUSIONS Different stages of 131I therapy exert various effects on gut microecology, which play an essential role in regulating radiotoxicity and predicting the therapeutic response.
Collapse
Affiliation(s)
- Ganghua Lu
- Clinical Nuclear Medicine Center, Imaging Clinical Medical Center, Institute of Nuclear Medicine, Institute of Clinical Mass Spectrometry Applied Research Center, Department of Nuclear Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Dingwei Gao
- Clinical Nuclear Medicine Center, Imaging Clinical Medical Center, Institute of Nuclear Medicine, Institute of Clinical Mass Spectrometry Applied Research Center, Department of Nuclear Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Yixian Liu
- Department of Gynecology and Obstetrics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Xiaqing Yu
- Clinical Nuclear Medicine Center, Imaging Clinical Medical Center, Institute of Nuclear Medicine, Institute of Clinical Mass Spectrometry Applied Research Center, Department of Nuclear Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China.
| | - Wen Jiang
- Clinical Nuclear Medicine Center, Imaging Clinical Medical Center, Institute of Nuclear Medicine, Institute of Clinical Mass Spectrometry Applied Research Center, Department of Nuclear Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China.
- Department of Nuclear Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
| | - Zhongwei Lv
- Clinical Nuclear Medicine Center, Imaging Clinical Medical Center, Institute of Nuclear Medicine, Institute of Clinical Mass Spectrometry Applied Research Center, Department of Nuclear Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China.
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, 200003, China.
| |
Collapse
|
10
|
Kerr CP, Sheehan-Klenk J, Grudzinski JJ, Adam DP, Nguyen TPT, Ferreira CA, Bates AM, Jin WJ, Kwon O, Olson AP, Lin W, Hyun M, Jagodinsky JC, Powers M, Sriramaneni RN, Clark PA, Shea AG, Rojas HC, Choi C, Massey CF, Zangl LM, Pinchuk AN, Aluicio-Sarduy E, Kim K, Engle JW, Hernandez R, Bednarz BP, Weichert JP, Morris ZS. Effects of clinically relevant radionuclides on the activation of a type I interferon response by radiopharmaceuticals in syngeneic murine tumor models. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.10.602990. [PMID: 39071353 PMCID: PMC11275738 DOI: 10.1101/2024.07.10.602990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Radiopharmaceutical therapies (RPT) activate a type I interferon (IFN1) response in tumor cells. We hypothesized that the timing and amplitude of this response varies by isotope. We compared equal doses delivered by 90 Y, 177 Lu, and 225 Ac in vitro as unbound radionuclides and in vivo when chelated to NM600, a tumor-selective alkylphosphocholine. Response in murine MOC2 head and neck carcinoma and B78 melanoma was evaluated by qPCR and flow cytometry. Therapeutic response to 225 Ac-NM600+anti-CTLA4+anti-PD-L1 immune checkpoint inhibition (ICI) was evaluated in wild-type and stimulator of interferon genes knockout (STING KO) B78. The timing and magnitude of IFN1 response correlated with radionuclide half-life and linear energy transfer. CD8 + /Treg ratios increased in tumors 7 days after 90 Y- and 177 Lu-NM600 and day 21 after 225 Ac-NM600. 225 Ac-NM600+ICI improved survival in mice with WT but not with STING KO tumors, relative to monotherapies. Immunomodulatory effects of RPT vary with radioisotope and promote STING-dependent enhanced response to ICIs in murine models. Teaser This study describes the time course and nature of tumor immunomodulation by radiopharmaceuticals with differing physical properties.
Collapse
|
11
|
Waked A, Crabbé M, Neirinckx V, Pérez SR, Wellens J, Rogister B, Benotmane MA, Vermeulen K. Preclinical evaluation of CXCR4 peptides for targeted radionuclide therapy in glioblastoma. EJNMMI Radiopharm Chem 2024; 9:52. [PMID: 39008219 PMCID: PMC11250742 DOI: 10.1186/s41181-024-00282-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024] Open
Abstract
BACKGROUND Glioblastoma (GBM), is the most fatal form of brain cancer, with a high tendency for recurrence despite combined treatments including surgery, radiotherapy, and chemotherapy with temozolomide. The C-X-C chemokine receptor 4 (CXCR4) plays an important role in tumour radioresistance and recurrence, and is considered as an interesting GBM target. TRT holds untapped potential for GBM treatment, with CXCR4-TRT being a promising strategy for recurrent GBM treatment. Our study focuses on the preclinical assessment of different 177Lu-labelled CXCR4-targeting peptides, CTCE-9908, DV1-K-DV3, and POL3026 for GBM treatment and exploring some of the radiobiological mechanisms underlying these therapies. RESULTS All three DOTA-conjugated peptides could be radiolabelled with 177Lu with > 95% radiochemical yield. Binding studies show high specific binding of [177Lu]Lu-DOTA-POL3026 to U87-CXCR4 + cells, with 42% of the added activity binding to the membrane at 1 nM, and 6.5% internalised into the cells. In the presence of the heterologous CXCR4 blocking agent, AMD11070, membrane binding was reduced by 95%, a result confirmed by quantitative in vitro autoradiography of orthotopic GBM xenograft sections. An activity-dependent decrease in cell viability was observed for [177Lu]Lu-DOTA-DV1-K-DV3 and [177Lu]Lu-DOTA-POL3026, along with a slight increase in the induction of apoptotic markers. Additionally, the expression of γH2AX increased in a time-and activity-dependent manner. Ex vivo biodistribution studies with [177Lu]Lu-DOTA-POL3026 show uptake in the tumour reaching a SUV of 1.9 at 24 h post-injection, with higher uptake in the kidneys, lungs, spleen, and liver. Dosimetry estimations show an absorbed dose of 0.93 Gy/MBq in the tumour. A blocking study with AMD11070 showed a 38% reduction in tumour uptake, with no significant reduction observed in µSPECT imaging. Although no brain uptake was observed in the ex vivo biodistribution study, autoradiography on U87-CXCR4 + tumour inoculated mouse brain slices shows non-specific binding in the brain, next to high specific binding to the tumour. CONCLUSIONS In conclusion, we compared different 177Lu-radiolabelled CXCR4-targeting peptides for their binding potential in GBM, and demonstrated their varied cytotoxic action against GBM cells in vitro, with POL3026 being the most promising, causing considerable DNA damage. Though the peptide's systemic biodistribution remains to be improved, our data demonstrate the potential of [177Lu]Lu-DOTA-POL3026 for CXCR4-TRT in the context of GBM.
