1
|
Bogdanovic B, Hugonnet F, Montemagno C. Theranostics in Hematological Malignancies: Cutting-Edge Advances in Diagnosis and Targeted Therapy. Cancers (Basel) 2025; 17:1247. [PMID: 40227793 PMCID: PMC11987953 DOI: 10.3390/cancers17071247] [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: 03/12/2025] [Revised: 04/02/2025] [Accepted: 04/04/2025] [Indexed: 04/15/2025] Open
Abstract
Hematologic malignancies, including leukemia, lymphoma, and multiple myeloma, continue to challenge clinicians with complex treatment regimens that often involve significant side effects and limited success, especially in advanced stages. Recent advancements in nuclear medicine have introduced theranostic strategies that merge diagnostic imaging with targeted therapeutic approaches, offering the potential for more precise and personalized treatment. A key area of progress lies in the development of alpha-emitting radiopharmaceuticals, such as 225Ac, 211At, or 212Pb, which can deliver potent radiation directly to tumor cells, sparing healthy tissue and minimizing collateral damage. In parallel with these therapeutic advancements, molecular imaging using radiolabeled agents enables better disease monitoring, assessment of treatment efficacy, and personalized management of patients with hematologic malignancies. The integration of diagnostic imaging with radiotherapy allows for a more tailored approach, where treatment can be adjusted based on real-time information about tumor progression and response. This review examines the recent strides made in both the development of radiopharmaceuticals and their applications in molecular imaging, with a focus on the potential to improve precision, reduce toxicity, and optimize patient outcomes. The synergy between targeted therapy and molecular imaging represents a transformative shift in the management of hematologic malignancies. As these technologies evolve, they are poised to redefine treatment paradigms, offering new hope for patients and potentially improving survival rates with more effective and less toxic treatment options.
Collapse
Affiliation(s)
- Bojana Bogdanovic
- Laboratory of Bioclinical Radiopharmaceutics, University Grenoble Alpes, INSERM, CHU Grenoble Alpes, 38000 Grenoble, France;
| | - Florent Hugonnet
- Nuclear Medicine Department, Centre Hospitalier Princesse Grace, 98000 Monaco, Monaco;
| | - Christopher Montemagno
- Laboratory of Bioclinical Radiopharmaceutics, University Grenoble Alpes, INSERM, CHU Grenoble Alpes, 38000 Grenoble, France;
- Biomedical Department, Centre Scientifique de Monaco, 98000 Monaco, Monaco
| |
Collapse
|
2
|
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
Affiliation(s)
- Donald Bunjes
- Department of Internal Medicine III, Ulm University Hospital, Ulm, Germany
| |
Collapse
|
3
|
Tosato M, Favaretto C, Kleynhans J, Burgoyne AR, Gestin JF, van der Meulen NP, Jalilian A, Köster U, Asti M, Radchenko V. Alpha Atlas: Mapping global production of α-emitting radionuclides for targeted alpha therapy. Nucl Med Biol 2024; 142-143:108990. [PMID: 39809026 DOI: 10.1016/j.nucmedbio.2024.108990] [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: 11/09/2024] [Revised: 12/06/2024] [Accepted: 12/17/2024] [Indexed: 01/16/2025]
Abstract
Targeted Alpha Therapy has shown great promise in cancer treatment, sparking significant interest over recent decades. However, its broad adoption has been impeded by the scarcity of alpha-emitters and the complexities related to their use. The availability of these radionuclides is often constrained by the intricate production processes and purification, as well as regulatory and logistical challenges. Moreover, the high cost and technical difficulties associated with handling and applying alpha-emitting radionuclides pose additional barriers to their clinical implementation. This Alpha Atlas provides an in-depth overview of the leading alpha-particle emitting radionuclide candidates for clinical use, focusing on their production processes and supply chains. By mapping the current facilities that produce and supply these radionuclides, this atlas aims to assist researchers, clinicians, and industries in initiating or scaling up the applications of alpha-emitters. The Alpha Atlas aspires to act as a strategic guide, facilitating collaboration and driving forward the integration of these potent therapeutic agents into cancer treatment practices.
Collapse
Affiliation(s)
- Marianna Tosato
- Radiopharmaceutical Chemistry Laboratory (RACHEL), Nuclear Medicine Unit, AUSL-IRCCS Reggio Emilia, 42123 Reggio Emilia, Italy.
| | - Chiara Favaretto
- Radiopharmacy and Cyclotron Department, IRCCS Sacro Cuore Don Calabria, Negrar 37024, Verona, Italy
| | - Janke Kleynhans
- Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Andrew R Burgoyne
- Oak Ridge National Laboratory, 1 Bethel Valley Road, Oak Ridge, TN 37830, United States
| | - Jean-François Gestin
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, 44000 Nantes, France
| | - Nicholas P van der Meulen
- PSI Center for Life Sciences, 5232 Villigen-PSI, Switzerland; PSI Center for Nuclear Engineering and Sciences, 5232 Villigen-PSI, Switzerland
| | - Amirreza Jalilian
- Department of Nuclear Safety and Security, International Atomic Energy Agency, 1220 Vienna, Austria
| | - Ulli Köster
- Institut Laue-Langevin, 38042 Grenoble, France
| | - Mattia Asti
- Radiopharmaceutical Chemistry Laboratory (RACHEL), Nuclear Medicine Unit, AUSL-IRCCS Reggio Emilia, 42123 Reggio Emilia, Italy
| | - Valery Radchenko
- Life Sciences Division, TRIUMF, BC V6T 2A3 Vancouver, British Columbia, Canada; Department of Chemistry, University of British Columbia, V6T 1Z1 Vancouver, British Columbia, Canada
| |
Collapse
|
4
|
Allen KH, Frank C, Jiao R, Malo ME, Bello M, De Nardo L, Meléndez-Alafort L, Dadachova E. In Vitro and In Vivo Comparison of Random versus Site-Specific Conjugation of Bifunctional Chelating Agents to the CD33-Binding Antibody for Use in Alpha- and Beta-Radioimmunotherapy. ACS OMEGA 2024; 9:50000-50011. [PMID: 39713652 PMCID: PMC11656351 DOI: 10.1021/acsomega.4c09450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/18/2024] [Accepted: 11/22/2024] [Indexed: 12/24/2024]
Abstract
Radiometal chelator conjugation is a cornerstone of radioimmunotherapy (RIT). Continued interest in selective placement of chelators remains an active topic of discussion in the field. With several simple site-specific methods being recently reported, it was of interest to investigate the benefits and potential drawbacks of the site-specific method with a full comparison to a more typical random conjugation method that is currently utilized in clinical applications. In this study, the conjugation methods were evaluated side by side to determine the utility of both methods using commercially available random and site-specific conjugation reagents by performing antigen binding; radiolabeling with 64Cu, 177Lu, and 225Ac radioisotopes; antibody-conjugate stability, cytotoxicity, in vivo distribution, pharmacokinetics analyses, and dosimetry to gather a whole data set for preclinical investigation. Evaluation revealed that both methods performed similarly during most experiments with the site-specific method, resulting in higher binding capacity of the antibody conjugate via flow cytometry. Radiolabeling was not significantly different between two methods, while stability showed that the site-specifically conjugated antibody was somewhat more stable at 37 °C in human serum over 1 week. In vitro experiments demonstrated less cell killing with the random conjugation method, while in vivo experiments showed no statistical differences in tumor uptake between conjugation methods. Dosimetry calculations were performed using the acquired PET/CT data and showed that apart from the liver, there was no significant difference in radiation doses delivered by either antibody conjugate. These results demonstrate that both methods are viable for future work, while the site-specific method offers several potential advantages and, in some cases, improved efficacy.
Collapse
Affiliation(s)
- Kevin
J. H. Allen
- College
of Pharmacy and Nutrition, University of
Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Connor Frank
- College
of Pharmacy and Nutrition, University of
Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Rubin Jiao
- College
of Pharmacy and Nutrition, University of
Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Mackenzie E. Malo
- College
of Pharmacy and Nutrition, University of
Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Michele Bello
- Department
of Physics and Astronomy, University of
Padua, Via Marzolo 8, Padua I-35131, Italy
| | - Laura De Nardo
- Department
of Physics and Astronomy, University of
Padua, Via Marzolo 8, Padua I-35131, Italy
| | - Laura Meléndez-Alafort
- Immunology
and Molecular Oncology Diagnostics Unit, Veneto Institute of Oncology IOV-IRCCS, Via Gattamelata 64, Padua 35128, Italy
| | - Ekaterina Dadachova
- College
of Pharmacy and Nutrition, University of
Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| |
Collapse
|
5
|
Zuo D, Wang H, Yu B, Li Q, Gan L, Chen W. Astatine-211 and actinium-225: two promising nuclides in targeted alpha therapy. Acta Biochim Biophys Sin (Shanghai) 2024; 57:327-343. [PMID: 39587859 PMCID: PMC11986457 DOI: 10.3724/abbs.2024206] [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: 07/11/2024] [Accepted: 10/22/2024] [Indexed: 11/27/2024] Open
Abstract
Nuclear medicine therapy offers a promising approach for tumor treatment, as the energy emitted during radionuclide decay causes irreparable damage to tumor cells. Notably, α-decay exhibits an even more significant destructive potential. By conjugating α-nuclides with antibodies or small-molecule inhibitors, targeted alpha therapy (TAT) can enhance tumor destruction while minimizing toxic side effects, making TAT an increasingly attractive antineoplastic strategy. Astatine-211 ( 211At) and actinium-225 ( 225Ac) have emerged as highly effective agents in TAT due to their exceptional physicochemical properties and biological effects. In this review, we highlight the applications of 211At-/ 225Ac-radiopharmaceuticals, particularly in specific tumor targets, such as prostate-specific membrane antigen (PSMA) in prostate cancers, cluster of differentiation (CD) in hematological malignancies, human epidermal growth factor receptor-2 (HER2) in ovarian cancers, and somatostatin receptor (SSTR) in neuroendocrine tumors. We synthesize the progress from preclinical and clinical trials to provide insights into the promising potential of 211At-/ 225Ac-radiopharmaceuticals for future treatments.
