1
|
Sancho L, Roteta A, Torres I, de Arcocha M, Ramos R, Domínguez ML, Rosales JJ, Prieto E, Quincoces G. State of the art and future perspectives of new radionuclides in Nuclear Medicine. Part II. Rev Esp Med Nucl Imagen Mol 2025; 44:500128. [PMID: 40147757 DOI: 10.1016/j.remnie.2025.500128] [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: 02/04/2025] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/29/2025]
Abstract
The state of the art and future perspectives of new radionuclides in Nuclear Medicine continue to evolve, driven by the development of isotopes with innovative applications in theragnostics. In this second part of the continuing education series, the clinical and therapeutic applications of terbium, actinium, and bismuth are analyzed in depth. The use of the four terbium isotopes (terbium-149, terbium-152, terbium-155, and terbium-161) is described, offering a versatile system for both diagnosis and treatment due to their chemical similarity to lutetium-177, along with the challenges related to their production and availability. Additionally, actinium-225, a powerful alpha-emitting radionuclide, is reviewed for its growing role in Targeted Alpha Therapy (TAT), particularly in prostate cancer and neuroendocrine tumors. Finally, bismuth-213, derived from actinium-225, is analyzed for its short half-life, making it a viable option for localized and selective therapies. Despite technical and production challenges, these radionuclides are driving the evolution of precision medicine, expanding therapeutic and diagnostic possibilities in Nuclear Medicine.
Collapse
Affiliation(s)
- L Sancho
- Servicio de Medicina Nuclear, Clínica Universidad de Navarra, Madrid, Spain
| | - A Roteta
- Servicio de Medicina Nuclear, Hospital Universitario de Donostia, San Sebastián, Spain
| | - I Torres
- Servicio de Medicina Nuclear, Hospital La Fe, Valencia, Spain
| | - M de Arcocha
- Unidad de Radiofarmacia, Servicio de Medicina Nuclear, Hospital Universitario Marqués de Valdecilla, Santander, Spain; Grupo de Imagen Molecular IDIVAL, Santander, Spain
| | - R Ramos
- Unidad de Radiofarmacia, Servicio de Medicina Nuclear, Clínica Universidad de Navarra, Madrid, Spain
| | - M L Domínguez
- Servicio de Medicina Nuclear, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - J J Rosales
- Servicio de Medicina Nuclear, Clínica Universidad de Navarra, Pamplona, Spain
| | - E Prieto
- Servicio de Radiofísica y Protección Radiológica, Clínica Universidad de Navarra, Pamplona, Spain.
| | - G Quincoces
- Unidad de Radiofarmacia, Servicio de Medicina Nuclear, Clínica Universidad de Navarra, Pamplona, Spain
| |
Collapse
|
2
|
Grewal US, Gbolahan OB, Takalkar AM, Halperin DM. Alpha radioligand therapy in neuroendocrine neoplasms: current landscape and spotlight on RYZ101. Future Oncol 2025; 21:1357-1363. [PMID: 40162745 PMCID: PMC12051522 DOI: 10.1080/14796694.2025.2485650] [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: 10/31/2024] [Accepted: 03/25/2025] [Indexed: 04/02/2025] Open
Abstract
The therapeutic landscape for neuroendocrine tumors (NETs) has rapidly evolved over the last three decades with the influx of a myriad of treatment options, such as somatostatin analogs (SSAs), targeted therapies, alkylating chemotherapies, and radioligand therapy (RLT). While the advent and regulatory approval of beta emitting RLT such as 177Lu-DOTATATE has offered a valuable therapeutic option for patients with NETs, there is still very significant room to tap the maximal therapeutic potential of RLT. Alpha RLT agents such as RYZ101 (225Ac-DOTATATE) offer a potential advantage over beta RLT due to more complex double-stranded DNA breaks and targeted cytotoxicity, as well as potential for minimizing off-target side-effects. Existing pre-clinical and clinical data suggest promising safety and efficacy of RYZ101 among patients with NETs. The ongoing phase 1b/3 trial ACTION-1 is poised to compare the safety and efficacy of RYZ101 against standard care therapies in advanced gastroenteropancreatic NETs (Ki67 ≤ 20%), after disease progression of prior 177Lu-SSA therapy. Yet, other alpha RLT agents are currently being investigated in both RLT-naïve as well-pre-treated patient populations. While long-term safety and efficacy data are awaited, alpha RLT agents such as RYZ101 offer a unique opportunity to enhance the therapeutic promise of RLT in NETs.
Collapse
Affiliation(s)
- Udhayvir. S. Grewal
- Division of Hematology, Oncology and Bone and Marrow Transplantation, University of Iowa Health Care, Iowa City, IA, USA
- Holden Comprehensive Cancer Center, Iowa City, IA, USA
| | - Olumide. B. Gbolahan
- Division of Medical Oncology, Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Amol. M. Takalkar
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
- Nuclear Medicine and Molecular Imaging, Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Daniel. M. Halperin
- Division of Medical Oncology, Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| |
Collapse
|
3
|
Zhang Y, Coghi P, Ren Z, Hosmane NS, Zhu Y. Comparison of Radionuclide Drug Conjugates With Boron Neutron Capture Therapy: An Overview of Targeted Charged Particle Radiation Therapy. Med Res Rev 2025; 45:867-886. [PMID: 39690514 DOI: 10.1002/med.22093] [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: 06/24/2024] [Revised: 09/11/2024] [Accepted: 11/25/2024] [Indexed: 12/19/2024]
Abstract
Targeted charged alpha- and beta-particle therapies are currently being used in clinical radiation treatments as newly developed methods for either killing or controlling tumor cell growth. The alpha particles can be generated either through a nuclear decay reaction or in situ by a nuclear fission reaction such as the boron neutron capture reaction. Different strategies have been employed to improve the selectivity and delivery of radiation dose to tumor cells based on the source of the clinically used alpha particles. As a result, the side effects of the treatment can be minimized. The increasing attention and research efforts on targeted alpha-particle therapy have been fueled by exciting results of both academic research and clinical trials. It is highly anticipated that alpha-particle therapy will improve the efficacy of treating malignant tumors. In this overview, we compare radionuclide drug conjugates (RDC) with boron neutron capture therapy (BNCT) to present recent developments in targeted alpha-particle therapy.
Collapse
Affiliation(s)
| | - Paolo Coghi
- Laboratory for Drug Discovery from Natural Resources & Industrialization, School of Pharmacy, Macau University of Science and Technology, Macau, China
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Zimo Ren
- Laboratory for Drug Discovery from Natural Resources & Industrialization, School of Pharmacy, Macau University of Science and Technology, Macau, China
| | - Narayan S Hosmane
- Department of Chemistry and Biochemistry, Northern Illinois University, DeKalb, Illinois, USA
| | | |
Collapse
|
4
|
Yamaguchi N, Wei JJ, Isomoto H. Clinical application of targeted α-emitter therapy in gastroenteropancreatic neuroendocrine neoplasms. J Gastroenterol 2025:10.1007/s00535-025-02241-z. [PMID: 40220045 DOI: 10.1007/s00535-025-02241-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 02/28/2025] [Indexed: 04/14/2025]
Abstract
Effective therapeutic strategies for advanced gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs) remain challenging, including a lack of response to therapy and post-treatment relapse. The rapid development of targeted radionuclide therapy (TRT) offers promising data for patients with somatostatin receptor (SSTR)-expressing tumors. This approach exhibits more advantages than somatostatin analog (SSA) therapy, which is primarily effective for well-differentiated and slow-growing GEP-NENs. Fortunately, some clinical studies on peptide receptor radionuclide therapy (PRRT) labeled with α-emitting radionuclides for GEP-NENs patients showed effective results for those with more advanced GEP-NENs, or those with malignant metastasis. For the improvement of clinical efficacy and the decline in the incidence of treatment-related relapse, recent progress in developing novel techniques and effective disease management strategies for optimal targeting has led to the emergence of targeted alpha therapy (TAT) in GEP-NENs patients. For instance, labeled technology and combination therapy could contribute to significantly improved long-term outcomes. However, the exact dosimetry for precision oncology, the shortage of radionuclides, and the stability of disease control are still under careful consideration. More high-quality, large-scale prospective studies are essential for obtaining valuable evidence on challenging problems and for further exploration.
Collapse
Affiliation(s)
- Naoyuki Yamaguchi
- Department of Endoscopy, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, Nagasaki, 852-8501, Japan
| | - Jing-Jing Wei
- Department of Endoscopy, the First Affiliated Hospital of Fujian Medical University, Cha Zhong Road No.20, Tai Jiang District, Fuzhou, 350004, Fujian, China.
- Department of Endoscopy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, Fujian, China.
- Division of Gastroenterology and Nephrology, Department of Multidisciplinary Internal Medicine, School of Medicine, Faculty of Medicine, Tottori University, Yonago, 683-8504, Japan.
| | - Hajime Isomoto
- Division of Gastroenterology and Nephrology, Department of Multidisciplinary Internal Medicine, School of Medicine, Faculty of Medicine, Tottori University, Yonago, 683-8504, Japan
| |
Collapse
|
5
|
Lacerda S, de Kruijff RM, Djanashvili K. The Advancement of Targeted Alpha Therapy and the Role of Click Chemistry Therein. Molecules 2025; 30:1296. [PMID: 40142070 PMCID: PMC11944744 DOI: 10.3390/molecules30061296] [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: 01/06/2025] [Revised: 02/21/2025] [Accepted: 03/11/2025] [Indexed: 03/28/2025] Open
Abstract
Recent years have seen a swift rise in the use of α-emitting radionuclides such as 225Ac and 223Ra as various radiopharmaceuticals to treat (micro)metastasized tumors. They have shown remarkable effectiveness in clinical practice owing to the highly cytotoxic α-particles that are emitted, which have a very short range in tissue, causing mainly double-stranded DNA breaks. However, it is essential that both chelation and targeting strategies are optimized for their successful translation to clinical application, as α-emitting radionuclides have distinctly different features compared to β--emitters, including their much larger atomic radius. Furthermore, upon α-decay, any daughter nuclide irrevocably breaks free from the targeting molecule, known as the recoil effect, dictating the need for faster targeting to prevent healthy tissue toxicity. In this review we provide a brief overview of the current status of targeted α-therapy and highlight innovations in α-emitter-based chelator design, focusing on the role of click chemistry to allow for fast complexation to biomolecules at mild labeling conditions. Finally, an outlook is provided on different targeting strategies and the role that pre-targeting can play in targeted alpha therapy.
Collapse
Affiliation(s)
- Sara Lacerda
- Centre de Biophysique Moléculaire, CNRS UPR 4301, Université d’Orléans, Rue Charles Sadron, 45071 Orléans, France;
| | - Robin M. de Kruijff
- Department of Radiation Science and Technology, Delft University of Technology, Mekelweg 15, 2629 JB Delft, The Netherlands;
| | - Kristina Djanashvili
- Department of Radiation Science and Technology, Delft University of Technology, Mekelweg 15, 2629 JB Delft, The Netherlands;
- Department of Biotechnology, Delft University of Technology, Van der Maasweg 9, 2629 HZ Delft, The Netherlands
| |
Collapse
|
6
|
Mapanao AK, Busslinger SD, Mehta A, Kegler K, Favaretto C, Grundler PV, Talip Z, Köster U, Johnston K, Schibli R, van der Meulen NP, Müller C. Preclinical investigation of [ 149Tb]Tb-DOTATATE and [ 149Tb]Tb-DOTA-LM3 for tumor-targeted alpha therapy. Eur J Nucl Med Mol Imaging 2025; 52:1383-1398. [PMID: 39743617 DOI: 10.1007/s00259-024-07035-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 12/15/2024] [Indexed: 01/04/2025]
Abstract
PURPOSE Terbium-149 is a short-lived α-particle emitter, potentially useful for tumor-targeted therapy. The aim of this study was to investigate terbium-149 in combination with the somatostatin receptor (SSTR) agonist DOTATATE and the SSTR antagonist DOTA-LM3. The radiopeptides were evaluated to compare their therapeutic efficacy in vitro and in vivo. METHODS Terbium-149 was produced at ISOLDE/CERN and chemically purified at the Paul Scherrer Institute. Radiolabeling of somatostatin analogues with [149Tb]TbCl3 was performed under standard labeling conditions at pH 4.5. Cell viability (MTT) and survival assays (colony forming) assays were performed after 16-18 h exposure of SSTR-positive AR42J rat pancreatic tumor cells to various activity concentrations of [149Tb]Tb-DOTATATE and [149Tb]Tb-DOTA-LM3. DNA double-strand breaks were determined using immunofluorescence imaging of γ-H2A.X and 53BP1. Therapy studies were performed with AR42J tumor-bearing mice injected with 1 × 5 MBq or 2 × 5 MBq of the respective radiopeptide. The tolerability of up to 40 MBq [149Tb]Tb-DOTATATE or 40 MBq [149Tb]Tb-DOTA-LM3 was assessed with regard to undesired effects to the bone marrow and kidneys in immunocompetent mice without tumors. RESULTS The radiolabeling of peptides was achieved at molar activities of up to 20 MBq/nmol at ≥ 98% radiochemical purity. AR42J cell viability was reduced in an activity-dependent manner, with [149Tb]Tb-DOTA-LM3 being slightly more potent than [149Tb]Tb-DOTATATE (EC50: 0.5 vs. 1.2 kBq/mL). Both radiopeptides induced a similar number of γ-H2A.X and 53BP1 foci per nuclei, which indicated DNA damage in AR42J tumor cells. Injection of tumor-bearing mice with 1 × 5 MBq radiopeptide resulted in median survival times of 16.5 days and 19 days for [149Tb]Tb-DOTATATE and [149Tb]Tb-DOTA-LM3, respectively, as compared to only 8 days for untreated control mice. Application of 2 × 5 MBq of the radiopeptides further extended the median survival times to 30 days and 29 days, respectively. The blood cell counts and values for blood plasma biomarkers of treated mice without tumors were similar to those of untreated controls. Renal accumulation of [99mTc]Tc-DMSA was similar in all mice, indicating normal kidney function. CONCLUSION 149Tb-based radiopeptides effectively reduced the viability of tumor cells in vitro as well as the tumor growth in mice without causing relevant adverse events, irrespective of whether the SSTR agonist or antagonist was employed. These data encourage further preclinical application of terbium-149 to evaluate its potential in combination with other tumor-targeting agents.