Collapse
Affiliation(s)
- Anthony Waked
- Nuclear Medical Applications Institute, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
- Laboratory of Nervous System Disorders and Therapy, GIGA Neurosciences, Université de Liège, Liège, Belgium
| | - Melissa Crabbé
- Nuclear Medical Applications Institute, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
| | - Virginie Neirinckx
- Laboratory of Nervous System Disorders and Therapy, GIGA Neurosciences, Université de Liège, Liège, Belgium
| | - Sunay Rodriguez Pérez
- Nuclear Medical Applications Institute, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
| | - Jasmien Wellens
- Nuclear Medical Applications Institute, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
| | - Bernard Rogister
- Laboratory of Nervous System Disorders and Therapy, GIGA Neurosciences, Université de Liège, Liège, Belgium
- Neurology Department, CHU de Liège, Liège, Belgium
| | - M Abderrafi Benotmane
- Nuclear Medical Applications Institute, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
| | - Koen Vermeulen
- Nuclear Medical Applications Institute, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium.
| |
Collapse
|
12
|
Trusova V, Karnaukhov I, Zelinsky A, Borts B, Ushakov I, Sidenko L, Gorbenko G. Radiolabeling of bionanomaterials with technetium 99m: current state and future prospects. Nanomedicine (Lond) 2024; 19:1569-1580. [PMID: 39011593 PMCID: PMC11321418 DOI: 10.1080/17435889.2024.2368454] [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: 03/04/2024] [Accepted: 06/12/2024] [Indexed: 07/17/2024] Open
Abstract
Radiolabeling of bionanomaterials with technetium-99m (99mTc) has become a promising approach in combining the benefits of nanotechnology and nuclear medicine for diagnostic and therapeutic purposes. This review is intended to provide a comprehensive overview of the state-of-the-art of radiolabeling of bionanomaterials with 99mTc, highlighting the synthesis methods, labeling mechanisms, biological evaluation, physicochemical characterization and clinical applications of 99mTc-labeled bionanomaterials. Various types of nanomaterials are considered in the review, including lipid- and protein-based nanosystems, dendrimers and polymeric nanomaterials. Moreover, the review assesses the challenges presented by this emerging field, such as stability of the radiolabel, potential toxicity of the nanomaterials and regulatory aspects. Finally, promising future perspectives and areas of research development in 99mTc-labeled bionanomaterials are discussed.
Collapse
Affiliation(s)
- Valeriya Trusova
- Department of Medical Physics & Biomedical Nanotechnologies, V.N. Karazin Kharkiv National University, 4 Svobody Sq., Kharkiv61022, Ukraine
| | - Ivan Karnaukhov
- National Science Center “Kharkov Institute of Physics and Technology”, Kharkiv, Ukraine
| | - Andrey Zelinsky
- National Science Center “Kharkov Institute of Physics and Technology”, Kharkiv, Ukraine
| | - Borys Borts
- National Science Center “Kharkov Institute of Physics and Technology”, Kharkiv, Ukraine
| | - Igor Ushakov
- National Science Center “Kharkov Institute of Physics and Technology”, Kharkiv, Ukraine
| | - Larysa Sidenko
- National Science Center “Kharkov Institute of Physics and Technology”, Kharkiv, Ukraine
| | - Galyna Gorbenko
- Department of Medical Physics & Biomedical Nanotechnologies, V.N. Karazin Kharkiv National University, 4 Svobody Sq., Kharkiv61022, Ukraine
| |
Collapse
|
13
|
Maitz CA, Bryan JN. The role of companion animal models in radiopharmaceutical development and translation. Vet Comp Oncol 2024; 22:165-173. [PMID: 38439693 DOI: 10.1111/vco.12969] [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] [Revised: 01/29/2024] [Accepted: 02/14/2024] [Indexed: 03/06/2024]
Abstract
Advancements in molecular imaging and drug targeting have created a renaissance in the development of radiopharmaceuticals for therapy and theranostics. While some radiopharmaceuticals, such as Na[131I]I, have been used clinically for decades, new agents are being approved using small-molecules, peptides, and antibodies for targeting. As these agents are being developed, the need to understand dosimetry and biologic effects of the systemically delivered radiotherapy becomes more important, particularly as highly potent radiopharmaceuticals using targeted alpha therapy become clinically utilized. As the processes being targeted become more complex, and the radiobiology of different particulate radiation becomes more diverse, models that better recapitulate human cancer and geometry are necessary. Companion animals develop many of the same types of cancer, carrying many of the same genetic drivers as those seen in people, and the scale and geometry of tumours in dogs more closely mimics those in humans than murine tumour models. Key translational challenges in oncology, such as alterations in tumour microenvironment, hypoxia, heterogeneity, and geometry are addressed by companion animal models. This review paper will provide background on radiopharmaceutical targeting techniques, review the use of radiopharmaceuticals in companion animal oncology, and explore the translational value of treating these patients in terms of dosimetry, treatment outcomes, and normal tissue complication rates.
Collapse
Affiliation(s)
- Charles A Maitz
- Comparative Oncology Radiobiology and Epigenetics Laboratory, Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, Missouri, USA
| | - Jeffrey N Bryan
- Comparative Oncology Radiobiology and Epigenetics Laboratory, Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, Missouri, USA
| |
Collapse
|
14
|
Dai D, Yu J, Gou W, Yang S, Li Y, Wang Z, Yang Z, Huang T, Li P, Zhu T, Hou W, Zhao Y, Xu W, Li Y. Novel CDK19-Targeted Radiotracers: A Potential Design Strategy to Improve the Pharmacokinetics and Tumor Uptake. J Med Chem 2024; 67:6726-6737. [PMID: 38570733 DOI: 10.1021/acs.jmedchem.4c00281] [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: 04/05/2024]
Abstract
Cyclin-dependent kinase 19 (CDK19) is overexpressed in prostate cancer, making it an attractive target for both imaging and therapy. Since little is known about the optimized approach for radioligands of nuclear proteins, linker optimization strategies were used to improve pharmacokinetics and tumor absorption, including the adjustment of the length, flexibility/rigidity, and hydrophilicity/lipophilicity of linkers. Molecular docking was conducted for virtual screening and followed by IC50 determination. Both BALB/c mice and P-16 xenografts were used for tissue distribution and PET/CT imaging. The ligand 68Ga-10c demonstrated high absorption in tumor 5 min after injection and sustains long-term imaging within 3 h. Furthermore, 68Ga-10c exhibited slow clearance within the tumor and was predominantly metabolized in both the liver and kidneys, showing the potential to alleviate metabolic pressure and enhance tissue safety. Therefore, the linker optimization strategy is well suited for CDK19 and provides a reference for the radioactive ligands of other nuclear targets.