Collapse
Affiliation(s)
- Dashan Zuo
- Institute of Modern PhysicsChinese Academy of SciencesLanzhou730000China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in MedicineGansu ProvinceLanzhou730000China
- University of Chinese Academy of SciencesBeijing100049China
| | - Hui Wang
- Institute of Modern PhysicsChinese Academy of SciencesLanzhou730000China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in MedicineGansu ProvinceLanzhou730000China
| | - Boyi Yu
- Institute of Modern PhysicsChinese Academy of SciencesLanzhou730000China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in MedicineGansu ProvinceLanzhou730000China
| | - Qiang Li
- Institute of Modern PhysicsChinese Academy of SciencesLanzhou730000China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in MedicineGansu ProvinceLanzhou730000China
- University of Chinese Academy of SciencesBeijing100049China
- Lanhai Nuclear Medical Research CenterPutian351153China
| | - Lu Gan
- Institute of Modern PhysicsChinese Academy of SciencesLanzhou730000China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in MedicineGansu ProvinceLanzhou730000China
- University of Chinese Academy of SciencesBeijing100049China
| | - Weiqiang Chen
- Institute of Modern PhysicsChinese Academy of SciencesLanzhou730000China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in MedicineGansu ProvinceLanzhou730000China
- University of Chinese Academy of SciencesBeijing100049China
- Lanhai Nuclear Medical Research CenterPutian351153China
| |
Collapse
|
6
|
Costa IM, Firth G, Kim J, Banu A, Pham TT, Sunassee K, Langdon S, De Santis V, Vass L, Schettino G, Fruhwirth GO, Terry SYA. In Vitro and Preclinical Systematic Dose-Effect Studies of Auger Electron- and β Particle-Emitting Radionuclides and External Beam Radiation for Cancer Treatment. Int J Radiat Oncol Biol Phys 2024; 120:1124-1134. [PMID: 38797497 PMCID: PMC7616868 DOI: 10.1016/j.ijrobp.2024.05.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 04/09/2024] [Accepted: 05/17/2024] [Indexed: 05/29/2024]
Abstract
PURPOSE Despite a rise in clinical use of radiopharmaceutical therapies, the biological effects of radionuclides and their relationship with absorbed radiation dose are poorly understood. Here, we set out to define this relationship for Auger electron emitters [99mTc]TcO4- and [123I]I- and β--particle emitter [188Re]ReO4-. Studies were carried out using genetically modified cells that permitted direct radionuclide comparisons. METHODS AND MATERIALS Triple-negative MDA-MB-231 breast cancer cells expressing the human sodium iodide symporter (hNIS) and green fluorescent protein (GFP; MDA-MB-231.hNIS-GFP) were used. In vitro radiotoxicity of [99mTc]TcO4-, [123I]I-, and [188Re]ReO4- was determined using clonogenic assays. Radionuclide uptake, efflux, and subcellular location were used to calculate nuclear absorbed doses using the Medical Internal Radiation Dose (MIRD) formalism. In vivo studies were performed using female NSG mice bearing orthotopic MDA-MB-231.hNIS-GFP tumors and compared with X-ray-treated (12.6-15 Gy) and untreated cohorts. Absorbed dose per unit activity in tumors and sodium iodide symporter-expressing organs was extrapolated to reference human adult models using OLINDA/EXM. RESULTS [99mTc]TcO4- and [123I]I- reduced the survival fraction only in hNIS-expressing cells, whereas [188Re]ReO4- reduced survival fraction in hNIS-expressing and parental cells. [123I]I- required 2.4- and 1.5-fold lower decays/cell to achieve 37% survival compared with [99mTc]TcO4- and [188Re]ReO4-, respectively, after 72 hours of incubation. Additionally, [99mTc]TcO4-, [123I]I-, and [188Re]ReO4- had superior cell killing effectiveness in vitro compared with X-rays. In vivo, X-ray led to a greater median survival compared with [188Re]ReO4- and [123I]I- (54 days vs 45 and 43 days, respectively). Unlike the X-ray cohort, no metastases were visualized in the radionuclide-treated cohorts. Extrapolated human absorbed doses of [188Re]ReO4- to a 1 g tumor were 13.8- and 11.2-fold greater than for [123I]I- in female and male models, respectively. CONCLUSIONS This work reports reference dose-effect data using cell and tumor models for [99mTc]TcO4-, [123I]I-, and [188Re]ReO4- for the first time. We further demonstrate the tumor-controlling effects of [123I]I- and [188Re]ReO4- in comparison with external beam radiation therapy.
Collapse
Affiliation(s)
- Ines M Costa
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom; Imaging Therapies and Cancer Group, Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
| | - George Firth
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Jana Kim
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Arshiya Banu
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Truc T Pham
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Kavitha Sunassee
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Sophie Langdon
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Vittorio De Santis
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Laurence Vass
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Giuseppe Schettino
- Medical Radiation Science Group, National Physical Laboratory, Teddington, United Kingdom.
| | - Gilbert O Fruhwirth
- Imaging Therapies and Cancer Group, Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom.
| | - Samantha Y A Terry
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom.
| |
Collapse
|
7
|
Peng X, Tang F, Li Y, Bai J, Li L, Zhang L. Combination of BCL-2 inhibitors and immunotherapy: a promising therapeutic strategy for hematological malignancies. Discov Oncol 2024; 15:311. [PMID: 39060763 PMCID: PMC11282050 DOI: 10.1007/s12672-024-01161-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
The rapid development of high-throughput sequencing in recent years has facilitated great progress in the molecular-targeted therapy of hematological malignancies, including leukemia, lymphoma, and multiple myeloma. BCL-2 inhibitors are among the most important molecular-targeted agents. Immunotherapy for hematologic malignancy has rapidly increased in popularity in recent years and has been proven to improve the overall survival rate. However, few clinical studies have investigated combination therapy with BCL-2 inhibitors and immunotherapies, such as immune molecule-targeted drugs or immune cell adoptive therapy. In this review, we discuss the drug discovery process, current clinical application status, and resistance and tolerance issues associated with BCL-2 inhibitors. We emphasize their important role in regulating the immune system and propose that the combination of BCL-2 inhibitors with immunotherapy may be one of the most promising treatment methods for hematologic malignancies.
Collapse
Affiliation(s)
- Xiaohuan Peng
- Department of Hematology, The Second Hospital and Clinical Medical School, Lanzhou University, Lanzhou, China
- Key Laboratory of the Hematology of Gansu Province, The Second Hospital and Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Futian Tang
- Key Laboratory of the Digestive Tumor of Gansu Province, The Second Hospital and Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Yanhong Li
- Key Laboratory of the Hematology of Gansu Province, The Second Hospital and Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Jun Bai
- Key Laboratory of the Hematology of Gansu Province, The Second Hospital and Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Lijuan Li
- Department of Hematology, The Second Hospital and Clinical Medical School, Lanzhou University, Lanzhou, China.
- Key Laboratory of the Hematology of Gansu Province, The Second Hospital and Clinical Medical School, Lanzhou University, Lanzhou, China.
| | - Liansheng Zhang
- Department of Hematology, The Second Hospital and Clinical Medical School, Lanzhou University, Lanzhou, China.