Collapse
Affiliation(s)
- Ana Katrina Mapanao
- Center for Radiopharmaceutical Sciences, PSI Center for Life Sciences, Villigen-PSI, 5232, Switzerland
| | - Sarah D Busslinger
- Center for Radiopharmaceutical Sciences, PSI Center for Life Sciences, Villigen-PSI, 5232, Switzerland
| | - Avni Mehta
- Center for Radiopharmaceutical Sciences, PSI Center for Life Sciences, Villigen-PSI, 5232, Switzerland
| | | | - Chiara Favaretto
- Center for Radiopharmaceutical Sciences, PSI Center for Life Sciences, Villigen-PSI, 5232, Switzerland
- Division of Nuclear Medicine, University Hospital Basel, Basel, 4031, Switzerland
| | - Pascal V Grundler
- Center for Radiopharmaceutical Sciences, PSI Center for Life Sciences, Villigen-PSI, 5232, Switzerland
| | - Zeynep Talip
- Center for Radiopharmaceutical Sciences, PSI Center for Life Sciences, Villigen-PSI, 5232, Switzerland
- Laboratory of Radiochemistry, PSI Center for Nuclear Engineering and Sciences, Villigen-PSI, 5232, Switzerland
| | - Ulli Köster
- Institut Laue-Langevin, Grenoble, 38042, France
| | - Karl Johnston
- Physics Department, ISOLDE/CERN, Geneva, 1211, Switzerland
| | - Roger Schibli
- Center for Radiopharmaceutical Sciences, PSI Center for Life Sciences, Villigen-PSI, 5232, Switzerland
- Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, 8093, Switzerland
| | - Nicholas P van der Meulen
- Center for Radiopharmaceutical Sciences, PSI Center for Life Sciences, Villigen-PSI, 5232, Switzerland
- Laboratory of Radiochemistry, PSI Center for Nuclear Engineering and Sciences, Villigen-PSI, 5232, Switzerland
| | - Cristina Müller
- Center for Radiopharmaceutical Sciences, PSI Center for Life Sciences, Villigen-PSI, 5232, Switzerland.
- Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, 8093, Switzerland.
| |
Collapse
|
7
|
Navalkissoor S, Grossman A. Somatostatin receptor-linked α-particle therapy in neuroendocrine tumours. J Neuroendocrinol 2025; 37:e13463. [PMID: 39529416 DOI: 10.1111/jne.13463] [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: 06/19/2024] [Revised: 09/19/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
The incidence and prevalence of neuroendocrine tumours (NETs) are on the rise, but to date, only complete surgical resection is curative. Among the various therapeutic options for metastatic disease, peptide receptor radionuclide therapy (PRRT), linking a radioactive moiety to an octreotide derivative, has been shown to be highly efficacious and a well-tolerated therapy, improving progression-free survival and prolonging overall survival. Nevertheless, complete responses are rare, and the current β-particle emitters have non-optimal radiobiological properties. A new generation of α-particle-emitting radionuclides is being developed, with the advantages of very high energy and a short path length. We survey the most recent developments in this field, summarising the result of currently performed studies in this potentially ground-breaking novel form of therapy for NETs.
Collapse
Affiliation(s)
- Shaunak Navalkissoor
- Department of Nuclear Medicine, Royal Free London NHS Foundation Trust, London, UK
| | - Ashley Grossman
- NET UNIT, ENETS Centre of Excellence, Royal Free London NHS Foundation Trust, London, UK
| |
Collapse
|
8
|
Puranik AD, Dev ID, Prasad V. Frontiers in radiopharmaceuticals for neuroendocrine tumors. J Neuroendocrinol 2025; 37:e70006. [PMID: 39961670 DOI: 10.1111/jne.70006] [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: 11/07/2024] [Revised: 01/30/2025] [Accepted: 01/31/2025] [Indexed: 03/20/2025]
Abstract
Neuroendocrine tumors encompass a wide range of tumors which originate from neural crest cells. These tumors were thought to be rare tumors, however, with the advent of advanced diagnostic techniques along with better understanding of the clinical presentation and histology of these tumors, the incidence of these tumors is exponentially rising. As the incidence and detection rate of NENs increased, the concept of 'heterogeneity' came into picture, which in turn led to dual-tracer imaging with addition of FDG PET/CT. Despite an imaging-based decision-making approach for NENs, there is still a significant subset of patients where the imaging-based biomarkers fall short in disease assessment, prognostication and improving outcomes. Alternate pathways as well as better peptide vectors for targeting the somatostatin receptor need to be studied. In this article, we address the existing as well as emerging trends in radiopharmaceuticals used for NENs, which are likely to impact not just the diagnostic algorithms in future, but also management strategies.
Collapse
Affiliation(s)
- Ameya D Puranik
- Department of Nuclear Medicine and Molecular Imaging, Tata Memorial Hospital, Tata Memorial Center, Homi Bhabha National Institute, Mumbai, India
| | - Indraja D Dev
- Department of Nuclear Medicine and Molecular Imaging, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Center, Homi Bhabha National Institute, Mumbai, India
| | - Vikas Prasad
- Mallinckrodt Institute of Radiology, Washington University in Saint Louis, Saint Louis, Missouri, USA
| |
Collapse
|
9
|
Wen X, Zhao T, Yang H, Shi M, Wee XJ, Fu J, Lin M, Zhang Z, Zou M, Green D, Wu X, Chen X, Zhang J. Development of [ 225Ac]Ac‑LNC1011 for targeted alpha-radionuclide therapy of prostate cancer. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07155-9. [PMID: 39992401 DOI: 10.1007/s00259-025-07155-9] [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: 10/31/2024] [Accepted: 02/12/2025] [Indexed: 02/25/2025]
Abstract
PURPOSE Prostate-specific membrane antigen (PSMA) radioligand therapy (PRLT) has become a promising option for treating metastatic castration-resistant prostate cancer (mCRPC). Radioligands labelled with the 68Ga/177Lu theranostic pair have been most widely used in the clinic for diagnosis and therapy, respectively. This study aims to develop a novel PSMA-targeted radioligand, LNC1011, radiolabeled with alpha-emitter 225Ac, to optimise pharmacokinetic properties and assess its potential for targeted alpha therapy (TAT) in prostate cancer treatment. METHODS LNC1011 (Dan-PSMA) was synthesised based on a PSMA-binding ligand with the addition of a dansylated amino acid. Systematic radiochemical analyses were conducted to confirm the successful synthesis and radiolabelling of [225Ac]Ac-LNC1011. Cell uptake and competition binding assays were performed in PSMA-positive PC3-PIP tumour cells to evaluate the binding affinity and PSMA targeting specificity. The pharmacokinetics properties and tumour uptake were characterised by biodistribution studies using healthy mice and a PC3-PIP xenograft mouse model injected with [225Ac]Ac-LNC1011. Radioligand therapy studies and maximum tolerated dose (MTD) assays were conducted to systematically evaluate the therapeutic efficacy and the safety of [225Ac]Ac-LNC1011. RESULTS [225Ac]Ac-LNC1011 was successfully radiolabelled with high radiochemical purity (> 97%) and high stability within 96 h (radiochemical purity > 96%). The high binding affinity of LNC1011 (IC50 = 16.28 nM) to PSMA was comparable to that of PSMA-617 (IC50 = 27.93 nM). Biodistribution studies confirmed that [225Ac]Ac-LNC1011 had moderate blood elimination half-life (T1/2z = 13.4 ± 0.57 h), which was at an optimised level between [225Ac]Ac-PSMA-617 (T1/2z = 5.19 ± 0.12 h) and [225Ac]Ac-PSMA-EB-01 (T1/2z = 25.18 ± 2.78 h). In addition, high tumour uptake of [225Ac]Ac-LNC1011 was identified to be 38.28 ± 10.04%ID/g at 1 h post-injection. The specific uptake gradually increased and peaked at 24 h (80.57 ± 3.00%ID/g) and persisted at a high level up to 72 h post-injection (50.58 ± 5.37%ID/g). Targeted alpha therapy results showed the complete inhibition of PC3-PIP tumour growth after administration of a single dose of 1 µCi and 0.5 µCi of [225Ac]Ac-LNC1011 similar to 0.5 µCi [225Ac]Ac-PSMA-617. At the 0.1 µCi dose level, partial remission was observed for [225Ac]Ac-LNC1011, as recurrence was found 20 days after administration. In contrast, mice treated with 0.1 µCi [225Ac]Ac-PSMA-617 showed incomplete tumour inhibition under the same conditions. CONCLUSION [225Ac]Ac-LNC1011 was successfully radiolabelled with high radiochemical purity and stability. With significantly improved tumour uptake and retention over PSMA-617, [225Ac]Ac-LNC1011 showed significantly better therapeutic efficacy than [225Ac]Ac-PSMA-617 for targeted alpha therapy of prostate cancer.
Collapse
Affiliation(s)
- Xuejun Wen
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
- Yantai Lannacheng Biotechnology Co., Ltd., Yantai, 264100, China
- Yong Loo Lin School of Medicine, Theranostics Center of Excellence (TCE), National University of Singapore, 11 Biopolis Way, Helios, 138667, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Tianzhi Zhao
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
- Yong Loo Lin School of Medicine, Theranostics Center of Excellence (TCE), National University of Singapore, 11 Biopolis Way, Helios, 138667, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Hongzhang Yang
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
- Yong Loo Lin School of Medicine, Theranostics Center of Excellence (TCE), National University of Singapore, 11 Biopolis Way, Helios, 138667, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Mengqi Shi
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
- Yong Loo Lin School of Medicine, Theranostics Center of Excellence (TCE), National University of Singapore, 11 Biopolis Way, Helios, 138667, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Xin Jie Wee
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
| | - Jiayu Fu
- Yantai Lannacheng Biotechnology Co., Ltd., Yantai, 264100, China
| | - Min Lin
- Yantai Lannacheng Biotechnology Co., Ltd., Yantai, 264100, China
| | - Zhenyue Zhang
- Yantai Lannacheng Biotechnology Co., Ltd., Yantai, 264100, China
| | - Maosheng Zou
- Yantai Lannacheng Biotechnology Co., Ltd., Yantai, 264100, China
| | - David Green
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
| | - Xiaoming Wu
- Yantai Lannacheng Biotechnology Co., Ltd., Yantai, 264100, China.
- College of Nuclear Science and Technology, Harbin Engineering University, Harbin, 150001, China.
| | - Xiaoyuan Chen
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore.
- Yong Loo Lin School of Medicine, Theranostics Center of Excellence (TCE), National University of Singapore, 11 Biopolis Way, Helios, 138667, Singapore.
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore.
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore.
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, 138673, Singapore.
- Department of Chemical and Biomolecular Engineering, Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117575, Singapore.
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore, 117544, Singapore.
| | - Jingjing Zhang
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore.
- Yong Loo Lin School of Medicine, Theranostics Center of Excellence (TCE), National University of Singapore, 11 Biopolis Way, Helios, 138667, Singapore.
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore.
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore.
| |
Collapse
|
10
|
Das S, Berlin J. Systemic Therapy Improvements Will Render Locoregional Treatments Obsolete for Patients with Cancer with Liver Metastases. Hematol Oncol Clin North Am 2025; 39:191-206. [PMID: 39510673 DOI: 10.1016/j.hoc.2024.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
Hepatic metastases are a major cause of morbidity and mortality for patients with cancer. Apart from curative resection, which offers patients the potential for long-term survival, an array of locoregional therapies, with limited evidence of improving survival, are used to treat them. The authors use examples from the realm of gastrointestinal cancer, largely focusing on the experience of patients with neuroendocrine cancer, hepatobiliary cancer, and colorectal cancer, to suggest that current systemic therapies offer, at minimum, similar survival outcomes for patients compared with these locoregional approaches.
Collapse
Affiliation(s)
- Satya Das
- Department of Medicine, Division of Hematology Oncology, Vanderbilt University Medical Center, 777 Preston Research Building, 2220 Pierce Avenue, Nashville, TN 37232, USA.
| | - Jordan Berlin
- Department of Medicine, Division of Hematology Oncology, Vanderbilt University Medical Center, 777 Preston Research Building, 2220 Pierce Avenue, Nashville, TN 37232, USA. https://twitter.com/jordanberlin5
| |
Collapse
|
11
|
Lee DY, Kim YI. Efficiency and Safety of Targeted Alpha Therapy in Metastatic Neuroendocrine Tumors: A Meta-analysis. Clin Nucl Med 2025; 50:e1-e6. [PMID: 39169519 DOI: 10.1097/rlu.0000000000005404] [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: 08/23/2024]
Abstract
PURPOSE Despite the effectiveness of 177 Lu-based peptide receptor radionuclide therapy in treating metastatic neuroendocrine tumors (NETs), disease progression posttreatment remains a significant challenge. Targeted alpha therapy (TAT) has emerged as a promising option for patients experiencing such progression. This study aims to assess the therapeutic efficiency and toxicity of TAT in patients with metastatic NET through a meta-analysis. PATIENTS AND METHODS We conducted a comprehensive search of PubMed, Embase, Cochrane Library, and CINAHL using relevant keywords. The analysis focused on the pooled proportions of objective response rate (ORR) and disease control rate (DCR) to determine therapeutic efficiency. We also evaluated the incidence of serious hematologic and renal adverse events (grade 3 or 4) to assess toxicity. A subgroup analysis was performed to identify factors influencing therapeutic outcomes. RESULTS Our meta-analysis included 7 studies comprising 162 patients. The results showed that TAT achieved ORR of 49.5% (95% confidence interval [CI]: 41.7%-57.4%) and DCR of 87.0% (95% CI: 72.1%-96.8%). The incidences of hematologic and renal toxicities were low, at 2.1% (95% CI: 0.5%-5.5%) and 3.4% (95% CI: 1.2%-7.3%), respectively. Subgroup analysis indicated consistent therapeutic efficiency across different variables, including prior 177 Lu-based peptide receptor radionuclide therapy treatment, 225 Ac-based TAT, absence of radiosensitizer, and methods of response evaluation, with ORR ranging from 46.6% to 57.1% and DCR from 82.0% to 91.5%. CONCLUSIONS TAT is an effective treatment for metastatic NET, demonstrating substantial disease control and response rates with minimal toxicity. These findings suggest that TAT is a viable therapeutic alternative for patients with metastatic NET.