Collapse
Affiliation(s)
- Dong Dai
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for China, Tianjin 300060, China
- Department of Molecular Medicine, Tianjin Cancer Hospital Airport Hospital, National Clinical Research Center for Cancer, Tianjin 300308, China
| | - Jiang Yu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Wenfeng Gou
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Shuangmeng Yang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Yanli Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Ziyang Wang
- Department of Molecular Medicine, Tianjin Cancer Hospital Airport Hospital, National Clinical Research Center for Cancer, Tianjin 300308, China
| | - Zhao Yang
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for China, Tianjin 300060, China
| | - Ting Huang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Panfeng Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Tong Zhu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Wenbin Hou
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Yu Zhao
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Wengui Xu
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for China, Tianjin 300060, China
| | - Yiliang Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| |
Collapse
|
15
|
Shea AG, Idrissou MB, Torres AI, Chen T, Hernandez R, Morris ZS, Sodji QH. Immunological effects of radiopharmaceutical therapy. FRONTIERS IN NUCLEAR MEDICINE (LAUSANNE, SWITZERLAND) 2024; 4:1331364. [PMID: 39355211 PMCID: PMC11440989 DOI: 10.3389/fnume.2024.1331364] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 03/14/2024] [Indexed: 10/03/2024]
Abstract
Radiation therapy (RT) is a pillar of cancer therapy used by more than half of all cancer patients. Clinically, RT is mostly delivered as external beam radiation therapy (EBRT). However, the scope of EBRT is limited in the metastatic setting, where all sites of disease need to be irradiated. Such a limitation is attributed to radiation-induced toxicities, for example on bone marrow and hematologic toxicities, resulting from a large EBRT field. Radiopharmaceutical therapy (RPT) has emerged as an alternative to EBRT for the irradiation of all sites of metastatic disease. While RPT can reduce tumor burden, it can also impact the immune system and anti-tumor immunity. Understanding these effects is crucial for predicting and managing treatment-related hematological toxicities and optimizing their integration with other therapeutic modalities, such as immunotherapies. Here, we review the immunomodulatory effects of α- and β-particle emitter-based RPT on various immune cell lines, such as CD8+ and CD4+ T cells, natural killer (NK) cells, and regulatory T (Treg) cells. We briefly discuss Auger electron-emitter (AEE)-based RPT, and finally, we highlight the combination of RPT with immune checkpoint inhibitors, which may offer potential therapeutic synergies for patients with metastatic cancers.
Collapse
Affiliation(s)
- Amanda G. Shea
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Malick Bio Idrissou
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Ana Isabel Torres
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Tessa Chen
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Reiner Hernandez
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Zachary S. Morris
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Quaovi H. Sodji
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
16
|
Ramonaheng K, Qebetu M, Ndlovu H, Swanepoel C, Smith L, Mdanda S, Mdlophane A, Sathekge M. Activity quantification and dosimetry in radiopharmaceutical therapy with reference to 177Lutetium. FRONTIERS IN NUCLEAR MEDICINE (LAUSANNE, SWITZERLAND) 2024; 4:1355912. [PMID: 39355215 PMCID: PMC11440950 DOI: 10.3389/fnume.2024.1355912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 03/12/2024] [Indexed: 10/03/2024]
Abstract
Radiopharmaceutical therapy has been widely adopted owing primarily to the development of novel radiopharmaceuticals. To fully utilize the potential of these RPTs in the era of precision medicine, therapy must be optimized to the patient's tumor characteristics. The vastly disparate dosimetry methodologies need to be harmonized as the first step towards this. Multiple factors play a crucial role in the shift from empirical activity administration to patient-specific dosimetry-based administrations from RPT. Factors such as variable responses seen in patients with presumably similar clinical characteristics underscore the need to standardize and validate dosimetry calculations. These efforts combined with ongoing initiatives to streamline the dosimetry process facilitate the implementation of radiomolecular precision oncology. However, various challenges hinder the widespread adoption of personalized dosimetry-based activity administration, particularly when compared to the more convenient and resource-efficient approach of empiric activity administration. This review outlines the fundamental principles, procedures, and methodologies related to image activity quantification and dosimetry with a specific focus on 177Lutetium-based radiopharmaceuticals.
Collapse
Affiliation(s)
- Keamogetswe Ramonaheng
- Department of Medical Physics and Radiobiology, Nuclear Medicine Research, Infrastructure (NuMeRI) NPC, Pretoria, South Africa
- Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria, South Africa
- Department of Nuclear Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Milani Qebetu
- Department of Medical Physics and Radiobiology, Nuclear Medicine Research, Infrastructure (NuMeRI) NPC, Pretoria, South Africa
- Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria, South Africa
| | - Honest Ndlovu
- Department of Medical Physics and Radiobiology, Nuclear Medicine Research, Infrastructure (NuMeRI) NPC, Pretoria, South Africa
- Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria, South Africa
- Department of Nuclear Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Cecile Swanepoel
- Department of Medical Physics and Radiobiology, Nuclear Medicine Research, Infrastructure (NuMeRI) NPC, Pretoria, South Africa
- Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria, South Africa
| | - Liani Smith
- Department of Medical Physics and Radiobiology, Nuclear Medicine Research, Infrastructure (NuMeRI) NPC, Pretoria, South Africa
- Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria, South Africa
| | - Sipho Mdanda
- Department of Medical Physics and Radiobiology, Nuclear Medicine Research, Infrastructure (NuMeRI) NPC, Pretoria, South Africa
- Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria, South Africa
- Department of Nuclear Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Amanda Mdlophane
- Department of Medical Physics and Radiobiology, Nuclear Medicine Research, Infrastructure (NuMeRI) NPC, Pretoria, South Africa
- Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria, South Africa
- Department of Nuclear Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Mike Sathekge
- Department of Medical Physics and Radiobiology, Nuclear Medicine Research, Infrastructure (NuMeRI) NPC, Pretoria, South Africa
- Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria, South Africa
- Department of Nuclear Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
17
|
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
|
18
|
Arbuznikova D, Eder M, Grosu AL, Meyer PT, Gratzke C, Zamboglou C, Eder AC. Towards Improving the Efficacy of PSMA-Targeting Radionuclide Therapy for Late-Stage Prostate Cancer-Combination Strategies. Curr Oncol Rep 2023; 25:1363-1374. [PMID: 37861915 PMCID: PMC10640479 DOI: 10.1007/s11912-023-01458-6] [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] [Accepted: 09/04/2023] [Indexed: 10/21/2023]
Abstract
PURPOSE OF REVIEW [177Lu]Lu-PSMA-617 is a radiopharmaceutical that emits beta-minus radiation and targets prostate-specific membrane antigen (PSMA)-positive prostate cancer. Despite its clinical success, there are still patients not showing sufficient response rates. This review compiles latest studies aiming at therapy improvement in [177Lu]Lu-PSMA-617-naïve and -resistant patients by alternative or combination treatments. RECENT FINDINGS A variety of agents to combine with [177Lu]Lu-PSMA-617 are currently under investigation including alpha radiation-emitting pharmaceuticals, radiosensitizers, taxane chemotherapeutics, androgen receptor pathway inhibitors, immune checkpoint inhibitors, and external beam radiation. Actinium-225 (225Ac)-labeled PSMA-targeting inhibitors are the most studied pharmaceuticals for combination therapy or as an alternative for treatment after progression under [177Lu]Lu-PSMA-617 therapy. Alpha emitters seem to have a potential of achieving a response to PSMA-targeting radionuclide therapy in both initial non-responders or responders to [177Lu]Lu-PSMA-617 later developing treatment resistance. Emerging evidence for immunostimulatory effects of radiopharmaceuticals and first prospective studies support the combination of [177Lu]Lu-PSMA-617 and immune checkpoint inhibition for late-stage prostate cancer.