- Key Laboratory of the Hematology of Gansu Province, The Second Hospital and Clinical Medical School, Lanzhou University, Lanzhou, China.
| |
Collapse
|
8
|
Tronchin S, Forster J, Hickson K, Bezak E. Modeling the effect of daughter migration on dosimetry estimates for unlabeled actinium-225. Med Phys 2024; 51:5032-5044. [PMID: 38197481 DOI: 10.1002/mp.16917] [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: 04/03/2023] [Revised: 12/05/2023] [Accepted: 12/13/2023] [Indexed: 01/11/2024] Open
Abstract
BACKGROUND Actinium-225 (225Ac) is an alpha emitting radionuclide which has demonstrated promising results in Targeted Alpha Therapy (TAT). A concern with 225Ac is that the decay energy can break the bond to the targeting vehicle, resulting in the release of free alpha-emitting daughter radionuclides in the body. PURPOSE The aim of this work is to develop a compartment model to describe the movement of unlabeled 225Ac in a human where the daughter isotopes of 225Ac have unique biokinetics. METHOD The ICRP Occupational Intake of Radionuclides reports were used to construct a compartment model for the 225Ac decay chain where the daughter isotopes of 225Ac are assigned their own unique transfer coefficients (TCs) between compartments. Computer simulations were performed for unlabeled 225Ac uniformly placed in the plasma and only the dose from alpha particles was considered. Absorbed doses to normal organs were determined for the liver, kidneys, bone, soft tissue, active marrow, and blood. Simulations were performed for the case when: (1) the daughters have unique biokinetics and (2) the daughters decay at the site of 225Ac. RESULTS When the daughters have unique biokinetics, the organs that receive the highest absorbed dose are the liver (male: 1466.6 mGy/MBq, female: 1885.7 mGy/MBq), bone (male: 293.6 mGy/MBq, female: 403.6 mGy/MBq) and kidneys (male: 260.8 mGy/MBq, female: 294.0 mGy/MBq). These doses were compared to the case when the daughters of 225Ac decay at the site of 225Ac. There was a 13.5% increase in kidney dose, a 0.8% decrease in liver dose, and <0.1% decrease in bone dose calculations when the daughters have unique biokinetics compared to assuming the daughters decay at the site of 225Ac. CONCLUSIONS The kidneys received a large dose estimate (260-295 mGy/MBq) as well as a considerable change in dose of +13.5% when the daughters have unique biokinetics compared to assuming the daughters decay at the site of 225Ac. Therefore, to accurately determine the kidney dose from unlabeled 225Ac in a human, the biokinetics of the daughter isotopes should be considered.
Collapse
Affiliation(s)
- Stephen Tronchin
- Department of Physics, The University of Adelaide, Adelaide, South Australia, Australia
| | - Jake Forster
- Department of Physics, The University of Adelaide, Adelaide, South Australia, Australia
- Medical Physics & Radiation Safety, South Australia Medical Imaging, Adelaide, South Australia, Australia
| | - Kevin Hickson
- Medical Physics & Radiation Safety, South Australia Medical Imaging, Adelaide, South Australia, Australia
- Allied Health & Human Performance, University of South Australia, Adelaide, South Australia, Australia
| | - Eva Bezak
- Department of Physics, The University of Adelaide, Adelaide, South Australia, Australia
- Allied Health & Human Performance, University of South Australia, Adelaide, South Australia, Australia
| |
Collapse
|
9
|
Kleynhans J, Ebenhan T, Cleeren F, Sathekge MM. Can current preclinical strategies for radiopharmaceutical development meet the needs of targeted alpha therapy? Eur J Nucl Med Mol Imaging 2024; 51:1965-1980. [PMID: 38676735 PMCID: PMC11139742 DOI: 10.1007/s00259-024-06719-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 04/15/2024] [Indexed: 04/29/2024]
Abstract
Preclinical studies are essential for effectively evaluating TAT radiopharmaceuticals. Given the current suboptimal supply chain of these radionuclides, animal studies must be refined to produce the most translatable TAT agents with the greatest clinical potential. Vector design is pivotal, emphasizing harmonious physical and biological characteristics among the vector, target, and radionuclide. The scarcity of alpha-emitting radionuclides remains a significant consideration. Actinium-225 and lead-212 appear as the most readily available radionuclides at this stage. Available animal models for researchers encompass xenografts, allografts, and PDX (patient-derived xenograft) models. Emerging strategies for imaging alpha-emitters are also briefly explored. Ultimately, preclinical research must address two critical aspects: (1) offering valuable insights into balancing safety and efficacy, and (2) providing guidance on the optimal dosing of the TAT agent.
Collapse
Affiliation(s)
- Janke Kleynhans
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, 3000, Belgium
| | - Thomas Ebenhan
- Department of Nuclear Medicine, University of Pretoria, and Steve Biko Academic Hospital, Pretoria, 0001, South Africa
- Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria, 0001, South Africa
| | - Frederik Cleeren
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, 3000, Belgium
| | - Mike Machaba Sathekge
- Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria, 0001, South Africa.
- Preclinical Imaging Facility, Nuclear Medicine Research Infrastructure, Pretoria, 0001, South Africa.
| |
Collapse
|
10
|
Munekane M, Fuchigami T, Ogawa K. Recent advances in the development of 225Ac- and 211At-labeled radioligands for radiotheranostics. ANAL SCI 2024; 40:803-826. [PMID: 38564087 PMCID: PMC11035452 DOI: 10.1007/s44211-024-00514-w] [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: 11/28/2023] [Accepted: 01/16/2024] [Indexed: 04/04/2024]
Abstract
Radiotheranostics utilizes a set of radioligands incorporating diagnostic or therapeutic radionuclides to achieve both diagnosis and therapy. Imaging probes using diagnostic radionuclides have been used for systemic cancer imaging. Integration of therapeutic radionuclides into the imaging probes serves as potent agents for radionuclide therapy. Among them, targeted alpha therapy (TAT) is a promising next-generation cancer therapy. The α-particles emitted by the radioligands used in TAT result in a high linear energy transfer over a short range, inducing substantial damage to nearby cells surrounding the binding site. Therefore, the key to successful cancer treatment with minimal side effects by TAT depends on the selective delivery of radioligands to their targets. Recently, TAT agents targeting biomolecules highly expressed in various cancer cells, such as sodium/iodide symporter, norepinephrine transporter, somatostatin receptor, αvβ3 integrin, prostate-specific membrane antigen, fibroblast-activation protein, and human epidermal growth factor receptor 2 have been developed and have made remarkable progress toward clinical application. In this review, we focus on two radionuclides, 225Ac and 211At, which are expected to have a wide range of applications in TAT. We also introduce recent fundamental and clinical studies of radiopharmaceuticals labeled with these radionuclides.
Collapse
Affiliation(s)
- Masayuki Munekane
- Graduate School of Medical Sciences, Kanazawa University, Kakuma-Machi, Kanazawa, Ishikawa, 920-1192, Japan
| | - Takeshi Fuchigami
- Graduate School of Medical Sciences, Kanazawa University, Kakuma-Machi, Kanazawa, Ishikawa, 920-1192, Japan.
| | - Kazuma Ogawa
- Graduate School of Medical Sciences, Kanazawa University, Kakuma-Machi, Kanazawa, Ishikawa, 920-1192, Japan.
- Institute for Frontier Science Initiative, Kanazawa University, Kakuma-Machi, Kanazawa, Ishikawa, 920-1192, Japan.
| |
Collapse
|
11
|
Salama MM, Aborehab NM, El Mahdy NM, Zayed A, Ezzat SM. Nanotechnology in leukemia: diagnosis, efficient-targeted drug delivery, and clinical trials. Eur J Med Res 2023; 28:566. [PMID: 38053150 DOI: 10.1186/s40001-023-01539-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 11/18/2023] [Indexed: 12/07/2023] Open
Abstract
Leukemia is a group of malignant disorders which affect the blood and blood-forming tissues in the bone marrow, lymphatic system, and spleen. Many types of leukemia exist; thus, their diagnosis and treatment are somewhat complicated. The use of conventional strategies for treatment such as chemotherapy and radiotherapy may develop many side effects and toxicity. Hence, modern research is concerned with the development of specific nano-formulations for targeted delivery of anti-leukemic drugs avoiding toxic effects on normal cells. Nanostructures can be applied not only in treatment but also in diagnosis. In this article, types of leukemia, its causes, diagnosis as well as conventional treatment of leukemia shall be reviewed. Then, the use of nanoparticles in diagnosis of leukemia and synthesis of nanocarriers for efficient delivery of anti-leukemia drugs being investigated in in vivo and clinical studies. Therefore, it may contribute to the discovery of novel and emerging nanoparticles for targeted treatment of leukemia with less side effects and toxicities.
Collapse
Affiliation(s)
- Maha M Salama
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo, 11562, Egypt
- Department of Pharmacognosy, Faculty of Pharmacy, The British University in Egypt, El Sherouk City, Suez Desert Road, Cairo, 11837, Egypt
| | - Nora M Aborehab
- Department of Biochemistry, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA), Giza, 12451, Egypt
| | - Nihal M El Mahdy
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA), Giza, 12451, Egypt
| | - Ahmed Zayed
- Department of Pharmacognosy, College of Pharmacy, Tanta University, Elguish Street (Medical Campus), Tanta, 31527, Egypt
| | - Shahira M Ezzat
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo, 11562, Egypt.