Collapse
Affiliation(s)
- Dong Yun Lee
- From the Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | | |
Collapse
|
12
|
Ding J, Qin S, Hou X, Zhang J, Yang M, Ma S, Zhu H, Feng Y, Yu F. Recent advances in emerging radiopharmaceuticals and the challenges in radiochemistry and analytical chemistry. Trends Analyt Chem 2025; 182:118053. [DOI: 10.1016/j.trac.2024.118053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
13
|
Lugat A, Chouin N, Chocteau F, Esnault M, Marionneau-Lambot S, Gouard S, Frampas É, Faivre-Chauvet A, Bourgeois M, Morgenstern A, Bruchertseifer F, Chérel M, Kraeber-Bodéré F, Ansquer C, Gaschet J. Survival impact of [ 225Ac]Ac-DOTATOC alpha-therapy in a preclinical model of pancreatic neuroendocrine tumor liver micrometastases. Eur J Nucl Med Mol Imaging 2025; 52:730-743. [PMID: 39269657 DOI: 10.1007/s00259-024-06918-0] [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: 04/23/2024] [Accepted: 09/02/2024] [Indexed: 09/15/2024]
Abstract
Although peptide radionuclide therapy (PRRT) using a somatostatin analog (SSA) radiolabeled with a beta- emitter: [177Lu]Lu-DOTATATE has shown a good clinical efficacy in neuroendocrine tumors (NETs), most of the patients only achieved tumoral stabilization and rare but severe long-term hematological toxicities have been reported. One of the promising options to improve PRRT is targeted alpha therapy. It is therefore essential to propose animal models that can mimic systemic spread disease, especially microscopic disease such as early stage of NET liver metastases to explore targeted alpha therapy. Herein, we report the evaluation of efficacy and toxicity of [225Ac]Ac-DOTATOC in an original preclinical murine model simulating the development of well-characterized liver metastases of pancreatic NETs with SSTR overexpression. METHODS A mouse model of liver metastases of pancreatic NETs was developed by intraportal injection of AR42J cells and explored using [68 Ga]Ga-DOTATOC and [18F]F-FDG PET/MRI. Biodistribution study and radiation dosimetry of [225Ac]Ac-DOTATOC were determined in subcutaneous tumor-bearing NMRI-nude mice. Efficacy and toxicity were determined by intravenous injection of increasing activities of [225Ac]Ac-DOTATOC 10 days after intraportal graft. RESULTS Liver tumors showed a high uptake of [68 Ga]Ga-DOTATOC and no uptake of [18F]F-FDG confirming the well-differentiated phenotype. All groups treated with [225Ac]Ac-DOTATOC showed a significant increase in overall survival compared with DOTATOC-treated mice, especially those treated with the highest activities: 53 days with 240 kBq (p = 0.0001), and 58 days with 2 × 120 kBq (p < 0.0001) vs 28 days with non-radiolabeled DOTATOC. On blood tests, a transient and moderate decreased in white blood cells count after treatment and no severe hepatic or renal toxicity were observed after treatment which was consistent with pathological and radiation dosimetry findings. CONCLUSION [225Ac]Ac-DOTATOC exhibit a favorable efficacy and toxicity profile in a mouse model of liver micrometastatic pancreatic NET.
Collapse
Affiliation(s)
- Alexandre Lugat
- Medical Oncology Department, Nantes University Hospital, 44000, Nantes, France
- Nuclear Medicine Department, Nantes University Hospital, 1, Place Alexis Ricordeau, 44000, Nantes, France
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, 8 Quai Moncousu, BP70721, Cedex 1, 44007, Nantes, France
| | - Nicolas Chouin
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, 8 Quai Moncousu, BP70721, Cedex 1, 44007, Nantes, France
| | - Florian Chocteau
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, 8 Quai Moncousu, BP70721, Cedex 1, 44007, Nantes, France
| | - Mathilde Esnault
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, 8 Quai Moncousu, BP70721, Cedex 1, 44007, Nantes, France
| | - Séverine Marionneau-Lambot
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, 8 Quai Moncousu, BP70721, Cedex 1, 44007, Nantes, France
| | - Sébastien Gouard
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, 8 Quai Moncousu, BP70721, Cedex 1, 44007, Nantes, France
| | - Éric Frampas
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, 8 Quai Moncousu, BP70721, Cedex 1, 44007, Nantes, France
- Central Department of Radiology and Medical Imaging, Nantes University Hospital, 44000, Nantes, France
| | - Alain Faivre-Chauvet
- Nuclear Medicine Department, Nantes University Hospital, 1, Place Alexis Ricordeau, 44000, Nantes, France
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, 8 Quai Moncousu, BP70721, Cedex 1, 44007, Nantes, France
| | - Mickaël Bourgeois
- Nuclear Medicine Department, Nantes University Hospital, 1, Place Alexis Ricordeau, 44000, Nantes, France
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, 8 Quai Moncousu, BP70721, Cedex 1, 44007, Nantes, France
| | - Alfred Morgenstern
- European Commission, Joint Research Centre, Directorate for Nuclear Safety and Security, Karlsruhe, Germany
| | - Frank Bruchertseifer
- European Commission, Joint Research Centre, Directorate for Nuclear Safety and Security, Karlsruhe, Germany
| | - Michel Chérel
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, 8 Quai Moncousu, BP70721, Cedex 1, 44007, Nantes, France
- Department of Nuclear Medicine, Institut de Cancérologie de L'Ouest (ICO) - Site Gauducheau, Saint-Herblain, France
| | - Françoise Kraeber-Bodéré
- Nuclear Medicine Department, Nantes University Hospital, 1, Place Alexis Ricordeau, 44000, Nantes, France
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, 8 Quai Moncousu, BP70721, Cedex 1, 44007, Nantes, France
| | - Catherine Ansquer
- Nuclear Medicine Department, Nantes University Hospital, 1, Place Alexis Ricordeau, 44000, Nantes, France
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, 8 Quai Moncousu, BP70721, Cedex 1, 44007, Nantes, France
| | - Joëlle Gaschet
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, 8 Quai Moncousu, BP70721, Cedex 1, 44007, Nantes, France.
| |
Collapse
|
14
|
Yoshimoto M, Washiyama K, Ohnuki K, Doi A, Inokuchi M, Kojima M, Miller BW, Yoshii Y, Inaki A, Fujii H. Long-Term Therapeutic Effects of 225Ac-DOTA-E[c(RGDfK)] 2 Induced by Radiosensitization via G2/M Arrest in Pancreatic Ductal Adenocarcinoma. Pharmaceutics 2024; 17:9. [PMID: 39861661 PMCID: PMC11768328 DOI: 10.3390/pharmaceutics17010009] [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: 11/29/2024] [Revised: 12/14/2024] [Accepted: 12/17/2024] [Indexed: 01/27/2025] Open
Abstract
Background: Alpha radionuclide therapy has emerged as a promising novel strategy for cancer treatment; however, the therapeutic potential of 225Ac-labeled peptides in pancreatic cancer remains uninvestigated. Methods: In the cytotoxicity study, tumor cells were incubated with 225Ac-DOTA-RGD2. DNA damage responses (γH2AX and 53BP1) were detected using flowcytometry or immunohistochemistry analysis. Biodistribution and therapeutic studies were carried out in BxPC-3-bearing mice. Results: 225Ac-DOTA-RGD2 demonstrated potent cytotoxicity against cells expressing αvβ3 or αvβ6 integrins and induced G2/M arrest and γH2AX expression as a marker of double-stranded DNA breaks. 225Ac-DOTA-RGD2 (20, 40, 65, or 90 kBq) showed favorable pharmacokinetics and remarkable tumor growth inhibition without severe side effects in the BxPC-3 mouse model. In vitro studies revealed that 5 and 10 kBq/mL of 225Ac-DOTA-RGD2 swiftly induced G2/M arrest and elevated γH2AX expression. Furthermore, to clarify the mechanism of successful tumor growth inhibition for a long duration in vivo, we investigated whether short-term high radiation exposure enhances radiation sensitivity. Initially, a 4 h induction treatment with 5 and 10 kBq/mL of 225Ac-DOTA-RGD2 enhanced both cytotoxicity and γH2AX expression with 0.5 kBq/mL of 225Ac-DOTA-RGD2 compared to a treatment with only 0.5 kBq/mL of 225Ac-DOTA-RGD2. Meanwhile, the γH2AX expression induced by 5 or 10 kBq/mL of 225Ac-DOTA-RGD2 alone decreased over time. Conclusions: These findings highlight the potential of using 225Ac-DOTA-RGD2 in the treatment of intractable pancreatic cancers, as its ability to induce G2/M cell cycle arrest enhances radiosensitization, resulting in notable growth inhibition.
Collapse
Affiliation(s)
- Mitsuyoshi Yoshimoto
- Division of Functional Imaging, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa 277-8577, Japan
| | - Kohshin Washiyama
- Advanced Clinical Research Center, Fukushima Global Medical Science Center, Fukushima Medical University, Fukushima 960-1295, Japan;
| | - Kazunobu Ohnuki
- Division of Functional Imaging, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa 277-8577, Japan
| | - Ayano Doi
- Division of Functional Imaging, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa 277-8577, Japan
| | - Miki Inokuchi
- Division of Functional Imaging, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa 277-8577, Japan
| | - Motohiro Kojima
- Division of Pathology, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa 277-8577, Japan
| | - Brian W. Miller
- Department of Radiation Oncology, College of Medicine, University of Arizona, Tucson, AZ 85719, USA
| | - Yukie Yoshii
- National Institute of Radiological Sciences, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
| | - Anri Inaki
- Division of Functional Imaging, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa 277-8577, Japan
| | - Hirofumi Fujii
- Division of Functional Imaging, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa 277-8577, Japan
| |
Collapse
|
15
|
Silindir-Gunay M, Ozolmez N. Adverse reactions to therapeutic radiopharmaceuticals. Appl Radiat Isot 2024; 214:111527. [PMID: 39332267 DOI: 10.1016/j.apradiso.2024.111527] [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: 06/18/2024] [Revised: 09/20/2024] [Accepted: 09/23/2024] [Indexed: 09/29/2024]
Abstract
Radiopharmaceuticals are drugs used in treatment or diagnosis that contain a radioactive part, usually a pharmaceutical part in their structure. Adverse drug reactions are harmful and unexpected responses that occur when administered at normal doses. Although radiopharmaceuticals are regarded as safe medical products, adverse reactions should not be ignored. More serious adverse reactions such as myelosuppression, pleural effusion, and death may develop in therapeutic radiopharmaceuticals due to their use at higher doses than those used in diagnosis. Therefore, monitoring adverse reactions and reporting them to health authorities is important. This review aims to provide information about adverse reactions that may be related to radiopharmaceuticals used in treatment.
Collapse
Affiliation(s)
- Mine Silindir-Gunay
- Department of Radiopharmacy, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey.
| | - Nur Ozolmez
- Department of Radiopharmacy, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey.
| |
Collapse
|
16
|
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
|
17
|
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
|
18
|
Ingham A, Wharton L, Koniar H, Merkens H, McNeil S, Sekar S, Osooly M, Rodríguez-Rodríguez C, Bénard F, Schaffer P, Yang H. Preclinical evaluation of [ 225Ac]Ac-crown-TATE - An alpha-emitting radiopharmaceutical for neuroendocrine tumors. Nucl Med Biol 2024; 138-139:108944. [PMID: 39154412 DOI: 10.1016/j.nucmedbio.2024.108944] [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: 04/08/2024] [Revised: 07/25/2024] [Accepted: 07/30/2024] [Indexed: 08/20/2024]
Abstract
BACKGROUND Targeted alpha therapy (TAT) of somatostatin receptor-2 (SSTR2) positive neuroendocrine tumors (NETs) involving Ac-225 ([225Ac]Ac-DOTA-TATE) has previously demonstrated improved therapeutic efficacy over conventional beta particle-emitting peptide receptor radionuclide therapy agents. DOTA-TATE requires harsh radiolabeling conditions for chelation of [225Ac]Ac3+, which can limit the achievable molar activities and thus therapeutic efficacy of such TAT treatments. Macropa-TATE was recently highlighted as a potential alternative to DOTA-TATE, owing to the mild radiolabeling conditions and high affinity toward [225Ac]Ac3+; however, elevated liver and kidney uptake were noted as a major limitation and a suitable imaging radionuclide is yet to be reported, which will be required for patient dosimetry studies and assessment of therapeutic benefit. Previously, [155Tb]Tb-crown-TATE has shown highly effective imaging of NETs in preclinical SPECT/CT studies, with high tumor uptake and low non-target accumulation; these favourable properties and the versatile coordination behavior of the crown chelator may therefore show promise for combination with Ac-225 for TAT. METHODS Crown-TATE was labeled with Ac-225, and radiochemical yield was analyzed as the function of crown-TATE concentration. LogD7.4 was measured as the indication of hydrophilicity. Free [225Ac]Ac3+ release from [225Ac]Ac-crown-TATE in human serum was studied. Biodistribution studies of [225Ac]Ac-crown-TATE in mice bearing AR42J tumors was evaluated at 1, 4, 24, 48, and 120 h, and the absorbed dose to major organs calculated. Therapy-monitoring studies with AR42J tumor bearing mice were undertaken using 30 kBq and 55 kBq doses of [225Ac]Ac-crown-TATE and compared to controls treated with PBS or crown-TATE. RESULTS [225Ac]Ac-crown-TATE was successfully prepared with high molar activity (640 kBq/nmol), and characterized as a moderately hydrophilic radioligand (LogD7.4 = -1.355 ± 0.135). No release of bound Ac-225 was observed over 9 days in human serum. Biodistribution studies of [225Ac]Ac-crown-TATE showed good initial tumor uptake (11.1 ± 1.7% IA/g at 4 h) which was sustained up to 120 h p.i. (6.92 ± 2.03% IA/g). Dosimetry calculations showed the highest absorbed dose was delivered to the tumors. Therapy monitoring studies demonstrated significant (log-rank test, P < 0.005) improved survival in both treatment groups compared to controls. CONCLUSIONS This preclinical study demonstrated the therapeutic efficacy of [225Ac]Ac-crown-TATE for treatment of NETs, and highlights the potential of using crown chelator for stable chelation of Ac-225 under mild conditions.
Collapse
Affiliation(s)
- Aidan Ingham
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, British Columbia V6T 2A3, Canada
| | - Luke Wharton
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, British Columbia V6T 2A3, Canada
| | - Helena Koniar
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, British Columbia V6T 2A3, Canada; Department of Physics and Astronomy, University of British Columbia, 6224 Agronomy Road, Vancouver, British Columbia V6T 1Z1, Canada
| | - Helen Merkens
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, British Columbia V5Z 1L3, Canada; Department of Radiology, UBC, University of British Columbia, Vancouver, British Columbia V5Z 1M9, Canada
| | - Scott McNeil
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, British Columbia V6T 2A3, Canada
| | - Sathiya Sekar
- Faculty of Pharmaceutical Sciences, University of British Columbia, 2405 Wesbrook Mall, Vancouver, British Columbia V6T 1Z3, Canada
| | - Maryam Osooly
- Faculty of Pharmaceutical Sciences, University of British Columbia, 2405 Wesbrook Mall, Vancouver, British Columbia V6T 1Z3, Canada
| | - Cristina Rodríguez-Rodríguez
- Department of Physics and Astronomy, University of British Columbia, 6224 Agronomy Road, Vancouver, British Columbia V6T 1Z1, Canada; Faculty of Pharmaceutical Sciences, University of British Columbia, 2405 Wesbrook Mall, Vancouver, British Columbia V6T 1Z3, Canada
| | - François Bénard
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, British Columbia V5Z 1L3, Canada; Department of Radiology, UBC, 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, UBC, University of British Columbia, Vancouver, British Columbia V5Z 1M9, 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.
| |
Collapse
|
19
|
Ramdhani D, Watabe H, Hardianto A, Janitra RS. Complexation of 3p- C-NETA with radiometal ions: A density functional theory study for targeted radioimmunotherapy. Heliyon 2024; 10:e34875. [PMID: 39144950 PMCID: PMC11320446 DOI: 10.1016/j.heliyon.2024.e34875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 07/13/2024] [Accepted: 07/17/2024] [Indexed: 08/16/2024] Open
Abstract
Bifunctional chelators (BFCs) are vital in the design of effective radiopharmaceuticals, as they are able to bind to both a radiometal ion and a targeting vector. The 3p-C-NETA or 4-[2-(bis-carboxy-methylamino)-5-(4-nitrophenyl)-entyl])-7-carboxymethyl-[1,4,7]tri-azonan-1-yl acetic acid is a novel and promising BFC, developed for diagnostic and therapeutic purposes. The binding affinity between the BFC and radiometal ion significantly impacts their effectiveness. Predicting the equilibrium constants for the formation of 1:1 radiometals/chelator complexes (log K1 values) is crucial for designing BFCs with improved affinity and selectivity for radiometals. The purpose of this study is to evaluate the complexation of Ga3+, Tb3+, Bi3+, and Ac3+ radiometal ions with 3p-C-NETA using density functional theory (B3LYP and M06-HF functional) and 6-311G(d)/SDD basis sets, where the 1,4,7,10-tetrazacyclodecane-1,4,7,10-tetracetic acid (DOTA) was employed as a benchmark. Formation of the [Ac3+(3p-C-NETA)(H2O)]- complexes is predicted to be markedly less stable compared to the other complexes, exhibiting the lowest chemical hardness and the highest chemical softness. Additionally, the chelation stability of the complexes is mainly determined by ligand-ion and ion-water interactions, which depend on the atomic charge and atomic radius of the metal ion.