Collapse
Affiliation(s)
- Daria Arbuznikova
- Department of Nuclear Medicine, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany
- Division of Radiopharmaceutical Development, German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany and German Cancer Research Center, Heidelberg, Germany
- Department of Radiation Oncology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Matthias Eder
- Department of Nuclear Medicine, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany.
- Division of Radiopharmaceutical Development, German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany and German Cancer Research Center, Heidelberg, Germany.
| | - Anca-Ligia Grosu
- Department of Radiation Oncology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Philipp T Meyer
- Department of Nuclear Medicine, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany
| | - Christian Gratzke
- Department of Urology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Constantinos Zamboglou
- Department of Radiation Oncology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ann-Christin Eder
- Department of Nuclear Medicine, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany
- Division of Radiopharmaceutical Development, German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany and German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
19
|
Abdollahi H, Saboury B, Soltani M, Shi K, Uribe C, Rahmim A. Radiopharmaceutical therapy on-a-chip: a perspective on microfluidic-driven digital twins towards personalized cancer therapies. Sci Bull (Beijing) 2023; 68:1983-1988. [PMID: 37573246 DOI: 10.1016/j.scib.2023.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/14/2023]
Affiliation(s)
- Hamid Abdollahi
- Department of Radiology, University of British Columbia, Vancouver V5Z 1M9, Canada; Department of Integrative Oncology, BC Cancer Research Institute, Vancouver V5Z 1L3, Canada
| | - Babak Saboury
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver V5Z 1L3, Canada; Department of Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda 20892, USA
| | - Madjid Soltani
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver V5Z 1L3, Canada; Department of Electrical & Computer Engineering, University of Waterloo, Waterloo N2L 3G1, Canada
| | - Kuangyu Shi
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern 3010, Switzerland; Computer Aided Medical Procedures and Augmented Reality, Institute of Informatics, Technical University of Munich, Munich 80333, Germany
| | - Carlos Uribe
- Department of Radiology, University of British Columbia, Vancouver V5Z 1M9, Canada; Department of Integrative Oncology, BC Cancer Research Institute, Vancouver V5Z 1L3, Canada; Functional Imaging, BC Cancer, Vancouver V5Z 4E6, Canada
| | - Arman Rahmim
- Department of Radiology, University of British Columbia, Vancouver V5Z 1M9, Canada; Department of Integrative Oncology, BC Cancer Research Institute, Vancouver V5Z 1L3, Canada; Department of Physics & Astronomy, University of British Columbia, Vancouver V6T 1Z1, Canada.
| |
Collapse
|
20
|
Patel D, Nguyen A, Fleeting C, Patel AB, Mumtaz M, Lucke-Wold B. Precision medicine in neurosurgery: The evolving role of theranostics. INNOSC THERANOSTICS & PHARMACOLOGICAL SCIENCES 2023; 6:417. [PMID: 37601162 PMCID: PMC10439809 DOI: 10.36922/itps.417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/22/2023]
Abstract
Theranostics in neurosurgery is a rapidly advancing field of precision medicine that combines diagnostic and therapeutic modalities to optimize patient outcomes. This approach has the potential to provide real-time feedback during therapy and diagnose a condition while simultaneously providing treatment. One such form of theranostics is focused ultrasound, which has been found to be effective in inducing neuroablation and neuromodulation and improving the efficacy of chemotherapy drugs by disrupting the blood-brain barrier. Targeted radionuclide therapy, which pairs positron emission tomography tracers with therapeutic effects and imaging modalities, is another promising form of theranostics for neurosurgery. Automated pathology analysis is yet another form of theranostics that can provide real-time feedback during the surgical resection of tumors. Electrical stimulation has also shown promise in optimizing therapies for patients with cerebral palsy. Overall, theranostics is a cost-effective way to optimize medical care for patients in neurosurgery. It is a relatively new field, but the advancements made so far show great promise for improving patient outcomes.
Collapse
Affiliation(s)
- Drashti Patel
- Department of Neurosurgery, University of Florida, College of Medicine, Gainesville, Florida, USA
| | - Andrew Nguyen
- Department of Neurosurgery, University of Florida, College of Medicine, Gainesville, Florida, USA
| | - Chance Fleeting
- Department of Neurosurgery, University of Florida, College of Medicine, Gainesville, Florida, USA
| | - Anjali B. Patel
- Department of Neurosurgery, University of Florida, College of Medicine, Gainesville, Florida, USA
| | - Mohammed Mumtaz
- Department of Neurosurgery, University of Florida, College of Medicine, Gainesville, Florida, USA
| | - Brandon Lucke-Wold
- Department of Neurosurgery, University of Florida, College of Medicine, Gainesville, Florida, USA
| |
Collapse
|
21
|
Giraudet AL, Vinceneux A, Pretet V, Paquet E, Lajusticia AS, Khayi F, Badel JN, Boyle H, Flechon A, Kryza D. Rationale for Prostate-Specific-Membrane-Antigen-Targeted Radionuclide Theranostic Applied to Metastatic Clear Cell Renal Carcinoma. Pharmaceuticals (Basel) 2023; 16:995. [PMID: 37513907 PMCID: PMC10383345 DOI: 10.3390/ph16070995] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 07/04/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023] Open
Abstract
Prostate-specific membrane antigen (PSMA), whose high expression has been demonstrated in metastatic aggressive prostate adenocarcinoma, is also highly expressed in the neovessels of various solid tumors, including clear cell renal cell carcinoma (ccRCC). In the VISION phase III clinical trial, PSMA-targeted radioligand therapy (PRLT) with lutetium 177 demonstrated a 4-month overall survival OS benefit compared to the best standard of care in heavily pretreated metastatic prostate cancer. Despite the improvement in the management of metastatic clear cell renal cell carcinoma (mccRCC) with antiangiogenic tyrosine kinase inhibitor (TKI) and immunotherapy, there is still a need for new treatments for patients who progress despite these drugs. In this study, we discuss the rationale of PRLT applied to the treavtment of mccRCC.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - David Kryza
- Lumen Nuclear Medicine Department, Hospices Civils de Lyon, 69437 Lyon, France
- UNIV Lyon-Université Claude Bernard Lyon 1, LAGEPP UMR 5007 CNRS Villeurbanne, 69100 Villeurbanne, France
| |
Collapse
|
22
|
Fernandes A, Oliveira A, Carvalho AL, Soares R, Barata P. Faecalibacterium prausnitzii in Differentiated Thyroid Cancer Patients Treated with Radioiodine. Nutrients 2023; 15:2680. [PMID: 37375584 DOI: 10.3390/nu15122680] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/05/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND Faecalibacterium prausnitzii, one of the most important bacteria of the human gut microbiota, produces butyrate (a short-chain fatty acid). Short-chain fatty acids are known to influence thyroid physiology and thyroid cancer's response to treatment. We aimed to analyze the relative abundance of Faecalibacterium prausnitzii on the gut microbiota of differentiated thyroid cancer patients compared to controls and its variation after radioiodine therapy (RAIT). METHODS Fecal samples were collected from 37 patients diagnosed with differentiated thyroid cancer before and after radioiodine therapy and from 10 volunteers. The abundance of F. prausnitzii was determined using shotgun metagenomics. RESULTS Our study found that the relative abundance of F. prausnitzii is significantly reduced in thyroid cancer patients compared to volunteers. We also found that there was a mixed response to RAIT, with an increase in the relative and absolute abundances of this bacterium in most patients. CONCLUSIONS Our study confirms that thyroid cancer patients present a dysbiotic gut microbiota, with a reduction in F. prausnitzii's relative abundance. In our study, radioiodine did not negatively affect F. prausnitzii, quite the opposite, suggesting that this bacterium might play a role in resolving radiation aggression issues.