- Department of Pharmacognosy, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA), Giza, 12451, Egypt.
| |
Collapse
|
12
|
Ocampo-García B, Cruz-Nova P, Jiménez-Mancilla N, Luna-Gutiérrez M, Oros-Pantoja R, Lara-Almazán N, Pérez-Velasco D, Santos-Cuevas C, Ferro-Flores G. 225Ac-iPSMA-RGD for Alpha-Therapy Dual Targeting of Stromal/Tumor Cell PSMA and Integrins. Int J Mol Sci 2023; 24:16553. [PMID: 38068876 PMCID: PMC10705946 DOI: 10.3390/ijms242316553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/07/2023] [Accepted: 11/08/2023] [Indexed: 12/18/2023] Open
Abstract
Prostate-specific membrane antigens (PSMAs) are frequently overexpressed in both tumor stromal endothelial cells and malignant cells (stromal/tumor cells) of various cancers. The RGD (Arg-Gly-Asp) peptide sequence can specifically detect integrins involved in tumor angiogenesis. This study aimed to preclinically evaluate the cytotoxicity, biokinetics, dosimetry, and therapeutic efficacy of 225Ac-iPSMA-RGD to determine its potential as an improved radiopharmaceutical for alpha therapy compared with the 225Ac-iPSMA and 225Ac-RGD monomers. HEHA-HYNIC-iPSMA-RGD (iPSMA-RGD) was synthesized and characterized by FT-IR, UV-vis, and UPLC mass spectroscopy. The cytotoxicity of 225Ac-iPSMA-RGD was assessed in HCT116 colorectal cancer cells. Biodistribution, biokinetics, and therapeutic efficacy were evaluated in nude mice with induced HCT116 tumors. In vitro results showed increased DNA double-strand breaks through ROS generation, cell apoptosis, and death in HCT116 cells treated with 225Ac-iPSMA-RGD. The results also demonstrated in vivo cytotoxicity in cancer cells after treatment with 225Ac-iPSMA-RGD and biokinetic and dosimetric properties suitable for alpha therapy, delivering ablative radiation doses up to 237 Gy/3.7 kBq to HCT116 tumors in mice. Given the phenotype of HCT116 cancer cells, the results of this study warrant further dosimetric and clinical studies to determine the potential of 225Ac-iPSMA-RGD in the treatment of colorectal cancer.
Collapse
Affiliation(s)
- Blanca Ocampo-García
- Department of Radioactive Materials, Instituto Nacional de Investigaciones Nucleares, Ocoyoacac 52750, Mexico; (P.C.-N.); (M.L.-G.); (N.L.-A.); (G.F.-F.)
| | - Pedro Cruz-Nova
- Department of Radioactive Materials, Instituto Nacional de Investigaciones Nucleares, Ocoyoacac 52750, Mexico; (P.C.-N.); (M.L.-G.); (N.L.-A.); (G.F.-F.)
| | | | - Myrna Luna-Gutiérrez
- Department of Radioactive Materials, Instituto Nacional de Investigaciones Nucleares, Ocoyoacac 52750, Mexico; (P.C.-N.); (M.L.-G.); (N.L.-A.); (G.F.-F.)
| | | | - Nancy Lara-Almazán
- Department of Radioactive Materials, Instituto Nacional de Investigaciones Nucleares, Ocoyoacac 52750, Mexico; (P.C.-N.); (M.L.-G.); (N.L.-A.); (G.F.-F.)
| | - Diana Pérez-Velasco
- Faculty of Chemistry, Universidad Autónoma del Estado de México, Toluca 50180, Mexico;
| | - Clara Santos-Cuevas
- Department of Radioactive Materials, Instituto Nacional de Investigaciones Nucleares, Ocoyoacac 52750, Mexico; (P.C.-N.); (M.L.-G.); (N.L.-A.); (G.F.-F.)
| | - Guillermina Ferro-Flores
- Department of Radioactive Materials, Instituto Nacional de Investigaciones Nucleares, Ocoyoacac 52750, Mexico; (P.C.-N.); (M.L.-G.); (N.L.-A.); (G.F.-F.)
| |
Collapse
|
13
|
Cao Q, Wu X, Zhang Q, Gong J, Chen Y, You Y, Shen J, Qiang Y, Cao G. Mechanisms of action of the BCL-2 inhibitor venetoclax in multiple myeloma: a literature review. Front Pharmacol 2023; 14:1291920. [PMID: 38026941 PMCID: PMC10657905 DOI: 10.3389/fphar.2023.1291920] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 10/18/2023] [Indexed: 12/01/2023] Open
Abstract
Abnormal cellular apoptosis plays a pivotal role in the pathogenesis of Multiple Myeloma (MM). Over the years, BCL-2, a crucial anti-apoptotic protein, has garnered significant attention in MM therapeutic research. Venetoclax (VTC), a small-molecule targeted agent, effectively inhibits BCL-2, promoting the programmed death of cancerous cells. While VTC has been employed to treat various hematological malignancies, its particular efficacy in MM has showcased its potential for broader clinical applications. In this review, we delve into the intricacies of how VTC modulates apoptosis in MM cells by targeting BCL-2 and the overarching influence of the BCL-2 protein family in MM apoptosis regulation. Our findings highlight the nuanced interplay between VTC, BCL-2, and MM, offering insights that may pave the way for optimizing therapeutic strategies. Through this comprehensive analysis, we aim to lay a solid groundwork for future explorations into VTC's clinical applications and the profound effects of BCL-2 on cellular apoptosis.
Collapse
Affiliation(s)
- Qiang Cao
- Department of Earth Sciences, Kunming University of Science and Technology, Kunming, China
- Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xinyan Wu
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Qi Zhang
- Undergraduate Department, Taishan University, Taian, China
| | - Junling Gong
- School of Public Health, Nanchang University, Nanchang, China
| | - Yuquan Chen
- Institute of Medical Information/Library, Chinese Academy of Medical Sciences, Beijing, China
| | - Yanwei You
- Division of Sports Science & Physical Education, Tsinghua University, Beijing, China
| | - Jun Shen
- Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yi Qiang
- Department of Earth Sciences, Kunming University of Science and Technology, Kunming, China
| | - Guangzhu Cao
- Department of Earth Sciences, Kunming University of Science and Technology, Kunming, China
| |
Collapse
|
14
|
Hassan M, Bokhari TH, Lodhi NA, Khosa MK, Usman M. A review of recent advancements in Actinium-225 labeled compounds and biomolecules for therapeutic purposes. Chem Biol Drug Des 2023; 102:1276-1292. [PMID: 37715360 DOI: 10.1111/cbdd.14311] [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: 03/16/2022] [Revised: 06/03/2023] [Accepted: 07/17/2023] [Indexed: 09/17/2023]
Abstract
In nuclear medicine, cancers that cannot be cured or can only be treated partially by traditional techniques like surgery or chemotherapy are killed by ionizing radiation as a form of therapeutic treatment. Actinium-225 is an alpha-emitting radionuclide that is highly encouraging as a therapeutic approach and more promising for targeted alpha therapy (TAT). Actinium-225 is the best candidate for tumor cells treatment and has physical characteristics such as high (LET) linear energy transfer (150 keV per μm), half-life (t1/2 = 9.92d), and short ranges (400-100 μm) which prevent the damage of normal healthy tissues. The introduction of various new radiopharmaceuticals and radioisotopes has significantly assisted the advancement of nuclear medicine. Ac-225 radiopharmaceuticals continuously demonstrate their potential as targeted alpha therapeutics. 225 Ac-labeled radiopharmaceuticals have confirmed their importance in medical and clinical areas by introducing [225 Ac]Ac-PSMA-617, [225 Ac]Ac-DOTATOC, [225 Ac]Ac-DOTA-substance-P, reported significantly improved response in patients with prostate cancer, neuroendocrine, and glioma, respectively. The development of these radiopharmaceuticals required a suitable buffer, incubation time, optimal pH, and reaction temperature. There is a growing need to standardize quality control (QC) testing techniques such as radiochemical purity (RCP). This review aims to summarize the development of the Ac-225 labeled compounds and biomolecules. The current state of their reported resulting clinical applications is also summarized as well.