Collapse
Affiliation(s)
- Danni Ramdhani
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
- Division of Radiation Protection and Safety Control, Cyclotron and Radioisotope Center (CYRIC), Tohoku University, Sendai, Japan
| | - Hiroshi Watabe
- Division of Radiation Protection and Safety Control, Cyclotron and Radioisotope Center (CYRIC), Tohoku University, Sendai, Japan
| | - Ari Hardianto
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Sumedang, Indonesia
| | - Regaputra S. Janitra
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Sumedang, Indonesia
| |
Collapse
|
20
|
Koniar H, Wharton L, Ingham A, Rodríguez-Rodríguez C, Kunz P, Radchenko V, Yang H, Rahmim A, Uribe C, Schaffer P. In vivoquantitative SPECT imaging of actinium-226: feasibility and proof-of-concept. Phys Med Biol 2024; 69:155003. [PMID: 38925140 DOI: 10.1088/1361-6560/ad5c37] [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: 04/24/2024] [Accepted: 06/26/2024] [Indexed: 06/28/2024]
Abstract
Objective.225Ac radiopharmaceuticals have tremendous potential for targeted alpha therapy, however,225Ac (t1/2= 9.9 d) lacks direct gamma emissions forin vivoimaging.226Ac (t1/2= 29.4 h) is a promising element-equivalent matched diagnostic radionuclide for preclinical evaluation of225Ac radiopharmaceuticals.226Ac has two gamma emissions (158 keV and 230 keV) suitable for SPECT imaging. This work is the first feasibility study forin vivoquantitative226Ac SPECT imaging and validation of activity estimation.Approach.226Ac was produced at TRIUMF (Vancouver, Canada) with its Isotope Separator and Accelerator (ISAC) facility. [226Ac]Ac3+was radiolabelled with the bioconjugate crown-TATE developed for therapeutic targeting of neuroendocrine tumours. Mice with AR42J tumour xenografts were injected with either 2 MBq of [226Ac]Ac-crown-TATE or 4 MBq of free [226Ac]Ac3+activity and were scanned at 1, 2.5, 5, and 24 h post injection in a preclinical microSPECT/CT. Quantitative SPECT images were reconstructed from the 158 keV and 230 keV photopeaks with attenuation, background, and scatter corrections. Image-based226Ac activity measurements were assessed from volumes of interest within tumours and organs of interest. Imaging data was compared withex vivobiodistribution measured via gamma counter.Main results. We present, to the best of our knowledge, the first everin vivoquantitative SPECT images of226Ac activity distributions. Time-activity curves derived from SPECT images quantify thein vivobiodistribution of [226Ac]Ac-crown-TATE and free [226Ac]Ac3+activity. Image-based activity measurements in the tumours and organs of interest corresponded well withex vivobiodistribution measurements.Significance. Here in, we established the feasibility ofin vivo226Ac quantitative SPECT imaging for accurate measurement of actinium biodistribution in a preclinical model. This imaging method could facilitate more efficient development of novel actinium labelled compounds by providing accurate quantitativein vivopharmacokinetic information essential for estimating toxicities, dosimetry, and therapeutic potency.
Collapse
Affiliation(s)
- Helena Koniar
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada
- Department of Physics and Astronomy, University of British Columbia, 6224 Agricultural Road, Vancouver, BC V6T 1Z1, Canada
| | - Luke Wharton
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada
| | - Aidan Ingham
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada
| | - Cristina Rodríguez-Rodríguez
- Department of Physics and Astronomy, University of British Columbia, 6224 Agricultural Road, Vancouver, BC V6T 1Z1, Canada
- Faculty of Pharmaceutical Sciences, University of British Columbia, 2405 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada
| | - Peter Kunz
- Accelerator Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada
- Department of Chemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC V5A 1S6, 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, 8888 University Drive, Burnaby, BC V5A 1S6, Canada
| | - Arman Rahmim
- Department of Physics and Astronomy, University of British Columbia, 6224 Agricultural Road, Vancouver, BC V6T 1Z1, Canada
- Department of Radiology, University of British Columbia, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada
- Department of Integrative Oncology, BC Cancer Research Centre, 675 W 10th Ave, Vancouver, BC, V5Z 1L3, Canada
| | - Carlos Uribe
- Department of Radiology, University of British Columbia, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada
- Department of Integrative Oncology, BC Cancer Research Centre, 675 W 10th Ave, Vancouver, BC, V5Z 1L3, Canada
- Department of Functional Imaging, BC Cancer, 600 West 10th Avenue, Vancouver, BC, V5Z 4E6, Canada
| | - Paul Schaffer
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada
- Department of Chemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC V5A 1S6, Canada
- Department of Radiology, University of British Columbia, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada
| |
Collapse
|
21
|
Asmundo L, Ambrosini V, Mojtahed A, Fanti S, Ferrone C, Hesami M, Sertic M, Najmi Z, Furtado FS, Dhami RS, Anderson MA, Samir A, Sharma A, Campana D, Ursprung S, Nikolau K, Domachevsky L, Blake MA, Norris EC, Clark JW, Catalano OA. Imaging of Neuroendocrine Neoplasms; Principles of Treatment Strategies. What Referring Clinicians Want to Know. J Comput Assist Tomogr 2024; 48:628-639. [PMID: 38626751 DOI: 10.1097/rct.0000000000001619] [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: 04/18/2024]
Abstract
ABSTRACT Neuroendocrine neoplasms (NENs) are a diverse group of tumors that express neuroendocrine markers and primarily affect the lungs and digestive system. The incidence of NENs has increased over time due to advancements in imaging and diagnostic techniques. Effective management of NENs requires a multidisciplinary approach, considering factors such as tumor location, grade, stage, symptoms, and imaging findings. Treatment strategies vary depending on the specific subtype of NEN. In this review, we will focus on treatment strategies and therapies including the information relevant to clinicians in order to undertake optimal management and treatment decisions, the implications of different therapies on imaging, and how to ascertain their possible complications and treatment effects.
Collapse
Affiliation(s)
| | | | - Amirkasra Mojtahed
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | | | - Cristina Ferrone
- Department of Surgery, Cedar-Sinai Health System, Los Angeles, CA
| | - Mina Hesami
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Madeleine Sertic
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Zahra Najmi
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Felipe S Furtado
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Ranjodh S Dhami
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Mark A Anderson
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Anthony Samir
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Amita Sharma
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Davide Campana
- Department of Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
| | - Stephan Ursprung
- Department of Radiology, University Hospital Tuebingen, Tuebingen, Germany
| | - Konstantin Nikolau
- Department of Radiology, University Hospital Tuebingen, Tuebingen, Germany
| | - Liran Domachevsky
- Department of Nuclear Medicine, The Chaim Sheba Medical Center, Tel Hashomer, Israel
| | - Michael A Blake
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Evan C Norris
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Jeffrey W Clark
- Department of Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Onofrio A Catalano
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
22
|
Bhimaniya S, Shah H, Jacene HA. Alpha-emitter Peptide Receptor Radionuclide Therapy in Neuroendocrine Tumors. PET Clin 2024; 19:341-349. [PMID: 38658229 DOI: 10.1016/j.cpet.2024.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Peptide receptor radionuclide therapy (PRRT) has become mainstream therapy of metastatic neuroendocrine tumors not controlled by somatostatin analog therapy. Currently, beta particle-emitting radiopharmaceuticals are the mainstay of PRRT. Alpha particle-emitting radiopharmaceuticals have a theoretic advantage over beta emitters in terms of improved therapeutic efficacy due to higher cancer cell death and lower nontarget tissue radiation-induced adverse events due to shorter path length of alpha particles. We discuss the available evidence for and the role of alpha particle PRRT.
Collapse
Affiliation(s)
- Sudhir Bhimaniya
- Harvard Medical School, Department of Radiology, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115, USA.
| | - Hina Shah
- Harvard Medical School, Department of Radiology, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115, USA
| | - Heather A Jacene
- Harvard Medical School, Department of Radiology, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115, USA
| |
Collapse
|
23
|
Zhang XB, Fan YB, Jing R, Getu MA, Chen WY, Zhang W, Dong HX, Dakal TC, Hayat A, Cai HJ, Ashrafizadeh M, Abd El-Aty AM, Hacimuftuoglu A, Liu P, Li TF, Sethi G, Ahn KS, Ertas YN, Chen MJ, Ji JS, Ma L, Gong P. Gastroenteropancreatic neuroendocrine neoplasms: current development, challenges, and clinical perspectives. Mil Med Res 2024; 11:35. [PMID: 38835066 DOI: 10.1186/s40779-024-00535-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 05/07/2024] [Indexed: 06/06/2024] Open
Abstract
Neuroendocrine neoplasms (NENs) are highly heterogeneous and potentially malignant tumors arising from secretory cells of the neuroendocrine system. Gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs) are the most common subtype of NENs. Historically, GEP-NENs have been regarded as infrequent and slow-growing malignancies; however, recent data have demonstrated that the worldwide prevalence and incidence of GEP-NENs have increased exponentially over the last three decades. In addition, an increasing number of studies have proven that GEP-NENs result in a limited life expectancy. These findings suggested that the natural biology of GEP-NENs is more aggressive than commonly assumed. Therefore, there is an urgent need for advanced researches focusing on the diagnosis and management of patients with GEP-NENs. In this review, we have summarized the limitations and recent advancements in our comprehension of the epidemiology, clinical presentations, pathology, molecular biology, diagnosis, and treatment of GEP-NETs to identify factors contributing to delays in diagnosis and timely treatment of these patients.
Collapse
Affiliation(s)
- Xian-Bin Zhang
- Department of General SurgeryInstitute of Precision Diagnosis and Treatment of Digestive System Tumors and Guangdong Provincial Key Laboratory of Chinese Medicine Ingredients and Gut Microbiomics, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, 518055, China
| | - Yi-Bao Fan
- Department of General SurgeryInstitute of Precision Diagnosis and Treatment of Digestive System Tumors and Guangdong Provincial Key Laboratory of Chinese Medicine Ingredients and Gut Microbiomics, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, 518055, China
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, 518060, China
| | - Rui Jing
- Department of Radiology, Second Hospital of Shandong University, Jinan, Shandong, 250000, China
| | - Mikiyas Amare Getu
- Department of General SurgeryInstitute of Precision Diagnosis and Treatment of Digestive System Tumors and Guangdong Provincial Key Laboratory of Chinese Medicine Ingredients and Gut Microbiomics, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, 518055, China
| | - Wan-Ying Chen
- Department of General SurgeryInstitute of Precision Diagnosis and Treatment of Digestive System Tumors and Guangdong Provincial Key Laboratory of Chinese Medicine Ingredients and Gut Microbiomics, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, 518055, China
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, 518060, China
| | - Wei Zhang
- Department of General SurgeryInstitute of Precision Diagnosis and Treatment of Digestive System Tumors and Guangdong Provincial Key Laboratory of Chinese Medicine Ingredients and Gut Microbiomics, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, 518055, China
| | - Hong-Xia Dong
- Department of Gastroenterology, General Hospital of Chinese PLA, Beijing, 100853, China
| | - Tikam Chand Dakal
- Department of Biotechnology, Mohanlal Sukhadia University, Udaipur, Rajasthan, 313001, India
| | - Akhtar Hayat
- Interdisciplinary Research Centre in Biomedical Materials (IRCBM), COMSATS University Islamabad, Lahore Campus, Lahore, 54000, Pakistan
| | - Hua-Jun Cai
- Department of General SurgeryInstitute of Precision Diagnosis and Treatment of Digestive System Tumors and Guangdong Provincial Key Laboratory of Chinese Medicine Ingredients and Gut Microbiomics, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, 518055, China
| | - Milad Ashrafizadeh
- Department of General SurgeryInstitute of Precision Diagnosis and Treatment of Digestive System Tumors and Guangdong Provincial Key Laboratory of Chinese Medicine Ingredients and Gut Microbiomics, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, 518055, China
| | - A M Abd El-Aty
- Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
- Department of Medical Pharmacology, Medical Faculty, Ataturk University, Erzurum, 25240, Turkey
| | - Ahmet Hacimuftuoglu
- Department of Medical Pharmacology, Medical Faculty, Ataturk University, Erzurum, 25240, Turkey
| | - Peng Liu
- Department of General SurgeryInstitute of Precision Diagnosis and Treatment of Digestive System Tumors and Guangdong Provincial Key Laboratory of Chinese Medicine Ingredients and Gut Microbiomics, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, 518055, China
| | - Tian-Feng Li
- Reproductive Medicine Center, Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong, 518055, China
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
| | - Kwang Seok Ahn
- Department of Science in Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Yavuz Nuri Ertas
- ERNAM-Nanotechnology Research and Application Center, Erciyes University, Kayseri, 38039, Türkiye
- Department of Biomedical Engineering, Erciyes University, Kayseri, 38280, Türkiye
- UNAM-National Nanotechnology Research Center, Bilkent University, Ankara, 06800, Türkiye
| | - Min-Jiang Chen
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, 323000, China
| | - Jian-Song Ji
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, 323000, China
| | - Li Ma
- Department of Epidemiology, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Peng Gong
- Department of General SurgeryInstitute of Precision Diagnosis and Treatment of Digestive System Tumors and Guangdong Provincial Key Laboratory of Chinese Medicine Ingredients and Gut Microbiomics, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, 518055, China.
| |
Collapse
|
24
|
Yang H, Zhang Y, Li H, Zhang Y, Feng Y, Yang X, Chen Y. Efficacy and Safety of 225 Ac-DOTATATE in the Treatment of Neuroendocrine Neoplasms With High SSTR Expression. Clin Nucl Med 2024; 49:505-512. [PMID: 38498615 DOI: 10.1097/rlu.0000000000005149] [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: 03/20/2024]
Abstract
PURPOSE We aimed to evaluate the efficacy and safety of 225 Ac-DOTATATE targeted α therapy (TAT) in various neuroendocrine neoplasms (NENs) with high somatostatin receptor (SSTR) expression. PATIENTS AND METHODS This single-center prospective study included 10 patients with histologically diagnosed NENs that exhibited increased SSTR expression on 68 Ga-DOTATATE PET/CT imaging. All patients received 225 Ac-DOTATATE TAT. The primary end points were molecular imaging-based response and disease control rate (DCR), measured using the slightly modified Positron Emission Tomography Response Criteria in Solid Tumors 1.0. The secondary end points were adverse event profiles and clinical responses. The adverse event profile was determined according to the Common Terminology Criteria for Adverse Events version 5.0. Clinical response was assessed using the EORTC QLQ-C30 v3.0 (European Organization for Research and Treatment of Cancer Core Quality of Life questionnaire version 3.0). RESULTS A molecular imaging-based partial response was observed in 40% of all patients, SD in 40%, PD in 20%, and DCR in 80%. The DCR was 83.3% (5/6) in patients who were previously treated with 177 Lu-DOTATATE. According to the EORTC QLQ-C30 v3.0 score, most symptoms improved after 225 Ac-DOTATATE treatment, with only diarrhea showing no improvement. Grade III/IV hematological, kidney, and liver toxicities were not observed. The median follow-up time was 14 months (7-22 months), and no deaths were reported. CONCLUSIONS This initial study suggests that 225 Ac-DOTATATE is a potentially promising option for treating NENs with elevated SSTR expression, with an acceptable toxicity profile and well-tolerated adverse effects.