Collapse
Affiliation(s)
- Ana Fernandes
- Department of Nuclear Medicine, Centro Hospitalar Universitário de São João, E.P.E., 4200-319 Porto, Portugal
| | - Ana Oliveira
- Department of Nuclear Medicine, Centro Hospitalar Universitário de São João, E.P.E., 4200-319 Porto, Portugal
| | - Ana Luísa Carvalho
- Department of Nuclear Medicine, Centro Hospitalar Universitário de São João, E.P.E., 4200-319 Porto, Portugal
| | - Raquel Soares
- Department of Biomedicine, Faculdade de Medicina da Universidade do Porto, 4200-319 Porto, Portugal
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - Pedro Barata
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- Department of Pharmaceutical Science, Faculdade de Ciências da Saúde da Universidade Fernando Pessoa, 4249-004 Porto, Portugal
- Department of Pathology, Centro Hospitalar Universitário do Porto, 4099-001 Porto, Portugal
| |
Collapse
|
23
|
Kauffman N, Singh SK, Morrison J, Zinn KR. Effective therapy with Bismuth-212 labeled macroaggregated albumin in orthotopic mouse breast tumor models. Front Chem 2023; 11:1204872. [PMID: 37234203 PMCID: PMC10206259 DOI: 10.3389/fchem.2023.1204872] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 05/02/2023] [Indexed: 05/27/2023] Open
Abstract
Intravascularly administered radiation therapy using beta (β-)-emitting radioisotopes has relied on either intravenously injected radiolabeled peptides that target cancer or radiolabeled microspheres that are trapped in the tumor following intra-arterial delivery. More recently, targeted intravenous radiopeptide therapies have explored the use of alpha (α)-particle emitting radioisotopes, but microspheres radiolabeled with α-particle emitters have not yet been studied. Here, FDA-approved macroaggregated albumin (MAA) particles were radiolabeled with Bismuth-212 (Bi-212-MAA) and evaluated using clonogenic and survival assays in vitro and using immune-competent mouse models of breast cancer. The in vivo biodistribution of Bi-212-MAA was investigated in Balb/c and C57BL/6 mice with 4T1 and EO771 orthotopic breast tumors, respectively. The same orthotopic breast cancer models were used to evaluate the treatment efficacy of Bi-212-MAA. Our results showed that macroaggregated albumin can be stably radiolabeled with Bi-212 and that Bi-212-MAA can deliver significant radiation therapy to reduce the growth and clonogenic potential of 4T1 and EO771 cells in vitro. Additionally, Bi-212-MAA treatment upregulated γH2AX and cleaved Caspase-3 expression in 4T1 cells. Biodistribution analyses showed 87-93% of the Bi-212-MAA remained in 4T1 and EO771 tumors 2 and 4 h after injection. Following single-tumor treatments with Bi-212-MAA there was a significant reduction in the growth of both 4T1 and EO771 breast tumors over the 18-day monitoring period. Overall, these findings showed that Bi-212-MAA was stably radiolabeled and inhibited breast cancer growth. Bi-212-MAA is an exciting platform to study α-particle therapy and will be easily translatable to larger animal models and human clinical trials.
Collapse
Affiliation(s)
- Nathan Kauffman
- Comparative Medicine and Integrative Biology, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, United States
| | - Satyendra Kumar Singh
- Department of Biomedical Engineering, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, United States
| | - James Morrison
- Advanced Radiology Services, Grand Rapids, MI, United States
| | - Kurt R. Zinn
- Departments of Radiology, Biomedical Engineering, Small Animal Clinical Sciences, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
24
|
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
|
25
|
Bellavia MC, Patel RB, Anderson CJ. Combined Targeted Radiopharmaceutical Therapy and Immune Checkpoint Blockade: From Preclinical Advances to the Clinic. J Nucl Med 2022; 63:1636-1641. [PMID: 36215570 PMCID: PMC12079708 DOI: 10.2967/jnumed.122.264373] [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: 05/08/2022] [Revised: 08/13/2022] [Indexed: 11/16/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized cancer care, but many patients with poorly immunogenic tumors fail to benefit. Preclinical studies have shown that external beam radiotherapy (EBRT) can synergize with ICI to prompt remarkable tumor regression and even eradication. However, EBRT is poorly suited to widely disseminated disease. Targeted radiopharmaceutical therapy (TRT) selectively delivers radiation to both the primary tumor and the metastatic sites, and promising results achieved with this approach have led to regulatory approval of certain agents (e.g., 177Lu-PSMA-617/Pluvicto for metastatic prostate cancer). To further improve therapeutic outcomes, combining TRT and ICI is a burgeoning research area, both preclinically and in clinical trials. Here we introduce basic TRT radiobiology and survey emerging and clinically translated TRT agents that have been combined with ICI.
Collapse
Affiliation(s)
- Michael C Bellavia
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Chemistry, University of Missouri, Columbia, Missouri
| | - Ravi B Patel
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Radiation Oncology, University of Pittsburgh, Pittsburgh, Pennsylvania; and
| | - Carolyn J Anderson
- Department of Chemistry, University of Missouri, Columbia, Missouri;
- Department of Radiology, University of Missouri, Columbia, Missouri
| |
Collapse
|
26
|
Sun J, Huangfu Z, Yang J, Wang G, Hu K, Gao M, Zhong Z. Imaging-guided targeted radionuclide tumor therapy: From concept to clinical translation. Adv Drug Deliv Rev 2022; 190:114538. [PMID: 36162696 DOI: 10.1016/j.addr.2022.114538] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 09/03/2022] [Accepted: 09/11/2022] [Indexed: 01/24/2023]
Abstract
Since the first introduction of sodium iodide I-131 for use with thyroid patients almost 80 years ago, more than 50 radiopharmaceuticals have reached the markets for a wide range of diseases, especially cancers. The nuclear medicine paradigm also shifts from solely molecular imaging or radionuclide therapy to imaging-guided radionuclide therapy, which is deemed a vital component of precision cancer therapy and an emerging medical modality for personalized medicine. The imaging-guided radionuclide therapy highlights the systematic integration of targeted nuclear diagnostics and radionuclide therapeutics. Regarding this, nuclear imaging serves to "visualize" the lesions and guide the therapeutic strategy, followed by administration of a precise patient specific dose of radiotherapeutics for treatment according to the absorbed dose to different organs and tumors calculated by dosimetry tools, and finally repeated imaging to predict the prognosis. This strategy leads to significantly enhanced therapeutic efficacy, improved patient outcomes, and manageable adverse events. In this review, we provide an overview of imaging-guided targeted radionuclide therapy for different tumors such as advanced prostate cancer and neuroendocrine tumors, with a focus on development of new radioligands and their preclinical and clinical results, and further discuss about challenges and future perspectives.