Collapse
Affiliation(s)
- Maria Hassan
- Department of Chemistry, Government College University, Faisalabad, Pakistan
| | | | - Nadeem Ahmed Lodhi
- Isotope Production Division, Pakistan institute of Nuclear Science & Technology (PINSTECH), Islamabad, Pakistan
| | | | - Muhammad Usman
- Department of Chemistry, Government College University, Faisalabad, Pakistan
| |
Collapse
|
15
|
Koniar H, Miller C, Rahmim A, Schaffer P, Uribe C. A GATE simulation study for dosimetry in cancer cell and micrometastasis from the 225Ac decay chain. EJNMMI Phys 2023; 10:46. [PMID: 37525027 PMCID: PMC10390455 DOI: 10.1186/s40658-023-00564-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 07/24/2023] [Indexed: 08/02/2023] Open
Abstract
BACKGROUND Radiopharmaceutical therapy (RPT) with alpha-emitting radionuclides has shown great promise in treating metastatic cancers. The successive emission of four alpha particles in the 225Ac decay chain leads to highly targeted and effective cancer cell death. Quantifying cellular dosimetry for 225Ac RPT is essential for predicting cell survival and therapeutic success. However, the leading assumption that all 225Ac progeny remain localized at their target sites likely overestimates the absorbed dose to cancer cells. To address limitations in existing semi-analytic approaches, this work evaluates S-values for 225Ac's progeny radionuclides with GATE Monte Carlo simulations. METHODS The cellular geometries considered were an individual cell (10 µm diameter with a nucleus of 8 µm diameter) and a cluster of cells (micrometastasis) with radionuclides localized in four subcellular regions: cell membrane, cytoplasm, nucleus, or whole cell. The absorbed dose to the cell nucleus was scored, and self- and cross-dose S-values were derived. We also evaluated the total absorbed dose with various degrees of radiopharmaceutical internalization and retention of the progeny radionuclides 221Fr (t1/2 = 4.80 m) and 213Bi (t1/2 = 45.6 m). RESULTS For the cumulative 225Ac decay chain, our self- and cross-dose nuclear S-values were both in good agreement with S-values published by MIRDcell, with per cent differences ranging from - 2.7 to - 8.7% for the various radionuclide source locations. Source location had greater effects on self-dose S-values than the intercellular cross-dose S-values. Cumulative 225Ac decay chain self-dose S-values increased from 0.167 to 0.364 GyBq-1 s-1 with radionuclide internalization from the cell surface into the cell. When progeny migration from the target site was modelled, the cumulative self-dose S-values to the cell nucleus decreased by up to 71% and 21% for 221Fr and 213Bi retention, respectively. CONCLUSIONS Our GATE Monte Carlo simulations resulted in cellular S-values in agreement with existing MIRD S-values for the alpha-emitting radionuclides in the 225Ac decay chain. To obtain accurate absorbed dose estimates in 225Ac studies, accurate understanding of daughter migration is critical for optimized injected activities. Future work will investigate other novel preclinical alpha-emitting radionuclides to evaluate therapeutic potency and explore realistic cellular geometries corresponding to targeted cancer cell lines.
Collapse
Affiliation(s)
- Helena Koniar
- Life Sciences Division, TRIUMF, Vancouver, BC, Canada.
- Department of Physics and Astronomy, University of British Columbia, Vancouver, BC, Canada.
| | - Cassandra Miller
- Department of Physics and Astronomy, University of British Columbia, Vancouver, BC, Canada
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Arman Rahmim
- Department of Physics and Astronomy, 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
| | - Paul Schaffer
- Life Sciences Division, TRIUMF, Vancouver, BC, Canada
- Department of Radiology, University of British Columbia, Vancouver, BC, Canada
- Department of Chemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Carlos Uribe
- Department of Radiology, University of British Columbia, Vancouver, BC, Canada
- Functional Imaging, BC Cancer, Vancouver, BC, Canada
| |
Collapse
|
16
|
Feuerecker B, Kratochwil C, Ahmadzadehfar H, Morgenstern A, Eiber M, Herrmann K, Pomykala KL. Clinical Translation of Targeted α-Therapy: An Evolution or a Revolution? J Nucl Med 2023; 64:685-692. [PMID: 37055224 DOI: 10.2967/jnumed.122.265353] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/10/2023] [Indexed: 04/15/2023] Open
Abstract
The field of radioligand therapy has advanced greatly in recent years, driven largely by β-emitting therapies targeting somatostatin receptor-expressing tumors and the prostate-specific membrane antigen. Now, more clinical trials are under way to evaluate α-emitting targeted therapies as potential next-generation theranostics with even higher efficacy due to their high linear energy and short range in human tissues. In this review, we summarize the important studies ranging from the first Food and Drug Administration-approved α-therapy, 223Ra-dichloride, for treatment of bone metastases in castration-resistant prostate cancer, including concepts in clinical translation such as targeted α-peptide receptor radiotherapy and 225Ac-PSMA-617 for treatment of prostate cancer, innovative therapeutic models evaluating new targets, and combination therapies. Targeted α-therapy is one of the most promising fields in novel targeted cancer therapy, with several early- and late-stage clinical trials for neuroendocrine tumors and metastatic prostate cancer already in progress, along with significant interest and investment in additional early-phase studies. Together, these studies will help us understand the short- and long-term toxicity of targeted α-therapy and potentially identify suitable therapeutic combination partners.
Collapse
Affiliation(s)
- Benedikt Feuerecker
- Department of Nuclear Medicine, Technische Universität München, München, Germany
- Department of Radiology, Technische Universität München, München, Germany
- German Cancer Consortium, partner sites München, Heidelberg, and Essen, Germany
- Department of Radiology, University Hospital, LMU München, München, Germany
| | - Clemens Kratochwil
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Hojjat Ahmadzadehfar
- Department of Nuclear Medicine, Klinikum Westfalen-Knappschaftskrankenhaus, Dortmund, Germany
| | | | - Matthias Eiber
- Department of Nuclear Medicine, Technische Universität München, München, Germany
| | - Ken Herrmann
- German Cancer Consortium, partner sites München, Heidelberg, and Essen, Germany
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany; and
| | - Kelsey L Pomykala
- Institute for Artificial Intelligence in Medicine, University Hospital Essen, Essen, Germany
| |
Collapse
|
17
|
Rubira L, Deshayes E, Santoro L, Kotzki PO, Fersing C. 225Ac-Labeled Somatostatin Analogs in the Management of Neuroendocrine Tumors: From Radiochemistry to Clinic. Pharmaceutics 2023; 15:1051. [PMID: 37111537 PMCID: PMC10146019 DOI: 10.3390/pharmaceutics15041051] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/18/2023] [Accepted: 03/22/2023] [Indexed: 04/29/2023] Open
Abstract
The widespread use of peptide receptor radionuclide therapy (PRRT) represents a major therapeutic breakthrough in nuclear medicine, particularly since the introduction of 177Lu-radiolabeled somatostatin analogs. These radiopharmaceuticals have especially improved progression-free survival and quality of life in patients with inoperable metastatic gastroenteropancreatic neuroendocrine tumors expressing somatostatin receptors. In the case of aggressive or resistant disease, the use of somatostatin derivatives radiolabeled with an alpha-emitter could provide a promising alternative. Among the currently available alpha-emitting radioelements, actinium-225 has emerged as the most suitable candidate, especially regarding its physical and radiochemical properties. Nevertheless, preclinical and clinical studies on these radiopharmaceuticals are still few and heterogeneous, despite the growing momentum for their future use on a larger scale. In this context, this report provides a comprehensive and extensive overview of the development of 225Ac-labeled somatostatin analogs; particular emphasis is placed on the challenges associated with the production of 225Ac, its physical and radiochemical properties, as well as the place of 225Ac-DOTATOC and 225Ac-DOTATATE in the management of patients with advanced metastatic neuroendocrine tumors.
Collapse
Affiliation(s)
- Léa Rubira
- Nuclear Medicine Department, Institut Régional du Cancer de Montpellier (ICM), University Montpellier, 34090 Montpellier, France
| | - Emmanuel Deshayes
- Nuclear Medicine Department, Institut Régional du Cancer de Montpellier (ICM), University Montpellier, 34090 Montpellier, France
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, University Montpellier, Institut Régional du Cancer de Montpellier (ICM), 34298 Montpellier, France
| | - Lore Santoro
- Nuclear Medicine Department, Institut Régional du Cancer de Montpellier (ICM), University Montpellier, 34090 Montpellier, France
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, University Montpellier, Institut Régional du Cancer de Montpellier (ICM), 34298 Montpellier, France
| | - Pierre Olivier Kotzki
- Nuclear Medicine Department, Institut Régional du Cancer de Montpellier (ICM), University Montpellier, 34090 Montpellier, France
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, University Montpellier, Institut Régional du Cancer de Montpellier (ICM), 34298 Montpellier, France
| | - Cyril Fersing
- Nuclear Medicine Department, Institut Régional du Cancer de Montpellier (ICM), University Montpellier, 34090 Montpellier, France
- IBMM, University Montpellier, CNRS, ENSCM, 34293 Montpellier, France
| |
Collapse
|
18
|
Ciotti G, Marconi G, Sperotto A, Giannini MB, Gottardi M, Martinelli G. Biological therapy in elderly patients with acute myeloid leukemia. Expert Opin Biol Ther 2023; 23:175-194. [PMID: 36715330 DOI: 10.1080/14712598.2023.2174015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
INTRODUCTION The introduction of target molecules and immunological therapies is changing the treatment landscape of acute myeloid leukemia (AML). AREAS COVERED We recapitulate the biological therapies that can be employed in the treatment of elderly patients with AML. Alongside small molecules inhibitors that target specific gene mutations, antibodies, tumor microenvironment modulators, and cellular therapies are being developed for the cure of the disease. Here, we report the biological activities, the efficacy and toxicities of humanized antibodies and antibody-drug conjugates that targets surface antigens as CD33 (gemtuzumab ozogamicine) or CD123 (pivekimab sunirine). We further explore mechanisms and effectiveness of medications that modify the microenvironment, such as glasdegib, or that harness the immune system against leukemia, such as CD47 antibody magrolimab, PD1/PDL1 inhibitors pembrolizumab and nivolumab, TIM3 inhibitor sabatolimab, T-cell and NK-cell engagers. Cellular therapies are considered, even if a large trial is still pending for the feasibility of the approach. In this scenario, a brief overview of the mechanism of action of target agents is provided, particularly with respect to their biological mechanisms. EXPERT OPINION Overall, this therapeutic armamentarium will constitute the basis for multimodal and personalized combinations that, in the idea of precision medicine, will enormously benefit elderly AML patients.