Collapse
Affiliation(s)
| | | | | | | | | | - Xiqun Yang
- Department of Dermatology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | | |
Collapse
|
25
|
Gao J, Li M, Yin J, Liu M, Wang H, Du J, Li J. The Different Strategies for the Radiolabeling of [ 211At]-Astatinated Radiopharmaceuticals. Pharmaceutics 2024; 16:738. [PMID: 38931860 PMCID: PMC11206656 DOI: 10.3390/pharmaceutics16060738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 05/10/2024] [Accepted: 05/22/2024] [Indexed: 06/28/2024] Open
Abstract
Astatine-211 (211At) has emerged as a promising radionuclide for targeted alpha therapy of cancer by virtue of its favorable nuclear properties. However, the limited in vivo stability of 211At-labeled radiopharmaceuticals remains a major challenge. This review provides a comprehensive overview of the current strategies for 211At radiolabeling, including nucleophilic and electrophilic substitution reactions, as well as the recent advances in the development of novel bifunctional coupling agents and labeling approaches to enhance the stability of 211At-labeled compounds. The preclinical and clinical applications of 211At-labeled radiopharmaceuticals, including small molecules, peptides, and antibodies, are also discussed. Looking forward, the identification of new molecular targets, the optimization of 211At production and quality control methods, and the continued evaluation of 211At-labeled radiopharmaceuticals in preclinical and clinical settings will be the key to realizing the full potential of 211At-based targeted alpha therapy. With the growing interest and investment in this field, 211At-labeled radiopharmaceuticals are poised to play an increasingly important role in future cancer treatment.
Collapse
Affiliation(s)
- Jie Gao
- China Institute for Radiation Protection, National Atomic Energy Agency Nuclear Technology (Nonclinical Evaluation of Radiopharmaceuticals) Research and Development Center, CNNC Key Laboratory on Radiotoxicology and Radiopharmaceutical Preclinical Evaluation, Taiyuan 030006, China; (J.G.); (M.L.); (J.Y.); (M.L.)
- China Institute of Atomic Energy, Beijing 102413, China;
| | - Mei Li
- China Institute for Radiation Protection, National Atomic Energy Agency Nuclear Technology (Nonclinical Evaluation of Radiopharmaceuticals) Research and Development Center, CNNC Key Laboratory on Radiotoxicology and Radiopharmaceutical Preclinical Evaluation, Taiyuan 030006, China; (J.G.); (M.L.); (J.Y.); (M.L.)
| | - Jingjing Yin
- China Institute for Radiation Protection, National Atomic Energy Agency Nuclear Technology (Nonclinical Evaluation of Radiopharmaceuticals) Research and Development Center, CNNC Key Laboratory on Radiotoxicology and Radiopharmaceutical Preclinical Evaluation, Taiyuan 030006, China; (J.G.); (M.L.); (J.Y.); (M.L.)
| | - Mengya Liu
- China Institute for Radiation Protection, National Atomic Energy Agency Nuclear Technology (Nonclinical Evaluation of Radiopharmaceuticals) Research and Development Center, CNNC Key Laboratory on Radiotoxicology and Radiopharmaceutical Preclinical Evaluation, Taiyuan 030006, China; (J.G.); (M.L.); (J.Y.); (M.L.)
- China Institute of Atomic Energy, Beijing 102413, China;
| | - Hongliang Wang
- First Hospital of Shanxi Medical University, Taiyuan 030001, China;
| | - Jin Du
- China Institute of Atomic Energy, Beijing 102413, China;
- China Isotope & Radiation Corporation, Beijing 100089, China
| | - Jianguo Li
- China Institute for Radiation Protection, National Atomic Energy Agency Nuclear Technology (Nonclinical Evaluation of Radiopharmaceuticals) Research and Development Center, CNNC Key Laboratory on Radiotoxicology and Radiopharmaceutical Preclinical Evaluation, Taiyuan 030006, China; (J.G.); (M.L.); (J.Y.); (M.L.)
| |
Collapse
|
26
|
Di Franco M, Zanoni L, Fortunati E, Fanti S, Ambrosini V. Radionuclide Theranostics in Neuroendocrine Neoplasms: An Update. Curr Oncol Rep 2024; 26:538-550. [PMID: 38581469 PMCID: PMC11063107 DOI: 10.1007/s11912-024-01526-5] [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] [Accepted: 03/26/2024] [Indexed: 04/08/2024]
Abstract
PURPOSE OF REVIEW This paper aims to address the latest findings in neuroendocrine tumor (NET) theranostics, focusing on new evidence and future directions of combined diagnosis with positron emission tomography (PET) and treatment with peptide receptor radionuclide therapy (PRRT). RECENT FINDINGS Following NETTER-1 trial, PRRT with [177Lu]Lu-DOTATATE was approved by FDA and EMA and is routinely employed in advanced G1 and G2 SST (somatostatin receptor)-expressing NET. Different approaches have been proposed so far to improve the PRRT therapeutic index, encompassing re-treatment protocols, combinations with other therapies and novel indications. Molecular imaging holds a potential added value in characterizing disease biology and heterogeneity using different radiopharmaceuticals (e.g., SST and FDG) and may provide predictive and prognostic parameters. Response assessment criteria are still an unmet need and new theranostic pairs showed preliminary encouraging results. PRRT for NET has become a paradigm of modern theranostics. PRRT holds a favorable toxicity profile, and it is associated with a prolonged time to progression, reduction of symptoms, and improved patients' quality of life. In light of further optimization, different new strategies have been investigated, along with the development of new radiopharmaceuticals.
Collapse
Affiliation(s)
- Martina Di Franco
- Nuclear Medicine, Alma Mater Studiorum, University of Bologna, Via Massarenti 9, 40138, Bologna, Italy.
| | - Lucia Zanoni
- Nuclear Medicine, IRCCS, Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
| | - Emilia Fortunati
- Nuclear Medicine, IRCCS, Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
| | - Stefano Fanti
- Nuclear Medicine, Alma Mater Studiorum, University of Bologna, Via Massarenti 9, 40138, Bologna, Italy
- Nuclear Medicine, IRCCS, Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
| | - Valentina Ambrosini
- Nuclear Medicine, Alma Mater Studiorum, University of Bologna, Via Massarenti 9, 40138, Bologna, Italy
- Nuclear Medicine, IRCCS, Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
| |
Collapse
|
27
|
Taunk NK, Escorcia FE, Lewis JS, Bodei L. Radiopharmaceuticals for Cancer Diagnosis and Therapy: New Targets, New Therapies-Alpha-Emitters, Novel Targets. Cancer J 2024; 30:218-223. [PMID: 38753757 PMCID: PMC11232930 DOI: 10.1097/ppo.0000000000000720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2024]
Abstract
ABSTRACT Radiopharmaceutical therapy has emerged as a promising approach for the treatment of various cancers. The exploration of novel targets such as tumor-specific antigens, overexpressed receptors, and intracellular biomolecules using antibodies, peptides, or small molecules has expanded the scope of radiopharmaceutical therapy, enabling precise and effective cancer treatment for an increasing number of tumor types. Alpha emitters, characterized by their high linear energy transfer and short path length, offer unique advantages in targeted therapy due to their potent cytotoxicity against cancer cells while sparing healthy tissues. This article reviews recent advancements in identifying novel targets for radiopharmaceutical therapy and applications in utilizing α-emitters for targeted treatment.
Collapse
Affiliation(s)
- Neil K. Taunk
- Department of Radiation Oncology and Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Freddy E. Escorcia
- Molecular Imaging Branch, Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Jason S. Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Lisa Bodei
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
28
|
Lawal IO, Abubakar SO, Ndlovu H, Mokoala KMG, More SS, Sathekge MM. Advances in Radioligand Theranostics in Oncology. Mol Diagn Ther 2024; 28:265-289. [PMID: 38555542 DOI: 10.1007/s40291-024-00702-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/06/2024] [Indexed: 04/02/2024]
Abstract
Theranostics with radioligands (radiotheranostics) has played a pivotal role in oncology. Radiotheranostics explores the molecular targets expressed on tumor cells to target them for imaging and therapy. In this way, radiotheranostics entails non-invasive demonstration of the in vivo expression of a molecular target of interest through imaging followed by the administration of therapeutic radioligand targeting the tumor-expressed molecular target. Therefore, radiotheranostics ensures that only patients with a high likelihood of response are treated with a particular radiotheranostic agent, ensuring the delivery of personalized care to cancer patients. Within the last decades, a couple of radiotheranostics agents, including Lutetium-177 DOTATATE (177Lu-DOTATATE) and Lutetium-177 prostate-specific membrane antigen (177Lu-PSMA), were shown to prolong the survival of cancer patients compared to the current standard of care leading to the regulatory approval of these agents for routine use in oncology care. This recent string of successful approvals has broadened the interest in the development of different radiotheranostic agents and their investigation for clinical translation. In this work, we present an updated appraisal of the literature, reviewing the recent advances in the use of established radiotheranostic agents such as radioiodine for differentiated thyroid carcinoma and Iodine-131-labeled meta-iodobenzylguanidine therapy of tumors of the sympathoadrenal axis as well as the recently approved 177Lu-DOTATATE and 177Lu-PSMA for differentiated neuroendocrine tumors and advanced prostate cancer, respectively. We also discuss the radiotheranostic agents that have been comprehensively characterized in preclinical studies and have shown some clinical evidence supporting their safety and efficacy, especially those targeting fibroblast activation protein (FAP) and chemokine receptor 4 (CXCR4) and those still being investigated in preclinical studies such as those targeting poly (ADP-ribose) polymerase (PARP) and epidermal growth factor receptor 2.
Collapse
Affiliation(s)
- Ismaheel O Lawal
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology and Imaging Sciences, Emory University, 1364 Clifton Road, NE, Atlanta, GA, 30322, USA.
- Department of Nuclear Medicine, University of Pretoria, Pretoria, 0001, South Africa.
| | - Sofiullah O Abubakar
- Department of Radiology and Nuclear Medicine, Sultan Qaboos Comprehensive Cancer Care and Research Center, Muscat, Oman
| | - Honest Ndlovu
- Department of Nuclear Medicine, University of Pretoria, Pretoria, 0001, South Africa
- Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria, 0001, South Africa
| | - Kgomotso M G Mokoala
- Department of Nuclear Medicine, University of Pretoria, Pretoria, 0001, South Africa
- Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria, 0001, South Africa
| | - Stuart S More
- Department of Nuclear Medicine, University of Pretoria, Pretoria, 0001, South Africa
- Division of Nuclear Medicine, Department of Radiation Medicine, University of Cape Town, Cape Town, 7700, South Africa
| | - Mike M Sathekge
- Department of Nuclear Medicine, University of Pretoria, Pretoria, 0001, South Africa
- Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria, 0001, South Africa
| |
Collapse
|
29
|
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] [MESH Headings] [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
|
30
|
Gape PMD, Schultz MK, Stasiuk GJ, Terry SYA. Towards Effective Targeted Alpha Therapy for Neuroendocrine Tumours: A Review. Pharmaceuticals (Basel) 2024; 17:334. [PMID: 38543120 PMCID: PMC10974115 DOI: 10.3390/ph17030334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 02/22/2024] [Accepted: 02/26/2024] [Indexed: 04/01/2024] Open
Abstract
This review article explores the evolving landscape of Molecular Radiotherapy (MRT), emphasizing Peptide Receptor Radionuclide Therapy (PRRT) for neuroendocrine tumours (NETs). The primary focus is on the transition from β-emitting radiopharmaceuticals to α-emitting agents in PRRT, offering a critical analysis of the radiobiological basis, clinical applications, and ongoing developments in Targeted Alpha Therapy (TAT). Through an extensive literature review, the article delves into the mechanisms and effectiveness of PRRT in targeting somatostatin subtype 2 receptors, highlighting both its successes and limitations. The discussion extends to the emerging paradigm of TAT, underlining its higher potency and specificity with α-particle emissions, which promise enhanced therapeutic efficacy and reduced toxicity. The review critically evaluates preclinical and clinical data, emphasizing the need for standardised dosimetry and a deeper understanding of the dose-response relationship in TAT. The review concludes by underscoring the significant potential of TAT in treating SSTR2-overexpressing cancers, especially in patients refractory to β-PRRT, while also acknowledging the current challenges and the necessity for further research to optimize treatment protocols.