Collapse
Affiliation(s)
- Juan Sun
- College of Pharmaceutical Sciences, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China; Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China
| | - Zhenyuan Huangfu
- College of Pharmaceutical Sciences, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China; Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China
| | - Jiangtao Yang
- College of Pharmaceutical Sciences, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China; Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China
| | - Guanglin Wang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, People's Republic of China.
| | - Kuan Hu
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Sciences, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan.
| | - Mingyuan Gao
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, People's Republic of China
| | - Zhiyuan Zhong
- College of Pharmaceutical Sciences, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China; Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China.
| |
Collapse
|
27
|
Ionizing Radiation from Radiopharmaceuticals and the Human Gut Microbiota: An Ex Vivo Approach. Int J Mol Sci 2022; 23:ijms231810809. [PMID: 36142722 PMCID: PMC9506506 DOI: 10.3390/ijms231810809] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/04/2022] [Accepted: 09/11/2022] [Indexed: 11/17/2022] Open
Abstract
This study aimed to determine the effect of three widely used radiopharmaceuticals with intestinal excretion on selected relevant bacteria that are part of the human gut microbiota, using an ex vivo approach. Fecal samples obtained from healthy volunteers were analyzed. Each sample was divided into four smaller aliquots. One served as the non-irradiated control. The other three were homogenized with three radiopharmaceutical solutions ([131I]NaI, [99mTc]NaTcO4, and [223Ra]RaCl2). Relative quantification of each taxa was determined by the 2−ΔΔC method, using the ribosomal gene 16S as an internal control (primers 534/385). Twelve fecal samples were analysed: three controls and nine irradiated. Our experiment showed fold changes in all analyzed taxa with all radiopharmaceuticals, but results were more significant with I-131, ranging from 1.87–83.58; whereas no relevant differences were found with Tc-99m and Ra-223, ranging from 0.98–1.58 and 0.83–1.97, respectively. This study corroborates limited existing research on how ionizing radiation changes the gut microbiota composition, providing novel data regarding the ex vivo effect of radiopharmaceuticals. Our findings justify the need for future larger scale projects.
Collapse
|
28
|
Kiess AP, Hobbs RF, Bednarz B, Knox SJ, Meredith R, Escorcia FE. ASTRO's Framework for Radiopharmaceutical Therapy Curriculum Development for Trainees. Int J Radiat Oncol Biol Phys 2022; 113:719-726. [PMID: 35367328 DOI: 10.1016/j.ijrobp.2022.03.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 03/08/2022] [Accepted: 03/13/2022] [Indexed: 10/18/2022]
Abstract
In 2017, the American Society for Radiation Oncology (ASTRO) board of directors prioritized radiopharmaceutical therapy (RPT) as a leading area for new therapeutic development, and the ASTRO RPT workgroup was created. Herein, the workgroup has developed a framework for RPT curriculum development upon which education leaders can build to integrate this modality into radiation oncology resident education. Through this effort, the workgroup aims to provide a guide to ensure robust training in an emerging therapeutic area within the context of existing radiation oncology training in radiation biology, medical physics, and clinical radiation oncology. The framework first determines the core RPT knowledge required to select patients, prescribe, safely administer, and manage related adverse events. Then, it defines the most important topics for preparing residents for clinical RPT planning and delivery. This framework is designed as a tool to supplement the current training that exists for radiation oncology residents. The final document was approved by the ASTRO board of directors in the fall of 2021.
Collapse
Affiliation(s)
- Ana P Kiess
- Department of Radiation Oncology, Johns Hopkins University, Baltimore, Maryland.
| | - Robert F Hobbs
- Department of Radiation Oncology, Johns Hopkins University, Baltimore, Maryland
| | - Bryan Bednarz
- Department of Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin
| | - Susan J Knox
- Department of Radiation Oncology, Stanford University Medical Center, Stanford, California
| | - Ruby Meredith
- Department of Radiation Oncology, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Freddy E Escorcia
- Molecular Imaging Branch, Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
29
|
Melis DR, Burgoyne AR, Ooms M, Gasser G. Bifunctional chelators for radiorhenium: past, present and future outlook. RSC Med Chem 2022; 13:217-245. [PMID: 35434629 PMCID: PMC8942221 DOI: 10.1039/d1md00364j] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 01/14/2022] [Indexed: 01/16/2023] Open
Abstract
Targeted radionuclide therapy (TRNT) is an ever-expanding field of nuclear medicine that provides a personalised approach to cancer treatment while limiting toxicity to normal tissues. It involves the radiolabelling of a biological targeting vector with an appropriate therapeutic radionuclide, often facilitated by the use of a bifunctional chelator (BFC) to stably link the two entities. The radioisotopes of rhenium, 186Re (t 1/2 = 90 h, 1.07 MeV β-, 137 keV γ (9%)) and 188Re (t 1/2 = 16.9 h, 2.12 MeV β-, 155 keV γ (15%)), are particularly attractive for radiotherapy because of their convenient and high-abundance β--particle emissions as well as their imageable γ-emissions and chemical similarity to technetium. As a transition metal element with multiple oxidation states and coordination numbers accessible for complexation, there is great opportunity available when it comes to developing novel BFCs for rhenium. The purpose of this review is to provide a recap on some of the past successes and failings, as well as show some more current efforts in the design of BFCs for 186/188Re. Future use of these radionuclides for radiotherapy depends on their cost-effective availability and this will also be discussed. Finally, bioconjugation strategies for radiolabelling biomolecules with 186/188Re will be touched upon.