Collapse
Affiliation(s)
- Giulia Ciotti
- Onco Hematology, Department of Oncology, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Giovanni Marconi
- IRCCS Istituto Romagnolo Per Lo Studio Dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Alessandra Sperotto
- Onco Hematology, Department of Oncology, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Maria B Giannini
- IRCCS Istituto Romagnolo Per Lo Studio Dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Michele Gottardi
- Onco Hematology, Department of Oncology, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Giovanni Martinelli
- IRCCS Istituto Romagnolo Per Lo Studio Dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| |
Collapse
|
19
|
Wharton L, Zhang C, Zeisler J, Rodríguez-Rodríguez C, Osooly M, Radchenko V, Yang H, Lin KS, Bénard F, Schaffer P, Orvig C. H 3TPAN-Triazole-Bn-NH 2: Tripicolinate Clicked-Bifunctional Chelate for [ 225Ac]/[ 111In] Theranostics. Bioconjug Chem 2022; 33:2381-2397. [PMID: 36378809 DOI: 10.1021/acs.bioconjchem.2c00465] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
A new, high-denticity, bifunctional ligand─H3TPAN-triazole-Bn-NH2─has been synthesized and studied in complexation with [225Ac]Ac3+ and [111In]In3+ for radiopharmaceutical applications. The bifunctional chelator is readily synthesized, using a high-yielding four-step prep, which is highly adaptable and allows for straightforward incorporation of different covalent linkers using CuI-catalyzed alkyne-azide cycloaddition (click) chemistry. Nuclear magnetic resonance (NMR) studies of H3TPAN-triazole-Bn-NH2 with La3+ and In3+ metal ions show the formation of a single, asymmetric complex with each ion in solution, corroborated by density functional theory (DFT) calculations. Radiolabeling studies with [225Ac]Ac3+ and [111In]In3+ showed highly effective complexation, achieving quantitative radiochemical conversions at low ligand concentrations (<10-6 M) under mild conditions (RT, 10 min), which is further accompanied by high stability in human serum. The bioconjugate─H3TPAN-triazole-Bn-Aoc-Pip-Nle-CycMSHhex─was prepared for targeting of MC1R-positive tumors, and the corresponding 111In-radiolabeled tracer was studied in vivo. SPECT/CT and biodistribution studies in C57BL/6J mice bearing B16-F10 tumors were performed, with the radiotracer showing good in vivo stability; tumor uptake was achieved. This work highlights a new promising and versatile bifunctional chelator, easily prepared and encouraging for 225Ac/111In theranostics.
Collapse
Affiliation(s)
- Luke Wharton
- Medicinal Inorganic Chemistry Group, Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, British Columbia V6T 1Z1, Canada.,Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, British Columbia V6T 2A3, Canada
| | - Chengcheng Zhang
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, British Columbia V5Z 1L3, Canada
| | - Jutta Zeisler
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, British Columbia V5Z 1L3, Canada
| | - Cristina Rodríguez-Rodríguez
- Faculty of Pharmaceutical Sciences, University of British Columbia, 2405 Wesbrook Mall, Vancouver, British Columbia V6T 1Z3, Canada.,Department of Physics and Astronomy, University of British Columbia, 6224 Agronomy Road, Vancouver, British Columbia V6T 1Z1, Canada
| | - Maryam Osooly
- Faculty of Pharmaceutical Sciences, University of British Columbia, 2405 Wesbrook Mall, Vancouver, British Columbia V6T 1Z3, Canada
| | - Valery Radchenko
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, British Columbia V6T 2A3, Canada.,Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, British Columbia V6T 1Z1, Canada
| | - Hua Yang
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, British Columbia V6T 2A3, Canada.,Department of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - Kuo-Shyan Lin
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, British Columbia V5Z 1L3, Canada.,Department of Radiology, University of British Columbia, Vancouver, British Columbia V5Z 1M9, Canada
| | - François Bénard
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, British Columbia V5Z 1L3, Canada.,Department of Radiology, University of British Columbia, Vancouver, British Columbia V5Z 1M9, Canada
| | - Paul Schaffer
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, British Columbia V6T 2A3, Canada.,Department of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada.,Department of Radiology, University of British Columbia, Vancouver, British Columbia V5Z 1M9, Canada
| | - Chris Orvig
- Medicinal Inorganic Chemistry Group, Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, British Columbia V6T 1Z1, Canada
| |
Collapse
|
20
|
Allen KJH, Jiao R, Li J, Beckford-Vera DR, Dadachova E. In Vitro and In Vivo Characterization of 89Zirconium-Labeled Lintuzumab Molecule. Molecules 2022; 27:molecules27196589. [PMID: 36235126 PMCID: PMC9571394 DOI: 10.3390/molecules27196589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/27/2022] [Accepted: 09/30/2022] [Indexed: 11/13/2022] Open
Abstract
Objective: Positron emission tomography (PET) imaging is a powerful non-invasive method to determine the in vivo behavior of biomolecules. Determining biodistribution and pharmacokinetic (PK) properties of targeted therapeutics can enable a better understanding of in vivo drug mechanisms such as tumor uptake, off target accumulation and clearance. Zirconium-89 (89Zr) is a readily available tetravalent PET-enabling radiometal that has been used to evaluate the biodistribution and PK of monoclonal antibodies. In the current study, we performed in vitro and in vivo characterization of 89Zr-lintuzumab, a radiolabeled anti-CD33 antibody, as a model to evaluate the in vivo binding properties in preclinical models of AML. Methods: Lintuzumab was conjugated to p-SCN-Bn-deferoxamine (DFO) and labeled with 89Zr using a 5:1 µCi:µg specific activity at 37 °C for 1h. The biological activity of 89Zr-lintuzumab was evaluated in a panel of CD33 positive cells using flow cytometry. Fox Chase SCID mice were injected with 2 × 106 OCI-AML3 cells into the right flank. After 12 days, a cohort of mice (n = 4) were injected with 89Zr-lintuzumab via tail vein. PET/CT scans of mice were acquired on days 1, 2, 3 and 7 post 89Zr-lintuzumab injection. To demonstrate 89Zr-lintuzumab specific binding to CD33 expressing tumors in vivo, a blocking study was performed. This cohort of mice (n = 4) was injected with native lintuzumab and 24 h later 89Zr-lintuzumab was administered. This group was imaged 3 and 7 days after injection of 89Zr-lintuzumab. A full ex vivo biodistribution study on both cohorts was performed on day 7. The results from the PET image and ex vivo biodistribution studies were compared. Results: Lintuzumab was successfully radiolabeled with 89Zr resulting in a 99% radiochemical yield. The 89Zr-lintuzumab radioconjugate specifically binds CD33 positive cells in a similar manner to native lintuzumab as observed by flow cytometry. PET imaging revealed high accumulation of 89Zr-lintuzumab in OCI-AML3 tumors within 24h post-injection of the radioconjugate. The 89Zr-lintuzumab high tumor uptake remains for up to 7 days. Tumor analysis of the PET data using volume of interest (VOI) showed significant blocking of 89Zr-lintuzumab in the group pre-treated with native lintuzumab (pre-blocked group), thus indicating specific targeting of CD33 on OCI-AML3 cells in vivo. The tumor uptake findings from the PET imaging study are in agreement with those from the ex vivo biodistribution results. Conclusions: PET imaging of 89Zr-lintuzumab shows high specific uptake in CD33 positive human OCI-AML3 tumors. The results from the image study agree with the observations from the ex vivo biodistribution study. Our findings collectively suggest that PET imaging using 89Zr-lintuzumab could be a powerful drug development tool to evaluate binding properties of anti-CD33 monoclonal antibodies in preclinical cancer models.