Collapse
Affiliation(s)
- Paul M. D. Gape
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London SE1 7EP, UK; (G.J.S.); (S.Y.A.T.)
| | - Michael K. Schultz
- Departments of Radiology, Radiation Oncology, Free Radical and Radiation Biology Program, University of Iowa, Iowa City, IA 52242, USA;
- Perspective Therapeutics, Coralville, IA 52241, USA
| | - Graeme J. Stasiuk
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London SE1 7EP, UK; (G.J.S.); (S.Y.A.T.)
| | - Samantha Y. A. Terry
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London SE1 7EP, UK; (G.J.S.); (S.Y.A.T.)
| |
Collapse
|
31
|
Lee D, Li M, Liu D, Baumhover NJ, Sagastume EA, Marks BM, Rastogi P, Pigge FC, Menda Y, Johnson FL, Schultz MK. Structural modifications toward improved lead-203/lead-212 peptide-based image-guided alpha-particle radiopharmaceutical therapies for neuroendocrine tumors. Eur J Nucl Med Mol Imaging 2024; 51:1147-1162. [PMID: 37955792 PMCID: PMC10881741 DOI: 10.1007/s00259-023-06494-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 10/26/2023] [Indexed: 11/14/2023]
Abstract
PURPOSE The lead-203 (203Pb)/lead-212 (212Pb) elementally identical radionuclide pair has gained significant interest in the field of image-guided targeted alpha-particle therapy for cancer. Emerging evidence suggests that 212Pb-labeled peptide-based radiopharmaceuticals targeting somatostatin receptor subtype 2 (SSTR2) may provide improved effectiveness compared to beta-particle-based therapies for neuroendocrine tumors (NETs). This study aims to improve the performance of SSTR2-targeted radionuclide imaging and therapy through structural modifications to Tyr3-octreotide (TOC)-based radiopharmaceuticals. METHODS New SSTR2-targeted peptides were designed and synthesized with the goal of optimizing the incorporation of Pb isotopes through the use of a modified cyclization technique; the introduction of a Pb-specific chelator (PSC); and the insertion of polyethylene glycol (PEG) linkers. The binding affinity of the peptides and the cellular uptake of 203Pb-labeled peptides were evaluated using pancreatic AR42J (SSTR2+) tumor cells and the biodistribution and imaging of the 203Pb-labeled peptides were assessed in an AR42J tumor xenograft mouse model. A lead peptide was identified (i.e., PSC-PEG2-TOC), which was then further evaluated for efficacy in 212Pb therapy studies. RESULTS The lead radiopeptide drug conjugate (RPDC) - [203Pb]Pb-PSC-PEG2-TOC - significantly improved the tumor-targeting properties, including receptor binding and tumor accumulation and retention as compared to [203Pb]Pb-DOTA0-Tyr3-octreotide (DOTATOC). Additionally, the modified RPDC exhibited faster renal clearance than the DOTATOC counterpart. These advantageous characteristics of [212Pb]Pb-PSC-PEG2-TOC resulted in a dose-dependent therapeutic effect with minimal signs of toxicity in the AR42J xenograft model. Fractionated administrations of 3.7 MBq [212Pb]Pb-PSC-PEG2-TOC over three doses further improved anti-tumor effectiveness, resulting in 80% survival (70% complete response) over 120 days in the mouse model. CONCLUSION Structural modifications to chelator and linker compositions improved tumor targeting and pharmacokinetics (PK) of 203/212Pb peptide-based radiopharmaceuticals for NET theranostics. These findings suggest that PSC-PEG2-TOC is a promising candidate for Pb-based targeted radionuclide therapy for NETs and other types of cancers that express SSTR2.
Collapse
Affiliation(s)
- Dongyoul Lee
- Department of Physics and Chemistry, Korea Military Academy, Seoul, Republic of Korea
| | - Mengshi Li
- Perspective Therapeutics, Inc., Coralville, IA, USA
| | - Dijie Liu
- Perspective Therapeutics, Inc., Coralville, IA, USA
| | | | | | | | - Prerna Rastogi
- Department of Pathology, The University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - F Christopher Pigge
- Department of Chemistry, The University of Iowa, ML B180 FRRBP, 500 Newton Road, Iowa City, IA, 52240, USA
| | - Yusuf Menda
- Department of Radiology, The University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | | | - Michael K Schultz
- Perspective Therapeutics, Inc., Coralville, IA, USA.
- Department of Chemistry, The University of Iowa, ML B180 FRRBP, 500 Newton Road, Iowa City, IA, 52240, USA.
- Department of Radiology, The University of Iowa Hospitals and Clinics, Iowa City, IA, USA.
- Department of Radiation Oncology, The University of Iowa Hospitals and Clinics, Iowa City, IA, USA.
| |
Collapse
|
32
|
Tikum AF, Ketchemen JP, Doroudi A, Nambisan AK, Babeker H, Njotu FN, Fonge H. Effectiveness of 225Ac-Labeled Anti-EGFR Radioimmunoconjugate in EGFR-Positive Kirsten Rat Sarcoma Viral Oncogene and BRAF Mutant Colorectal Cancer Models. J Nucl Med 2024:jnumed.123.266204. [PMID: 38360051 DOI: 10.2967/jnumed.123.266204] [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: 06/28/2023] [Revised: 01/05/2024] [Indexed: 02/17/2024] Open
Abstract
Eighty percent of colorectal cancers (CRCs) overexpress epidermal growth factor receptor (EGFR). Kirsten rat sarcoma viral oncogene (KRAS) mutations are present in 40% of CRCs and drive de novo resistance to anti-EGFR drugs. BRAF oncogene is mutated in 7%-10% of CRCs, with even worse prognosis. We have evaluated the effectiveness of [225Ac]Ac-macropa-nimotuzumab in KRAS mutant and in KRAS wild-type and BRAFV600E mutant EGFR-positive CRC cells in vitro and in vivo. Anti-CD20 [225Ac]Ac-macropa-rituximab was developed and used as a nonspecific radioimmunoconjugate. Methods: Anti-EGFR antibody nimotuzumab was radiolabeled with 225Ac via an 18-membered macrocyclic chelator p-SCN-macropa. The immunoconjugate was characterized using flow cytometry, radioligand binding assay, and high-performance liquid chromatography, and internalization was studied using live-cell imaging. In vitro cytotoxicity was evaluated in 2-dimensional monolayer EGFR-positive KRAS mutant DLD-1, SW620, and SNU-C2B; in KRAS wild-type and BRAFV600E mutant HT-29 CRC cell lines; and in 3-dimensional spheroids. Dosimetry was studied in healthy mice. The in vivo efficacy of [225Ac]Ac-macropa-nimotuzumab was evaluated in mice bearing DLD-1, SW620, and HT-29 xenografts after treatment with 3 doses of 13 kBq/dose administered 10 d apart. Results: In all cell lines, in vitro studies showed enhanced cytotoxicity of [225Ac]Ac-macropa-nimotuzumab compared with nimotuzumab and controls. The inhibitory concentration of 50% in the DLD-1 cell line was 1.8 nM for [225Ac]Ac-macropa-nimotuzumab versus 84.1 nM for nimotuzumab. Similarly, the inhibitory concentration of 50% was up to 79-fold lower for [225Ac]Ac-macropa-nimotuzumab than for nimotuzumab in KRAS mutant SNU-C2B and SW620 and in KRAS wild-type and BRAFV600E mutant HT-29 CRC cell lines. A similar trend was observed for 3-dimensional spheroids. Internalization peaked 24-48 h after incubation and depended on EGFR expression. In the [225Ac]Ac-macropa-nimotuzumab group, 3 of 7 mice bearing DLD-1 tumors had complete remission. Median survival was 40 and 34 d for mice treated with phosphate-buffered saline and [225Ac]Ac-macropa-rituximab (control), respectively, whereas it was not reached for the [225Ac]Ac-macropa-nimotuzumab group (>90 d). Similarly, median survival of mice bearing HT-29 xenografts was 16 and 12.5 d for those treated with [225Ac]Ac-macropa-rituximab and phosphate-buffered saline, respectively, and was not reached for those treated with [225Ac]Ac-macropa-nimotuzumab (>90 d). One of 7 mice bearing HT-29 xenografts and treated using [225Ac]Ac-macropa-nimotuzumab had complete remission. Compared with untreated mice, [225Ac]Ac-macropa-nimotuzumab more than doubled (16 vs. 41 d) the median survival of mice bearing SW620 xenografts. Conclusion: [225Ac]Ac-macropa-nimotuzumab is effective against KRAS mutant and BRAFV600E mutant CRC models.
Collapse
Affiliation(s)
- Anjong Florence Tikum
- Department of Medical Imaging, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Jessica P Ketchemen
- Department of Medical Imaging, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Alireza Doroudi
- Department of Medical Imaging, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Anand K Nambisan
- Department of Medical Imaging, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Hanan Babeker
- Department of Medical Imaging, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada; and
| | - Fabrice Ngoh Njotu
- Department of Medical Imaging, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Humphrey Fonge
- Department of Medical Imaging, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada;
- Department of Medical Imaging, Royal University Hospital Saskatoon, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
33
|
Zhang T, Lei H, Chen X, Dou Z, Yu B, Su W, Wang W, Jin X, Katsube T, Wang B, Zhang H, Li Q, Di C. Carrier systems of radiopharmaceuticals and the application in cancer therapy. Cell Death Discov 2024; 10:16. [PMID: 38195680 PMCID: PMC10776600 DOI: 10.1038/s41420-023-01778-3] [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: 10/17/2023] [Revised: 12/04/2023] [Accepted: 12/13/2023] [Indexed: 01/11/2024] Open
Abstract
Radiopharmaceuticals play a vital role in cancer therapy. The carrier of radiopharmaceuticals can precisely locate and guide radionuclides to the target, where radionuclides kill surrounding tumor cells. Effective application of radiopharmaceuticals depends on the selection of an appropriate carrier. Herein, different types of carriers of radiopharmaceuticals and the characteristics are briefly described. Subsequently, we review radiolabeled monoclonal antibodies (mAbs) and their derivatives, and novel strategies of radiolabeled mAbs and their derivatives in the treatment of lymphoma and colorectal cancer. Furthermore, this review outlines radiolabeled peptides, and novel strategies of radiolabeled peptides in the treatment of neuroendocrine neoplasms, prostate cancer, and gliomas. The emphasis is given to heterodimers, bicyclic peptides, and peptide-modified nanoparticles. Last, the latest developments and applications of radiolabeled nucleic acids and small molecules in cancer therapy are discussed. Thus, this review will contribute to a better understanding of the carrier of radiopharmaceuticals and the application in cancer therapy.
Collapse
Affiliation(s)
- Taotao Zhang
- Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China
- College of Life Sciences, University of Chinese Academy of Sciences, 101408, Beijing, China
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, 101408, Beijing, China
| | - Huiwen Lei
- Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China
- College of Life Sciences, University of Chinese Academy of Sciences, 101408, Beijing, China
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, 101408, Beijing, China
| | - Xiaohua Chen
- Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China
- College of Life Sciences, University of Chinese Academy of Sciences, 101408, Beijing, China
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, 101408, Beijing, China
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, 516029, China
| | - Zhihui Dou
- Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China
- College of Life Sciences, University of Chinese Academy of Sciences, 101408, Beijing, China
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, 101408, Beijing, China
| | - Boyi Yu
- Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China
- College of Life Sciences, University of Chinese Academy of Sciences, 101408, Beijing, China
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, 101408, Beijing, China
| | - Wei Su
- Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China
- College of Life Sciences, University of Chinese Academy of Sciences, 101408, Beijing, China
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, 101408, Beijing, China
| | - Wei Wang
- College of Life Science, Northwest Normal University, Lanzhou, 730000, China
| | - Xiaodong Jin
- Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, 516029, China
| | - Takanori Katsube
- National Institute of Radiological Sciences, National Institutes for Quantum Science and Technology, Chiba, 263-8555, Japan
| | - Bing Wang
- National Institute of Radiological Sciences, National Institutes for Quantum Science and Technology, Chiba, 263-8555, Japan
| | - Hong Zhang
- Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China.
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China.
- College of Life Sciences, University of Chinese Academy of Sciences, 101408, Beijing, China.
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, 101408, Beijing, China.
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, 516029, China.
| | - Qiang Li
- Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China.
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China.
- College of Life Sciences, University of Chinese Academy of Sciences, 101408, Beijing, China.
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, 101408, Beijing, China.
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, 516029, China.
| | - Cuixia Di
- Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China.
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China.
- College of Life Sciences, University of Chinese Academy of Sciences, 101408, Beijing, China.
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, 101408, Beijing, China.
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, 516029, China.
| |
Collapse
|
34
|
Sedlack AJH, Meyer C, Mench A, Winters C, Barbon D, Obrzut S, Mallak N. Essentials of Theranostics: A Guide for Physicians and Medical Physicists. Radiographics 2024; 44:e230097. [PMID: 38060426 DOI: 10.1148/rg.230097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
Radiopharmaceutical therapies (RPTs) are gaining increased interest with the recent emergence of novel safe and effective theranostic agents, improving outcomes for thousands of patients. The term theranostics refers to the use of diagnostic and therapeutic agents that share the same molecular target; a major step toward precision medicine, especially for oncologic applications. The authors dissect the fundamentals of theranostics in nuclear medicine. First, they explain the radioactive decay schemes and the characteristics of emitted electromagnetic radiation used for imaging, as well as particles used for therapeutic purposes, followed by the interaction of the different types of radiation with tissue. These concepts directly apply to clinical RPTs and play a major role in the efficacy and toxicity profile of different radiopharmaceutical agents. Personalized dosimetry is a powerful tool that can help estimate patient-specific absorbed doses, in tumors as well as normal organs. Dosimetry in RPT is an area of active investigation, as most of what we know about the relationship between delivered dose and tissue damage is extrapolated from external-beam radiation therapy; more research is needed to understand this relationship as it pertains to RPTs. Tumor heterogeneity is increasingly recognized as an important prognostic factor. Novel molecular imaging agents, often in combination with fluorine 18-fluorodeoxyglucose, are crucial for assessment of target expression in the tumor and potential hypermetabolic disease that may lack the molecular target expression. ©RSNA, 2023 Test Your Knowledge questions are available in the supplemental material.
Collapse
Affiliation(s)
- Andrew J H Sedlack
- From the Medical Scientist Training Program, Feinberg School of Medicine, Northwestern University, Chicago, Ill (A.J.H.S.); and Department of Diagnostic Radiology, Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, L340, Portland, OR 97239-3098 (C.M., A.M., C.W., D.B., S.O., N.M.)
| | - Catherine Meyer
- From the Medical Scientist Training Program, Feinberg School of Medicine, Northwestern University, Chicago, Ill (A.J.H.S.); and Department of Diagnostic Radiology, Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, L340, Portland, OR 97239-3098 (C.M., A.M., C.W., D.B., S.O., N.M.)
| | - Anna Mench
- From the Medical Scientist Training Program, Feinberg School of Medicine, Northwestern University, Chicago, Ill (A.J.H.S.); and Department of Diagnostic Radiology, Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, L340, Portland, OR 97239-3098 (C.M., A.M., C.W., D.B., S.O., N.M.)
| | - Celeste Winters
- From the Medical Scientist Training Program, Feinberg School of Medicine, Northwestern University, Chicago, Ill (A.J.H.S.); and Department of Diagnostic Radiology, Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, L340, Portland, OR 97239-3098 (C.M., A.M., C.W., D.B., S.O., N.M.)
| | - Dennis Barbon
- From the Medical Scientist Training Program, Feinberg School of Medicine, Northwestern University, Chicago, Ill (A.J.H.S.); and Department of Diagnostic Radiology, Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, L340, Portland, OR 97239-3098 (C.M., A.M., C.W., D.B., S.O., N.M.)
| | - Sebastian Obrzut
- From the Medical Scientist Training Program, Feinberg School of Medicine, Northwestern University, Chicago, Ill (A.J.H.S.); and Department of Diagnostic Radiology, Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, L340, Portland, OR 97239-3098 (C.M., A.M., C.W., D.B., S.O., N.M.)
| | - Nadine Mallak
- From the Medical Scientist Training Program, Feinberg School of Medicine, Northwestern University, Chicago, Ill (A.J.H.S.); and Department of Diagnostic Radiology, Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, L340, Portland, OR 97239-3098 (C.M., A.M., C.W., D.B., S.O., N.M.)
| |
Collapse
|
35
|
Puranik AD, Choudhury S, Ghosh S, Dev ID, Ramchandani V, Uppal A, Bhosale V, Palsapure A, Rungta R, Pandey R, Khatri S, George G, Satamwar Y, Maske R, Agrawal A, Shah S, Purandare NC, Rangarajan V. Tata Memorial Centre Evidence Based Use of Nuclear medicine diagnostic and treatment modalities in cancer. Indian J Cancer 2024; 61:S1-S28. [PMID: 38424680 DOI: 10.4103/ijc.ijc_52_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 01/31/2024] [Indexed: 03/02/2024]
Abstract
ABSTRACT PET/CT and radioisotope therapy are diagnostic and therapeutic arms of Nuclear Medicine, respectively. With the emergence of better technology, PET/CT has become an accessible modality. Diagnostic tracers exploring disease-specific targets has led the clinicians to look beyond FDG PET. Moreover, with the emergence of theranostic pairs of radiopharmaceuticals, radioisotope therapy is gradually making it's way into treatment algorithm of common cancers in India. We therefore would like to discuss in detail the updates in PET/CT imaging and radionuclide therapy and generate a consensus-driven evidence based document which would guide the practitioners of Oncology.