Collapse
Affiliation(s)
- Diana R Melis
- SCK CEN, Belgian Nuclear Research Centre Boeretang 200 BE-2400 Mol Belgium +1 865 341 1413 +32 14 33 32 83
- Chimie ParisTech, Laboratory for Inorganic Chemical Biology, PSL University F-75005 Paris France www.gassergroup.com +33 1 44 27 56 02
| | - Andrew R Burgoyne
- SCK CEN, Belgian Nuclear Research Centre Boeretang 200 BE-2400 Mol Belgium +1 865 341 1413 +32 14 33 32 83
| | - Maarten Ooms
- SCK CEN, Belgian Nuclear Research Centre Boeretang 200 BE-2400 Mol Belgium +1 865 341 1413 +32 14 33 32 83
| | - Gilles Gasser
- Chimie ParisTech, Laboratory for Inorganic Chemical Biology, PSL University F-75005 Paris France www.gassergroup.com +33 1 44 27 56 02
| |
Collapse
|
30
|
Wieczorek Villas Boas CA, de Jesus Silva J, Pereira Dias LA, Brandão Freire MR, Balieiro LM, Ferreira Dos Santos CS, Vivaldini BF, Benedetto R, Vieira DP, de Queiroz Souza Passos P, Marumo MH, Teixeira LFS, Bortoleti de Araújo E. In vitro and in vivo response of PSMA-617 radiolabeled with CA and NCA lutetium-177. Appl Radiat Isot 2021; 180:110064. [PMID: 34923290 DOI: 10.1016/j.apradiso.2021.110064] [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: 07/11/2021] [Revised: 12/09/2021] [Accepted: 12/09/2021] [Indexed: 11/02/2022]
Abstract
The PSMA-targeted radionuclide therapy has been explored since 2015 with radioisotope lutetium-177, whose β- emission range is adequate for micrometastases treatment. This radioisotope is obtained by two different production routes that directly affect the specific activity of lutetium-177 (non-carrier added and carrier added) and, consequently, the specific activity of radiopharmaceuticals, like 177Lu-PSMA-617. The influence of the specific activity of lutetium-177 on the properties of the radiopharmaceutical PSMA-617 was evaluated through pre-clinical studies. The in vitro study pointed to a lower constant of dissociation with non-carrier added lutetium-177 due to the difference in the specific activity. However, competition and internalization assays resulted in similar results for both lutetium-177. Based on these pre-clinical experiments, the total in vitro tumor cell binding and tumor uptake in vivo were similar, with no influence of the specific activity of the 177Lu-PSMA-617. Regardless the specific activity did not directly affect tumor uptake, the tumor/non-target organs ratios were higher for the radiopharmaceutical labeled with carrier added lutetium-177, which had the lowest specific activity.
Collapse
Affiliation(s)
- Cristian Antonio Wieczorek Villas Boas
- Instituto de Pesquisas Energéticas e Nucleares, IPEN/CNEN, Av Prof. Lineu Prestes, 2242 - Cidade Universitária, São Paulo, CEP 05508-000, Brazil; Department of Radiation Oncology, Washington University School of Medicine, 4511 Forest Park Ave, St Louis, MO, 63110, USA.
| | - Jefferson de Jesus Silva
- Instituto de Pesquisas Energéticas e Nucleares, IPEN/CNEN, Av Prof. Lineu Prestes, 2242 - Cidade Universitária, São Paulo, CEP 05508-000, Brazil
| | - Luís Alberto Pereira Dias
- Instituto de Pesquisas Energéticas e Nucleares, IPEN/CNEN, Av Prof. Lineu Prestes, 2242 - Cidade Universitária, São Paulo, CEP 05508-000, Brazil
| | - Maria Renata Brandão Freire
- Instituto de Pesquisas Energéticas e Nucleares, IPEN/CNEN, Av Prof. Lineu Prestes, 2242 - Cidade Universitária, São Paulo, CEP 05508-000, Brazil
| | - Luiza Mascarenhas Balieiro
- Instituto de Pesquisas Energéticas e Nucleares, IPEN/CNEN, Av Prof. Lineu Prestes, 2242 - Cidade Universitária, São Paulo, CEP 05508-000, Brazil
| | - Carolina Silva Ferreira Dos Santos
- Instituto de Pesquisas Energéticas e Nucleares, IPEN/CNEN, Av Prof. Lineu Prestes, 2242 - Cidade Universitária, São Paulo, CEP 05508-000, Brazil
| | - Bianca Franchesqueti Vivaldini
- Instituto de Pesquisas Energéticas e Nucleares, IPEN/CNEN, Av Prof. Lineu Prestes, 2242 - Cidade Universitária, São Paulo, CEP 05508-000, Brazil
| | - Raquel Benedetto
- Instituto de Pesquisas Energéticas e Nucleares, IPEN/CNEN, Av Prof. Lineu Prestes, 2242 - Cidade Universitária, São Paulo, CEP 05508-000, Brazil
| | - Daniel Perez Vieira
- Instituto de Pesquisas Energéticas e Nucleares, IPEN/CNEN, Av Prof. Lineu Prestes, 2242 - Cidade Universitária, São Paulo, CEP 05508-000, Brazil
| | - Priscila de Queiroz Souza Passos
- Instituto de Pesquisas Energéticas e Nucleares, IPEN/CNEN, Av Prof. Lineu Prestes, 2242 - Cidade Universitária, São Paulo, CEP 05508-000, Brazil
| | - Maria Helena Marumo
- Instituto de Pesquisas Energéticas e Nucleares, IPEN/CNEN, Av Prof. Lineu Prestes, 2242 - Cidade Universitária, São Paulo, CEP 05508-000, Brazil
| | - Luis Felipe S Teixeira
- Instituto de Pesquisas Energéticas e Nucleares, IPEN/CNEN, Av Prof. Lineu Prestes, 2242 - Cidade Universitária, São Paulo, CEP 05508-000, Brazil
| | - Elaine Bortoleti de Araújo
- Instituto de Pesquisas Energéticas e Nucleares, IPEN/CNEN, Av Prof. Lineu Prestes, 2242 - Cidade Universitária, São Paulo, CEP 05508-000, Brazil
| |
Collapse
|
31
|
Lassmann M, Eberlein U, Gear J, Konijnenberg M, Kunikowska J. Dosimetry for Radiopharmaceutical Therapy: The European Perspective. J Nucl Med 2021; 62:73S-79S. [PMID: 34857624 DOI: 10.2967/jnumed.121.262754] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 10/13/2021] [Indexed: 11/16/2022] Open
Abstract
This review presents efforts in Europe over the last few years with respect to standardization of quantitative imaging and dosimetry and comprises the results of several European research projects on practices regarding radiopharmaceutical therapies (RPTs). Because the European Union has regulatory requirements concerning dosimetry in RPTs, the European Association of Nuclear Medicine released a position paper in 2021 on the use of dosimetry under these requirements. The importance of radiobiology for RPTs is elucidated in another position paper by the European Association of Nuclear Medicine. Furthermore, how dosimetry interacts with clinical requirements is described, with several clinical examples. In the future, more efforts need to be undertaken to increase teaching and standardization efforts and to incorporate radiobiology for further individualizing patient treatment, with the aim of improving the outcome and safety of RPTs.