Collapse
Affiliation(s)
- Kevin J. H. Allen
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Rubin Jiao
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Jason Li
- Actinium Pharmaceuticals, New York, NY 10016, USA
| | | | - Ekaterina Dadachova
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
- Correspondence: ; Tel.: +1-306-966-5163
| |
Collapse
|
21
|
Wharton L, Jaraquemada-Peláez MDG, Zhang C, Zeisler J, Rodríguez-Rodríguez C, Osooly M, Radchenko V, Yang H, Lin KS, Bénard F, Schaffer P, Orvig C. H 4picoopa─Robust Chelate for 225Ac/ 111In Theranostics. Bioconjug Chem 2022; 33:1900-1921. [DOI: 10.1021/acs.bioconjchem.2c00364] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Luke Wharton
- Medicinal Inorganic Chemistry Group, Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, BC V6T 1Z1, Canada
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada
| | | | - Chengcheng Zhang
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada
| | - Jutta Zeisler
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada
| | - Cristina Rodríguez-Rodríguez
- Faculty of Pharmaceutical Sciences, University of British Columbia, 2405 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada
- Department of Physics and Astronomy, University of British Columbia, 6224 Agronomy Road, Vancouver, BC V6T 1Z1, Canada
| | - Maryam Osooly
- Faculty of Pharmaceutical Sciences, University of British Columbia, 2405 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada
| | - Valery Radchenko
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, BC V6T 1Z1, Canada
| | - Hua Yang
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada
- Department of Chemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - Kuo-Shyan Lin
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada
- Department of Radiology, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - François Bénard
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada
- Department of Radiology, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Paul Schaffer
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada
- Department of Chemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
- Department of Radiology, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Chris Orvig
- Medicinal Inorganic Chemistry Group, Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, BC V6T 1Z1, Canada
| |
Collapse
|
22
|
Walter RB. Where do we stand with radioimmunotherapy for acute myeloid leukemia? Expert Opin Biol Ther 2022; 22:555-561. [PMID: 35350938 PMCID: PMC9090441 DOI: 10.1080/14712598.2022.2060735] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/29/2022] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Despite the approval of several new drugs, deaths from acute myeloid leukemia (AML) remain common. Because of well-defined cell surface antigens, easy accessibility, and radiosensitivity of leukemia cells, there is long-standing interest in radiolabeled antibodies (radioimmunotherapy [RIT]) to complement or replace existing treatments and improve outcomes in AML. AREAS COVERED Targeting primarily CD33, CD45, or CD66, early RIT efforts have focused on β-emitters, including iodine-131 (131I) and yttrium-90, mostly to intensify conditioning therapy before allogeneic hematopoietic cell transplantation (HCT). An 131I-labeled CD45 antibody (Iomab-B [apamistamab-I131]) is currently studied in the registration-type phase 3 SIERRA trial (NCT02665065) for this purpose. Of growing interest as therapeutic payloads are α-particle emitting radionuclides such as actinium-225 (225Ac) or astatine-211 (211At) since they deliver substantially higher decay energies over a much shorter distance than β-emitters, rendering them more suitable for precise, potent, and efficient target cell killing while minimizing toxicity to surrounding bystander cells, possibly allowing use outside of HCT. Clinical efforts with 211At-labeled CD45 antibodies and 225Ac-labeled CD33 antibodies (e.g. 225Ac-lintuzumab [Actimab-A]) are ongoing. EXPERT OPINION A first anti-AML RIT may soon become available. This might propel further work to develop RIT-based treatments for AML, with many such efforts already ongoing.
Collapse
Affiliation(s)
- Roland B Walter
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Medicine, Division of Hematology, University of Washington, Seattle, WA, USA
- Department of Laboratory Medicine & Pathology, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| |
Collapse
|
23
|
Development and Functional Characterization of a Versatile Radio-/Immunotheranostic Tool for Prostate Cancer Management. Cancers (Basel) 2022; 14:cancers14081996. [PMID: 35454902 PMCID: PMC9027777 DOI: 10.3390/cancers14081996] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 04/08/2022] [Accepted: 04/11/2022] [Indexed: 12/12/2022] Open
Abstract
Simple Summary In previous studies, we described a modular Chimeric Antigen Receptor (CAR) T cell platform which we termed UniCAR. In contrast to conventional CARs, the interaction of UniCAR T cells does not occur directly between the CAR T cell and the tumor cell but is mediated via bispecific adaptor molecules so-called target modules (TMs). Here we present the development and functional characterization of a novel IgG4-based TM, directed to the tumor-associated antigen (TAA) prostate stem cell antigen (PSCA), which is overexpressed in prostate cancer (PCa). We show that this anti-PSCA IgG4-TM cannot only be used for (i) redirection of UniCAR T cells to PCa cells but also for (ii) positron emission tomography (PET) imaging, and (iii) alpha particle-based endoradiotherapy. For radiolabeling, the anti-PSCA IgG4-TM was conjugated with the chelator DOTAGA. PET imaging was performed using the 64Cu-labeled anti-PSCA IgG4-TM. According to PET imaging, the anti-PSCA IgG4-TM accumulates with high contrast in the PSCA-positive tumors of experimental mice without visible uptake in other organs. For endoradiotherapy the anti-PSCA IgG4-TM-DOTAGA conjugate was labeled with 225Ac3+. Targeted alpha therapy resulted in tumor control over 60 days after a single injection of the 225Ac-labeled TM. The favorable pharmacological profile of the anti-PSCA IgG4-TM, and its usage for (i) imaging, (ii) targeted alpha therapy, and (iii) UniCAR T cell immunotherapy underlines the promising radio-/immunotheranostic capabilities for the diagnostic imaging and treatment of PCa. Abstract Due to its overexpression on the surface of prostate cancer (PCa) cells, the prostate stem cell antigen (PSCA) is a potential target for PCa diagnosis and therapy. Here we describe the development and functional characterization of a novel IgG4-based anti-PSCA antibody (Ab) derivative (anti-PSCA IgG4-TM) that is conjugated with the chelator DOTAGA. The anti-PSCA IgG4-TM represents a multimodal immunotheranostic compound that can be used (i) as a target module (TM) for UniCAR T cell-based immunotherapy, (ii) for diagnostic positron emission tomography (PET) imaging, and (iii) targeted alpha therapy. Cross-linkage of UniCAR T cells and PSCA-positive tumor cells via the anti-PSCA IgG4-TM results in efficient tumor cell lysis both in vitro and in vivo. After radiolabeling with 64Cu2+, the anti-PSCA IgG4-TM was successfully applied for high contrast PET imaging. In a PCa mouse model, it showed specific accumulation in PSCA-expressing tumors, while no uptake in other organs was observed. Additionally, the DOTAGA-conjugated anti-PSCA IgG4-TM was radiolabeled with 225Ac3+ and applied for targeted alpha therapy. A single injection of the 225Ac-labeled anti-PSCA IgG4-TM was able to significantly control tumor growth in experimental mice. Overall, the novel anti-PSCA IgG4-TM represents an attractive first member of a novel group of radio-/immunotheranostics that allows diagnostic imaging, endoradiotherapy, and CAR T cell immunotherapy.
Collapse
|
24
|
Andreozzi F, Massaro F, Wittnebel S, Spilleboudt C, Lewalle P, Salaroli A. New Perspectives in Treating Acute Myeloid Leukemia: Driving towards a Patient-Tailored Strategy. Int J Mol Sci 2022; 23:3887. [PMID: 35409248 PMCID: PMC8999556 DOI: 10.3390/ijms23073887] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/27/2022] [Accepted: 03/28/2022] [Indexed: 12/16/2022] Open
Abstract
For decades, intensive chemotherapy (IC) has been considered the best therapeutic option for treating acute myeloid leukemia (AML), with no curative option available for patients who are not eligible for IC or who have had failed IC. Over the last few years, several new drugs have enriched the therapeutic arsenal of AML treatment for both fit and unfit patients, raising new opportunities but also new challenges. These include the already approved venetoclax, the IDH1/2 inhibitors enasidenib and ivosidenib, gemtuzumab ozogamicin, the liposomal daunorubicin/cytarabine formulation CPX-351, and oral azacitidine. Venetoclax, an anti BCL2-inhibitor, in combination with hypomethylating agents (HMAs), has markedly improved the management of unfit and elderly patients from the perspective of improved quality of life and better survival. Venetoclax is currently under investigation in combination with other old and new drugs in early phase trials. Recently developed drugs with different mechanisms of action and new technologies that have already been investigated in other settings (BiTE and CAR-T cells) are currently being explored in AML, and ongoing trials should determine promising agents, more synergic combinations, and better treatment strategies. Access to new drugs and inclusion in clinical trials should be strongly encouraged to provide scientific evidence and to define the future standard of treatment in AML.
Collapse
Affiliation(s)
- Fabio Andreozzi
- Hematology Department, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Bruxelles, Belgium; (F.M.); (S.W.); (C.S.); (P.L.); (A.S.)
| | - Fulvio Massaro
- Hematology Department, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Bruxelles, Belgium; (F.M.); (S.W.); (C.S.); (P.L.); (A.S.)