Collapse
Affiliation(s)
- Ameya D Puranik
- Department of Nuclear Medicine and Molecular Imaging, Tata Memorial Hospital and Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Jalloul W, Ghizdovat V, Stolniceanu CR, Ionescu T, Grierosu IC, Pavaleanu I, Moscalu M, Stefanescu C. Targeted Alpha Therapy: All We Need to Know about 225Ac's Physical Characteristics and Production as a Potential Theranostic Radionuclide. Pharmaceuticals (Basel) 2023; 16:1679. [PMID: 38139806 PMCID: PMC10747780 DOI: 10.3390/ph16121679] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/24/2023] [Accepted: 11/30/2023] [Indexed: 12/24/2023] Open
Abstract
The high energy of α emitters, and the strong linear energy transfer that goes along with it, lead to very efficient cell killing through DNA damage. Moreover, the degree of oxygenation and the cell cycle state have no impact on these effects. Therefore, α radioisotopes can offer a treatment choice to individuals who are not responding to β- or gamma-radiation therapy or chemotherapy drugs. Only a few α-particle emitters are suitable for targeted alpha therapy (TAT) and clinical applications. The majority of available clinical research involves 225Ac and its daughter nuclide 213Bi. Additionally, the 225Ac disintegration cascade generates γ decays that can be used in single-photon emission computed tomography (SPECT) imaging, expanding the potential theranostic applications in nuclear medicine. Despite the growing interest in applying 225Ac, the restricted global accessibility of this radioisotope makes it difficult to conduct extensive clinical trials for many radiopharmaceutical candidates. To boost the availability of 225Ac, along with its clinical and potential theranostic applications, this review attempts to highlight the fundamental physical properties of this α-particle-emitting isotope, as well as its existing and possible production methods.
Collapse
Affiliation(s)
- Wael Jalloul
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- North East Regional Innovative Cluster for Structural and Molecular Imaging (Imago-Mol), 700115 Iasi, Romania
| | - Vlad Ghizdovat
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- North East Regional Innovative Cluster for Structural and Molecular Imaging (Imago-Mol), 700115 Iasi, Romania
| | - Cati Raluca Stolniceanu
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- North East Regional Innovative Cluster for Structural and Molecular Imaging (Imago-Mol), 700115 Iasi, Romania
| | - Teodor Ionescu
- Department of Morpho-Functional Sciences (Pathophysiology), “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Irena Cristina Grierosu
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Ioana Pavaleanu
- Department of Mother and Child, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Mihaela Moscalu
- Department of Preventive Medicine and Interdisciplinarity, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Cipriana Stefanescu
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- North East Regional Innovative Cluster for Structural and Molecular Imaging (Imago-Mol), 700115 Iasi, Romania
| |
Collapse
|
37
|
Sudo H, Tsuji AB, Sugyo A, Harada Y, Nagayama S, Katagiri T, Nakamura Y, Higashi T. Head-to-head comparison of three chelates reveals DOTAGA promising for 225 Ac labeling of anti-FZD10 antibody OTSA101. Cancer Sci 2023; 114:4677-4690. [PMID: 37781962 PMCID: PMC10728013 DOI: 10.1111/cas.15978] [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: 05/11/2023] [Revised: 09/04/2023] [Accepted: 09/12/2023] [Indexed: 10/03/2023] Open
Abstract
To select the most suitable chelate for 225 Ac radiolabeling of the anti-FZD10 antibody OTSA101, we directly compared three chelates: S-2-(4-isothiocyanatobenzyl)-1,4,7,10-tetraazacyclododecane tetraacetic acid (p-SCN-Bn-DOTA), 2,2',2″-(10-(1-carboxy-4-((4-isothiocyanatobenzyl)amino)-4-oxobutyl)-1,4,7,10-tetraazacyclododecane-1,4,7-triyl) triacetic acid (p-SCN-Bn-DOTAGA), and 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid mono-N-hydroxysuccinimide ester (DO3A-NHS-ester). We evaluated the binding affinity of the chelate-conjugated OTSA101 antibodies, as well as the labeling efficiency and stability in murine serum of 225 Ac-labeled OTSA101 as in vitro properties. The biodistribution, intratumoral distribution, absorbed doses, and therapeutic effects of the chelate-conjugated OTSA101 antibodies were assessed in the synovial sarcoma mouse model SYO-1. Of the three conjugates, DOTAGA conjugation had the smallest impact on the binding affinity (p < 0.01). The labeling efficiencies of DOTAGA-OTSA101 and DO3A-OTSA101 were 1.8-fold higher than that of DOTA-OTSA101 (p < 0.01). The stabilities were similar between 225 Ac-labeled DOTA-OTSA101, DOTAGA-OTSA101, and DO3A-OTSA101in serum at 37 and 4°C. The dosimetric analysis based on the biodistribution revealed significantly higher tumor-absorbed doses by 225 Ac-labeled DOTA-OTSA101 and DOTAGA-OTSA101 compared with 225 Ac-DO3A-OTSA101 (p < 0.05). 225 Ac-DOTAGA-OTSA101 exhibited the highest tumor-to-bone marrow ratio, with bone marrow being the dose-limiting tissue. The therapeutic and adverse effects were not significantly different between the three conjugates. Our findings indicate that among the three evaluated chelates, DOTAGA appears to be the most promising chelate to produce 225 Ac-labeled OTSA101 with high binding affinity and high radiochemical yields while providing high absorbed doses to tumors and limited absorbed doses to bone marrow.
Collapse
Affiliation(s)
- Hitomi Sudo
- Department of Molecular Imaging and TheranosticsNational Institutes for Quantum Science and Technology (QST)ChibaJapan
| | - Atsushi B. Tsuji
- Department of Molecular Imaging and TheranosticsNational Institutes for Quantum Science and Technology (QST)ChibaJapan
| | - Aya Sugyo
- Department of Molecular Imaging and TheranosticsNational Institutes for Quantum Science and Technology (QST)ChibaJapan
| | | | | | - Toyomasa Katagiri
- Division of Genome MedicineInstitute of Advanced Medical Sciences, Tokushima UniversityTokushimaJapan
- National Institutes of Biomedical Innovation, Health and NutritionIbarakiJapan
| | - Yusuke Nakamura
- National Institutes of Biomedical Innovation, Health and NutritionIbarakiJapan
| | - Tatsuya Higashi
- Department of Molecular Imaging and TheranosticsNational Institutes for Quantum Science and Technology (QST)ChibaJapan
| |
Collapse
|
38
|
Han G, Hwang E, Lin F, Clift R, Kim D, Guest M, Bischoff E, Moran S, Li G. RYZ101 (Ac-225 DOTATATE) Opportunity beyond Gastroenteropancreatic Neuroendocrine Tumors: Preclinical Efficacy in Small-Cell Lung Cancer. Mol Cancer Ther 2023; 22:1434-1443. [PMID: 37616528 DOI: 10.1158/1535-7163.mct-23-0029] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 04/28/2023] [Accepted: 08/21/2023] [Indexed: 08/26/2023]
Abstract
Overexpression of somatostatin receptors (SSTR), particularly SSTR2, is found in gastroenteropancreatic neuroendocrine tumors (GEP-NET), and subsets of other solid tumors such as small-cell lung cancer (SCLC). SCLC accounts for approximately 13% to 15% of lung cancer and lacks effective therapeutic options. IHC analysis indicates that up to 50% of SCLC tumors are SSTR2-positive, with a substantial subset showing high and homogenous expression. Peptide receptor radionuclide therapy with radiolabeled somatostatin analogue, Lu-177 DOTATATE, has been approved for GEP-NETs. Different strategies aimed at improving outcomes, such as the use of alpha-emitting radioisotopes, are currently being investigated. RYZ101 (Ac-225 DOTATATE) is comprised of the alpha-emitting radioisotope actinium-225, chemical chelator DOTA, and octreotate (TATE), a somatostatin analogue. In the cell-based competitive radioligand binding assay, RAYZ-10001-La (lanthanum surrogate for RYZ101) showed high binding affinity (Ki = 0.057 nmol/L) to human SSTR2 and >600-fold selectivity against other SSTR subtypes. RAYZ-10001-La exhibited efficient internalization to SSTR2-positive cells. In multiple SSTR2-expressing SCLC xenograft models, single-dose intravenous RYZ101 3 μCi (0.111 MBq) or 4 μCi (0.148 MBq) significantly inhibited tumor growth, with deeper responses, including sustained regression, observed in the models with higher SSTR2 levels. The antitumor effect was further enhanced when RYZ101 was combined with carboplatin and etoposide at clinically relevant doses. In summary, RYZ101 is a highly potent, alpha-emitting radiopharmaceutical agent, and preclinical data demonstrate the potential of RYZ101 for the treatment of patients with SSTR-positive cancers.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Gary Li
- RayzeBio, Inc., San Diego, California
| |
Collapse
|
39
|
Liatsou I, Josefsson A, Yu J, Li Z, Davis K, Brayton C, Wang H, Hobbs RF, Sgouros G. Early Normal Tissue Effects and Bone Marrow Relative Biological Effectiveness for an Actinium 225-Labeled HER2/neu-Targeting Antibody. Int J Radiat Oncol Biol Phys 2023; 117:1028-1037. [PMID: 37331568 DOI: 10.1016/j.ijrobp.2023.06.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 05/16/2023] [Accepted: 06/11/2023] [Indexed: 06/20/2023]
Abstract
PURPOSE In this study we determined the dose-independent relative biological effectiveness (RBE2) of bone marrow for an anti-HER2/neu antibody labeled with the alpha-particle emitter actinium 225 (225Ac). Hematologic toxicity is often a consequence of radiopharmaceutical therapy (RPT) administration, and dosimetric guidance to the bone marrow is required to limit toxicity. METHODS AND MATERIALS Female neu/N transgenic mice (MMTV-neu) were intravenously injected with 0 to 16.65 kBq of the alpha-particle emitter labeled antibody, 225Ac-DOTA-7.16.4, and euthanized at 1 to 9 days after treatment. Complete blood counts were performed. Femurs and tibias were collected, and bone marrow was isolated from 1 femur and tibia and counted for radioactivity. Contralateral intact femurs were fixed, decalcified, and assessed by histology. Marrow cellularity was the biologic endpoint selected for RBE2 determination. For the reference radiation, both femurs of the mice were photon irradiated with 0 to 5 Gy using a small animal radiation research platform. RESULTS Response as measured by cellularity for the alpha-particle emitter RPT (αRPT) RPT and the external beam radiation therapy were linear and linear quadratic, respectively, as a function of absorbed dose. The resulting dose-independent RBE2 for bone marrow was 6. CONCLUSIONS As αRPT gains prominence, preclinical studies evaluating RBE in vivo will be important in relating to human experience with beta-particle emitter RPT. Such normal tissue RBE evaluations will help mitigate unexpected toxicity in αRPT.
Collapse
Affiliation(s)
- Ioanna Liatsou
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Anders Josefsson
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jing Yu
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Zhi Li
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Kaori Davis
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Cory Brayton
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Hao Wang
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Robert F Hobbs
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - George Sgouros
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
40
|
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
|
41
|
Morgan KA, Rudd SE, Noor A, Donnelly PS. Theranostic Nuclear Medicine with Gallium-68, Lutetium-177, Copper-64/67, Actinium-225, and Lead-212/203 Radionuclides. Chem Rev 2023; 123:12004-12035. [PMID: 37796539 DOI: 10.1021/acs.chemrev.3c00456] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/06/2023]
Abstract
Molecular changes in malignant tissue can lead to an increase in the expression levels of various proteins or receptors that can be used to target the disease. In oncology, diagnostic imaging and radiotherapy of tumors is possible by attaching an appropriate radionuclide to molecules that selectively bind to these target proteins. The term "theranostics" describes the use of a diagnostic tool to predict the efficacy of a therapeutic option. Molecules radiolabeled with γ-emitting or β+-emitting radionuclides can be used for diagnostic imaging using single photon emission computed tomography or positron emission tomography. Radionuclide therapy of disease sites is possible with either α-, β-, or Auger-emitting radionuclides that induce irreversible damage to DNA. This Focus Review centers on the chemistry of theranostic approaches using metal radionuclides for imaging and therapy. The use of tracers that contain β+-emitting gallium-68 and β-emitting lutetium-177 will be discussed in the context of agents in clinical use for the diagnostic imaging and therapy of neuroendocrine tumors and prostate cancer. A particular emphasis is then placed on the chemistry involved in the development of theranostic approaches that use copper-64 for imaging and copper-67 for therapy with functionalized sarcophagine cage amine ligands. Targeted therapy with radionuclides that emit α particles has potential to be of particular use in late-stage disease where there are limited options, and the role of actinium-225 and lead-212 in this area is also discussed. Finally, we highlight the challenges that impede further adoption of radiotheranostic concepts while highlighting exciting opportunities and prospects.