Collapse
Affiliation(s)
- Michael Lassmann
- Department of Nuclear Medicine, University of Würzburg, Würzburg, Germany
| | - Uta Eberlein
- Department of Nuclear Medicine, University of Würzburg, Würzburg, Germany;
| | - Jonathan Gear
- Joint Department of Physics, Royal Marsden NHS Foundation Trust and Institute of Cancer Research, London, United Kingdom
| | - Mark Konijnenberg
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands; and
| | - Jolanta Kunikowska
- Nuclear Medicine Department, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
32
|
Aerts A, Eberlein U, Holm S, Hustinx R, Konijnenberg M, Strigari L, van Leeuwen FWB, Glatting G, Lassmann M. EANM position paper on the role of radiobiology in nuclear medicine. Eur J Nucl Med Mol Imaging 2021; 48:3365-3377. [PMID: 33912987 PMCID: PMC8440244 DOI: 10.1007/s00259-021-05345-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 03/28/2021] [Indexed: 12/16/2022]
Abstract
With an increasing variety of radiopharmaceuticals for diagnostic or therapeutic nuclear medicine as valuable diagnostic or treatment option, radiobiology plays an important role in supporting optimizations. This comprises particularly safety and efficacy of radionuclide therapies, specifically tailored to each patient. As absorbed dose rates and absorbed dose distributions in space and time are very different between external irradiation and systemic radionuclide exposure, distinct radiation-induced biological responses are expected in nuclear medicine, which need to be explored. This calls for a dedicated nuclear medicine radiobiology. Radiobiology findings and absorbed dose measurements will enable an improved estimation and prediction of efficacy and adverse effects. Moreover, a better understanding on the fundamental biological mechanisms underlying tumor and normal tissue responses will help to identify predictive and prognostic biomarkers as well as biomarkers for treatment follow-up. In addition, radiobiology can form the basis for the development of radiosensitizing strategies and radioprotectant agents. Thus, EANM believes that, beyond in vitro and preclinical evaluations, radiobiology will bring important added value to clinical studies and to clinical teams. Therefore, EANM strongly supports active collaboration between radiochemists, radiopharmacists, radiobiologists, medical physicists, and physicians to foster research toward precision nuclear medicine.
Collapse
Affiliation(s)
- An Aerts
- Radiobiology Unit, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
| | - Uta Eberlein
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany.
| | - Sören Holm
- Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, University Hospital Copenhagen, Copenhagen, Denmark
| | - Roland Hustinx
- Division of Nuclear Medicine and Oncological Imaging, University Hospital of Liège, GIGA-CRC in vivo Imaging, University of Liège, Liège, Belgium
| | - Mark Konijnenberg
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Lidia Strigari
- Medical Physics Department, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Fijs W B van Leeuwen
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Gerhard Glatting
- Medical Radiation Physics, Department of Nuclear Medicine, Ulm University, Ulm, Germany
| | - Michael Lassmann
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
33
|
Artigas C, Mileva M, Flamen P, Karfis I. Targeted radionuclide therapy: an emerging field in solid tumours. Curr Opin Oncol 2021; 33:493-499. [PMID: 34183491 DOI: 10.1097/cco.0000000000000762] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
PURPOSE OF REVIEW Targeted radionuclide therapy (TRNT) is characterized by systemic administration of radiolabelled drugs, targeting specific molecular alterations expressed on the tumour cells. Small molecules, labelled with β- or α- emitting radioisotopes, are used to deliver radiation directly to the tumour sites. Pretreatment imaging to visualize whole body biodistribution of the target, using the same drugs labelled with positron or γ-emitting radionuclides, completes the concept of theranostic. This review will briefly summarize the current clinical research findings and applications of TRNT in solid tumours, mostly focusing on neuroendocrine and prostate neoplasms. RECENT FINDINGS Peptide receptor radionuclide therapy is a major component in the management of gastroentropancreatic neuroendocrine tumours, with favourable safety profile, quality-of-life improvement and survival benefit. On the NETTER-1 study, it proved to be more effective than high-dose long-acting-release octreotide, leading to its regulatory approval. Prostate-specific membrane antigen (PSMA) is an excellent target for TRNT in prostate cancer. 177Lu-PSMA radioligand therapy demonstrated higher response rates in patients with metastatic castration resistant prostate cancer, when compared with second-line chemotherapy. New developments, including targeting of fibroblast activation proteins overexpressed in the tumour stroma, show promising preliminary results in the theranostic setting. SUMMARY Recent research has demonstrated and consolidated the use of TRNT against well established targets in neuroendocrine tumours and prostate cancer. The identification of new promising molecular targets for TRNT, will further expand the theranostic applications of radionuclides in the field of nuclear medicine.
Collapse
Affiliation(s)
- Carlos Artigas
- Department of Nuclear Medicine, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | | | | | | |
Collapse
|
34
|
Saluja V, Mishra Y, Mishra V, Giri N, Nayak P. Dendrimers based cancer nanotheranostics: An overview. Int J Pharm 2021; 600:120485. [PMID: 33744447 DOI: 10.1016/j.ijpharm.2021.120485] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 02/26/2021] [Accepted: 03/09/2021] [Indexed: 12/12/2022]
Abstract
Cancer is a known deadliest disease that requires a judicious diagnostic, targeting, and treatment strategy for an early prognosis and selective therapy. The major pitfalls of the conventional approach are non-specificity in targeting, failure to precisely monitor therapy outcome, and cancer progression leading to malignancies. The unique physicochemical properties offered by nanotechnology derived nanocarriers have the potential to radically change the landscape of cancer diagnosis and therapeutic management. An integrative approach of utilizing both diagnostic and therapeutic functionality using a nanocarrier is termed as nanotheranostic. The nanotheranostics platform is designed in such a way that overcomes various biological barriers, efficiently targets the payload to the desired locus, and simultaneously supports planning, monitoring, and verification of treatment delivery to demonstrate an enhanced therapeutic efficacy. Thus, a nanotheranostic platform could potentially assist in drug targeting, image-guided focal therapy, drug release and distribution monitoring, predictionof treatment response, and patient stratification. A class of highly branched nanocarriers known as dendrimers is recognized as an advanced nanotheranostic platform that has the potential to revolutionize the oncology arena by its unique and exciting features. A dendrimer is a well-defined three-dimensional globular chemical architecture with a high level of monodispersity, amenability of precise size control, and surface functionalization. All the dendrimer properties exhibit a reproducible pharmacokinetic behavior that could ensure the desired biodistribution and efficacy. Dendrimers are thus being exploited as a nanotheranostic platform embodying a diverse class of therapeutic, imaging, and targeting moieties for cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Vikrant Saluja
- Faculty of Pharmaceutical Sciences, PCTE Group of Institutes, Ludhiana, Punjab, India; School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Yachana Mishra
- Department of Zoology, Shri Shakti Degree College, Sankhahari, Ghatampur, Kanpur Nagar, Uttar Pradesh, India
| | - Vijay Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India.
| | - Namita Giri
- College of Pharmacy, Ferris State University, Big Rapids, MI 49307, USA
| | - Pallavi Nayak
- Faculty of Pharmaceutical Sciences, PCTE Group of Institutes, Ludhiana, Punjab, India; School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| |
Collapse
|