- PhD Program in Clinical and Experimental Medicine, University of Modena and Reggio Emilia, 41121 Modena, Italy
| | - Sebastian Wittnebel
- Hematology Department, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Bruxelles, Belgium; (F.M.); (S.W.); (C.S.); (P.L.); (A.S.)
| | - Chloé Spilleboudt
- Hematology Department, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Bruxelles, Belgium; (F.M.); (S.W.); (C.S.); (P.L.); (A.S.)
| | - Philippe Lewalle
- Hematology Department, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Bruxelles, Belgium; (F.M.); (S.W.); (C.S.); (P.L.); (A.S.)
| | - Adriano Salaroli
- Hematology Department, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Bruxelles, Belgium; (F.M.); (S.W.); (C.S.); (P.L.); (A.S.)
| |
Collapse
|
25
|
Lim J, Sari-Ak D, Bagga T. Siglecs as Therapeutic Targets in Cancer. BIOLOGY 2021; 10:1178. [PMID: 34827170 PMCID: PMC8615218 DOI: 10.3390/biology10111178] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/03/2021] [Accepted: 11/08/2021] [Indexed: 02/06/2023]
Abstract
Hypersialylation is a common post-translational modification of protein and lipids found on cancer cell surfaces, which participate in cell-cell interactions and in the regulation of immune responses. Sialic acids are a family of nine-carbon α-keto acids found at the outermost ends of glycans attached to cell surfaces. Given their locations on cell surfaces, tumor cells aberrantly overexpress sialic acids, which are recognized by Siglec receptors found on immune cells to mediate broad immunomodulatory signaling. Enhanced sialylation exposed on cancer cell surfaces is exemplified as "self-associated molecular pattern" (SAMP), which tricks Siglec receptors found on leukocytes to greatly down-regulate immune responsiveness, leading to tumor growth. In this review, we focused on all 15 human Siglecs (including Siglec XII), many of which still remain understudied. We also highlighted strategies that disrupt the course of Siglec-sialic acid interactions, such as antibody-based therapies and sialic acid mimetics leading to tumor cell depletion. Herein, we introduced the central roles of Siglecs in mediating pro-tumor immunity and discussed strategies that target these receptors, which could benefit improved cancer immunotherapy.
Collapse
Affiliation(s)
- Jackwee Lim
- Singapore Immunology Network, A*STAR, 8a Biomedical Grove, Singapore 138648, Singapore;
| | - Duygu Sari-Ak
- Department of Medical Biology, School of Medicine, University of Health Sciences, Istanbul 34668, Turkey;
| | - Tanaya Bagga
- Singapore Immunology Network, A*STAR, 8a Biomedical Grove, Singapore 138648, Singapore;
| |
Collapse
|
26
|
Evaluation of Aminopolycarboxylate Chelators for Whole-Body Clearance of Free 225Ac: A Feasibility Study to Reduce Unexpected Radiation Exposure during Targeted Alpha Therapy. Pharmaceutics 2021; 13:pharmaceutics13101706. [PMID: 34683999 PMCID: PMC8540721 DOI: 10.3390/pharmaceutics13101706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/06/2021] [Accepted: 10/12/2021] [Indexed: 11/24/2022] Open
Abstract
Actinium-225 (225Ac) is a promising radionuclide used in targeted alpha therapy (TAT). Although 225Ac labeling of bifunctional chelating ligands is effective, previous in vivo studies reported that free 225Ac can be released from the drugs and that such free 225Ac is predominantly accumulated in the liver and could cause unexpected toxicity. To accelerate the clinical development of 225Ac TAT with a variety of drugs, preparing methods to deal with any unexpected toxicity would be valuable. The aim of this study was to evaluate the feasibility of various chelators for reducing and excreting free 225Ac and compare their chemical structures. Nine candidate chelators (D-penicillamine, dimercaprol, Ca-DTPA, Ca-EDTA, CyDTA, GEDTA TTHA, Ca-TTHA, and DO3A) were evaluated in vitro and in vivo. The biodistribution and dosimetry of free 225Ac were examined in mice before an in vivo chelating study. The liver exhibited pronounced 225Ac uptake, with an estimated human absorbed dose of 4.76 SvRBE5/MBq. Aminopolycarboxylate chelators with five and six carboxylic groups, Ca-DTPA and Ca-TTHA, significantly reduced 225Ac retention in the liver (22% and 30%, respectively). Significant 225Ac reductions were observed in the heart and remainder of the body with both Ca-DTPA and Ca-TTHA, and in the lung, kidney, and spleen with Ca-TTHA. In vitro interaction analysis supported the in vivo reduction ability of Ca-DTPA and Ca-TTHA. In conclusion, aminopolycarboxylate chelators with five and six carboxylic groups, Ca-DTPA and Ca-TTHA, were effective for whole-body clearance of free 225Ac. This feasibility study provides useful information for reducing undesirable radiation exposure from free 225Ac.
Collapse
|
27
|
Ingham A, Kostelnik TI, McNeil BL, Patrick BO, Choudhary N, Jaraquemada-Peláez MDG, Orvig C. Getting a lead on Pb 2+-amide chelators for 203/212Pb radiopharmaceuticals. Dalton Trans 2021; 50:11579-11595. [PMID: 34352061 DOI: 10.1039/d1dt01653a] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Amide-based chelators DTPAm, EGTAm and ampam were synthesized to investigate which chelator most ideally coordinates [nat/203Pb]Pb2+ ions for potential radiopharmaceutical applications. 1H NMR spectroscopy was used to study each metal-ligand complex in the solution state. The 1H NMR spectrum of [Pb(DTPAm)]2+ revealed minimal isomerization and fluxional behaviour compared to [Pb(EGTAm)]2+ and [Pb(ampam)]2+, both of which showed fewer spectral changes indicative of less static behaviour. The solid-state coordination properties of each complex were also examined from single crystal structures that were studied by X-ray diffraction (XRD). In the solid-state, octadentate DTPAm coordinated Pb2+ to form an eight-coordinate hemidirected complex; octadentate EGTAm coordinated Pb2+ forming a ten-coordinate holodirected complex with a bidentate NO3- ion also coordinated to the metal centre; decadentate ampam completely encapsulated the Pb2+ ion to form a ten-coordinate holodirected complex with a C2 axis of symmetry. Potentiometric titrations were carried out to assess the thermodynamic stability of each metal-ligand complex. The pM values obtained for [Pb(DTPAm)]2+, [Pb(EGTAm)]2+ and [Pb(ampam)]2+ were 9.7, 7.2 and 10.2, respectively. The affinity of each chelator for Pb2+ ions was tested by [203Pb]Pb2+ radiolabeling studies to evaluate their prospects as chelators for [203/212Pb]Pb2+-based radiopharmaceuticals. DTPAm radiolabeled [203Pb]Pb2+ ions achieving molar activities as high as 3.5 MBq μmol-1 within 15 minutes, at 25 °C, whereas EGTAm and ampam produced lower molar activities of 0.25 MBq μmol-1 within 30 minutes, at 37 °C. EGTAm and ampam were therefore deemed unsuitable for [203/212Pb]Pb2+-based radiopharmaceutical applications, while DTPAm warrants further studies.
Collapse
Affiliation(s)
- Aidan Ingham
- Medicinal Inorganic Chemistry Group, Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, British Columbia V6 T 1Z1, Canada.
| | | | | | | | | | | | | |
Collapse
|
28
|
Eychenne R, Chérel M, Haddad F, Guérard F, Gestin JF. Overview of the Most Promising Radionuclides for Targeted Alpha Therapy: The "Hopeful Eight". Pharmaceutics 2021; 13:pharmaceutics13060906. [PMID: 34207408 PMCID: PMC8234975 DOI: 10.3390/pharmaceutics13060906] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/02/2021] [Accepted: 06/08/2021] [Indexed: 12/11/2022] Open
Abstract
Among all existing radionuclides, only a few are of interest for therapeutic applications and more specifically for targeted alpha therapy (TAT). From this selection, actinium-225, astatine-211, bismuth-212, bismuth-213, lead-212, radium-223, terbium-149 and thorium-227 are considered as the most suitable. Despite common general features, they all have their own physical characteristics that make them singular and so promising for TAT. These radionuclides were largely studied over the last two decades, leading to a better knowledge of their production process and chemical behavior, allowing for an increasing number of biological evaluations. The aim of this review is to summarize the main properties of these eight chosen radionuclides. An overview from their availability to the resulting clinical studies, by way of chemical design and preclinical studies is discussed.
Collapse
Affiliation(s)
- Romain Eychenne
- Groupement d’Intérêt Public ARRONAX, 1 Rue Aronnax, F-44817 Saint-Herblain, France;
- Université de Nantes, Inserm, CNRS, Centre de Recherche en Cancérologie et Immunologie Nantes—Angers (CRCINA)—UMR 1232, ERL 6001, F-44000 Nantes, France; (M.C.); (F.G.)
- Correspondence: (R.E.); (J.-F.G.)
| | - Michel Chérel
- Université de Nantes, Inserm, CNRS, Centre de Recherche en Cancérologie et Immunologie Nantes—Angers (CRCINA)—UMR 1232, ERL 6001, F-44000 Nantes, France; (M.C.); (F.G.)
| | - Férid Haddad
- Groupement d’Intérêt Public ARRONAX, 1 Rue Aronnax, F-44817 Saint-Herblain, France;
- Laboratoire Subatech, UMR 6457, Université de Nantes, IMT Atlantique, CNRS, Subatech, F-44000 Nantes, France
| | - François Guérard
- Université de Nantes, Inserm, CNRS, Centre de Recherche en Cancérologie et Immunologie Nantes—Angers (CRCINA)—UMR 1232, ERL 6001, F-44000 Nantes, France; (M.C.); (F.G.)
| | - Jean-François Gestin
- Université de Nantes, Inserm, CNRS, Centre de Recherche en Cancérologie et Immunologie Nantes—Angers (CRCINA)—UMR 1232, ERL 6001, F-44000 Nantes, France; (M.C.); (F.G.)
- Correspondence: (R.E.); (J.-F.G.)
| |
Collapse
|