Collapse
Affiliation(s)
- Katherine A Morgan
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Melbourne 3010, Australia
| | - Stacey E Rudd
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Melbourne 3010, Australia
| | - Asif Noor
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Melbourne 3010, Australia
| | - Paul S Donnelly
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Melbourne 3010, Australia
| |
Collapse
|
42
|
Turner JH. Cancer Care by Committee to be Superseded by Personal Physician-Patient Partnership Informed by Artificial Intelligence. Cancer Biother Radiopharm 2023; 38:497-505. [PMID: 37366774 DOI: 10.1089/cbr.2023.0058] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2023] Open
Abstract
Multidisciplinary tumor boards (MTBs) have become the reference standard of cancer management, founded upon randomized controlled trial (RCT) evidence-based guidelines. The inordinate delays inherent in awaiting formal regulatory agency approvals of novel therapeutic agents, and the rigidities and nongeneralizability of this regimented approach, often deny cancer patients timely access to effective innovative treatment. Reluctance of MTBs to accept theranostic care of patients with advanced neuroendocrine tumors (NETs) and metastatic castrate-resistant prostate cancer resulted in decades of delay in the incorporation of 177Lu-octreotate and 177Lu-prostate-specific membrane antigen (PSMA) into routine clinical oncology practice. Recent developments in immunotherapy and molecular targeted precision therapy, based on N-of-One individual multifactorial genome analyses, have greatly increased the complexity of decision-making. Burgeoning specialist workload and tight time frames now threaten to overwhelm the logistically, and emotionally, demanding MTB system. It is hypothesized that the advent of advanced artificial intelligence technology and Chatbot natural language algorithms will shift the cancer care paradigm from a MTB management model toward a personal physician-patient shared-care partnership for real-world practice of precision individualized holistic oncology.
Collapse
Affiliation(s)
- J Harvey Turner
- Department of Nuclear Medicine, Fiona Stanley Fremantle Hospitals Group, The University of Western Australia, Murdoch, Australia
| |
Collapse
|
43
|
Hull A, Hsieh W, Borysenko A, Tieu W, Bartholomeusz D, Bezak E. Development of [ 225Ac]Ac-DOTA-C595 as radioimmunotherapy of pancreatic cancer: in vitro evaluation, dosimetric assessment and detector calibration. EJNMMI Radiopharm Chem 2023; 8:22. [PMID: 37679594 PMCID: PMC10484829 DOI: 10.1186/s41181-023-00209-z] [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: 07/20/2023] [Accepted: 08/28/2023] [Indexed: 09/09/2023] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignancy which may benefit from radioimmunotherapy. Previously, [177Lu]Lu-DOTA-C595 has been developed as a beta-emitting radioimmunoconjugate to target cancer-specific mucin 1 epitopes (MUC1-CE) overexpressed on PDAC. However, the therapeutic effect may be enhanced by using an alpha-emitting radionuclide such as Actinium-225 (Ac-225). The short range and high linear energy transfer of alpha particles provides dense cellular damage and can overcome typical barriers related to PDAC treatment such as hypoxia. Despite the added cytotoxicity of alpha-emitters, their clinical implementation can be complicated by their complex decay chains, recoil energy and short-range impeding radiation detection. In this study, we developed and evaluated [225Ac]Ac-DOTA-C595 as an alpha-emitting radioimmunotherapy against PDAC using a series of in vitro experiments and conducted a preliminary dosimetric assessment and cross-calibration of detectors for the clinical implementation of Ac-225. RESULTS Cell binding and internalisation of [225Ac]Ac-DOTA-C595 was rapid and greatest in cells with strong MUC1-CE expression. Over 99% of PDAC cells had positive yH2AX expression within 1 h of [225Ac]Ac-DOTA-C595 exposure, suggesting a high level of DNA damage. Clonogenic assays further illustrated the cytotoxicity of [225Ac]Ac-DOTA-C595 in a concentration-dependent manner. At low concentrations of [225Ac]Ac-DOTA-C595, cells with strong MUC1-CE expression had lower cell survival than cells with weak MUC1-CE expression, yet survival was similar between cell lines at high concentrations irrespective of MUC1-CE expression. A dosimetric assessment was performed to estimate the dose-rate of 1 kBq of [225Ac]Ac-DOTA-C595 with consideration to alpha particles. Total absorption of 1 kBq of Ac-225 was estimated to provide a dose rate of 17.5 mGy/h, confirmed via both detector measurements and calculations. CONCLUSION [225Ac]Ac-DOTA-C595 was shown to target and induce a therapeutic effect in MUC1-CE expressing PDAC cells.
Collapse
Affiliation(s)
- Ashleigh Hull
- Allied Health and Human Performance Academic Unit, University of South Australia, City East Campus, Cnr North Tce and Frome Road, Adelaide, SA, 5001, Australia.
- Department of PET, Nuclear Medicine & Bone Densitometry, Royal Adelaide Hospital, SA Medical Imaging, Adelaide, SA, 5000, Australia.
| | - William Hsieh
- Allied Health and Human Performance Academic Unit, University of South Australia, City East Campus, Cnr North Tce and Frome Road, Adelaide, SA, 5001, Australia
- Department of PET, Nuclear Medicine & Bone Densitometry, Royal Adelaide Hospital, SA Medical Imaging, Adelaide, SA, 5000, Australia
| | - Artem Borysenko
- Radiation Protection Branch, South Australian Environment Protection Authority, Adelaide, SA, 5000, Australia
| | - William Tieu
- School of Physical Sciences, The University of Adelaide, Adelaide, SA, 5000, Australia
| | - Dylan Bartholomeusz
- Department of PET, Nuclear Medicine & Bone Densitometry, Royal Adelaide Hospital, SA Medical Imaging, Adelaide, SA, 5000, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, 5000, Australia
| | - Eva Bezak
- Allied Health and Human Performance Academic Unit, University of South Australia, City East Campus, Cnr North Tce and Frome Road, Adelaide, SA, 5001, Australia
- School of Physical Sciences, The University of Adelaide, Adelaide, SA, 5000, Australia
| |
Collapse
|
44
|
Beydağı G, Alan Selçuk N, Kabasakal L. Alpha Peptide Receptor Radionuclide Therapy in Neuroendocrine Tumors. NUCLEAR MEDICINE SEMINARS 2023; 9:109-115. [DOI: 10.4274/nts.galenos.2023.0015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
45
|
Su D, Yang H, Qiu C, Chen Y. Peptide receptor radionuclide therapy in advanced Pheochromocytomas and Paragangliomas: a systematic review and meta-analysis. Front Oncol 2023; 13:1141648. [PMID: 37483516 PMCID: PMC10358840 DOI: 10.3389/fonc.2023.1141648] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 06/20/2023] [Indexed: 07/25/2023] Open
Abstract
Objective Peptide receptor radionuclide therapy (PRRT) for advanced pheochromocytomas and paragangliomas (PPGLs) has received increasing attention. The purpose of this article is to evaluate the efficacy and safety of PRRT in patients with metastatic or inoperable PPGLs by meta-analysis. Methods A literature search was conducted in PubMed, Embase, Scopus, and Cochrane Library databases up to November 2022. All articles on PRRT for PPGLs were searched, and appropriate data were included for analysis. The measures evaluated included objective response rate (ORR), disease control rate (DCR), clinical response rate, biochemical response rate, progression-free survival (PFS), overall survival (OS), and adverse events. Statistical analysis was performed using Stata 16.0 and the R programming language, data were combined using a random-effects model, and the results were presented using forest plots. Results A total of 20 studies with 330 patients were included in the analysis. The results showed that ORR and DCR were 20.0% (95% CI: 12.0%-28.0%) and 90.0% (95% CI: 85.0%-95.0%), respectively. Clinical and biochemical responses were 74.9% (95% CI: 56.3%-90.2%) and 69.5% (95%CI: 40.2%-92.9%). Median PFS and median OS were 31.79 (95% CI:21.25-42.33) months and 74.30 (95% CI: 0.75-147.84) months, respectively. Any grade of hematotoxicity and nephrotoxicity occurred in 22.3% (95% CI:12.5%-33.5%) and 4.3% (95% CI:0.2%-11.4%) patients. Grade 3-4 hemotoxicity occurred in 4.3% (95% CI:0.2%-11.4%) and grade 3-4 nephrotoxicity in 4/212 patients. Additionally, Treatment was discontinued in 9.0% (95% CI: 0.5%-23.3%) patients and one patient died as a result of a toxicity. Conclusion Patients with metastatic or inoperable PPGLs can be effectively treated with PRRT, and it has a favorable safety profile. Systematic review registration https://www.crd.york.ac.uk/PROSPERO, identifier CRD42022359232.
Collapse
Affiliation(s)
- Dan Su
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan, China
| | - Hongyu Yang
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan, China
| | - Chen Qiu
- Department of Ophthalmology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Yue Chen
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan, China
- Academician (expert) Workstation of Sichuan Province, Luzhou, Sichuan, China
| |
Collapse
|
46
|
Zhu T, Hsu JC, Guo J, Chen W, Cai W, Wang K. Radionuclide-based theranostics - a promising strategy for lung cancer. Eur J Nucl Med Mol Imaging 2023; 50:2353-2374. [PMID: 36929181 PMCID: PMC10272099 DOI: 10.1007/s00259-023-06174-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 02/25/2023] [Indexed: 03/18/2023]
Abstract
PURPOSE This review aims to provide a comprehensive overview of the latest literature on personalized lung cancer management using different ligands and radionuclide-based tumor-targeting agents. BACKGROUND Lung cancer is the leading cause of cancer-related deaths worldwide. Due to the heterogeneity of lung cancer, advances in precision medicine may enhance the disease management landscape. More recently, theranostics using the same molecule labeled with two different radionuclides for imaging and treatment has emerged as a promising strategy for systemic cancer management. In radionuclide-based theranostics, the target, ligand, and radionuclide should all be carefully considered to achieve an accurate diagnosis and optimal therapeutic effects for lung cancer. METHODS We summarize the latest radiotracers and radioligand therapeutic agents used in diagnosing and treating lung cancer. In addition, we discuss the potential clinical applications and limitations associated with target-dependent radiotracers as well as therapeutic radionuclides. Finally, we provide our views on the perspectives for future development in this field. CONCLUSIONS Radionuclide-based theranostics show great potential in tailored medical care. We expect that this review can provide an understanding of the latest advances in radionuclide therapy for lung cancer and promote the application of radioligand theranostics in personalized medicine.
Collapse
Affiliation(s)
- Tianxing Zhu
- Department of Respiratory Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, Zhejiang, China
- Lingang Laboratory, Shanghai, 200031, China
| | - Jessica C Hsu
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Jingpei Guo
- Department of Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, 519000, Guangdong, China
| | - Weiyu Chen
- Department of Respiratory Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, Zhejiang, China.
- International Institutes of Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, Zhejiang, China.
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI, 53705, USA.
| | - Kai Wang
- Department of Respiratory Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, Zhejiang, China.
| |
Collapse
|
47
|
Zhang X, Wakabayashi H, Hiromasa T, Kayano D, Kinuya S. Recent Advances in Radiopharmaceutical Theranostics of Pheochromocytoma and Paraganglioma. Semin Nucl Med 2023; 53:503-516. [PMID: 36641337 DOI: 10.1053/j.semnuclmed.2022.12.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/23/2022] [Accepted: 12/29/2022] [Indexed: 01/15/2023]
Abstract
As a rare kind of non-epithelial neuroendocrine neoplasms, paragangliomas (PGLs) exhibit various clinical characteristics with excessive catecholamine secretion and have been a research focus in recent years. Although several modalities are available nowadays, radiopharmaceuticals play an integral role in the management of PGLs. Theranostics utilises radiopharmaceuticals for diagnostic and therapeutic intentions by aiming at a specific target in tumour and has been considered a possible means in diagnosis, staging, monitoring and treatment planning. Numerous radiopharmaceuticals have been developed over the past decades. 123/131-Metaiodobenzylguanidine (123/131I-MIBG), the theranostics pair target on norepinephrine transporter system, has remained a fantastic protocol for patients with PGLs because of disease control with limited toxicity. The high-specific-activity 131I-MIBG was authorised by the Food and Drug Administration as a systemic treatment method for metastatic PGLs in 2018. Afterward, peptide receptor radionuclide therapy, which uses radiolabelled somatostatin (SST) analogues, has been exploited as a superior substitute. 68Ga-somatostatin analogue (SSA) PET showed significant performance in diagnosing PGLs than MIBG scintigraphy, especially in patients with head and neck PGLs or SDHx mutation. 90Y/177Lu-DOTA-SSA is highly successful and has preserved favourable safety with mounting evidence regarding objective response, disease stabilisation, symptomatic and hormonal management and quality of life preservation. Besides the ordinary beta emitters, alpha-emitters such as 211At-MABG and 225Ac-DOTATATE have been investigated intensively in recent years. However, many studies are still in the pre-clinical stage, and more research is necessary. This review summarises the developments and recent advances in radiopharmaceutical theranostics of PGLs.
Collapse
Affiliation(s)
- Xue Zhang
- Department of Nuclear Medicine, Kanazawa University Hospital, Kanazawa, Ishikawa, Japan
| | - Hiroshi Wakabayashi
- Department of Nuclear Medicine, Kanazawa University Hospital, Kanazawa, Ishikawa, Japan.
| | - Tomo Hiromasa
- Department of Nuclear Medicine, Kanazawa University Hospital, Kanazawa, Ishikawa, Japan
| | - Daiki Kayano
- Department of Nuclear Medicine, Kanazawa University Hospital, Kanazawa, Ishikawa, Japan
| | - Seigo Kinuya
- Department of Nuclear Medicine, Kanazawa University Hospital, Kanazawa, Ishikawa, Japan
| |
Collapse
|
48
|
Lepareur N, Ramée B, Mougin-Degraef M, Bourgeois M. Clinical Advances and Perspectives in Targeted Radionuclide Therapy. Pharmaceutics 2023; 15:1733. [PMID: 37376181 DOI: 10.3390/pharmaceutics15061733] [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: 05/19/2023] [Revised: 06/09/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Targeted radionuclide therapy has become increasingly prominent as a nuclear medicine subspecialty. For many decades, treatment with radionuclides has been mainly restricted to the use of iodine-131 in thyroid disorders. Currently, radiopharmaceuticals, consisting of a radionuclide coupled to a vector that binds to a desired biological target with high specificity, are being developed. The objective is to be as selective as possible at the tumor level, while limiting the dose received at the healthy tissue level. In recent years, a better understanding of molecular mechanisms of cancer, as well as the appearance of innovative targeting agents (antibodies, peptides, and small molecules) and the availability of new radioisotopes, have enabled considerable advances in the field of vectorized internal radiotherapy with a better therapeutic efficacy, radiation safety and personalized treatments. For instance, targeting the tumor microenvironment, instead of the cancer cells, now appears particularly attractive. Several radiopharmaceuticals for therapeutic targeting have shown clinical value in several types of tumors and have been or will soon be approved and authorized for clinical use. Following their clinical and commercial success, research in that domain is particularly growing, with the clinical pipeline appearing as a promising target. This review aims to provide an overview of current research on targeting radionuclide therapy.
Collapse
Affiliation(s)
- Nicolas Lepareur
- Comprehensive Cancer Center Eugène Marquis, 35000 Rennes, France
- Inserm, INRAE, Institut NUMECAN (Nutrition, Métabolismes et Cancer)-UMR 1317, Univ Rennes, 35000 Rennes, France
| | - Barthélémy Ramée
- Nuclear Medicine Department, Nantes University Hospital, 44000 Nantes, France
| | - Marie Mougin-Degraef
- Nuclear Medicine Department, Nantes University Hospital, 44000 Nantes, France
- Inserm, CNRS, CRCI2NA (Centre de Recherche en Cancérologie et Immunologie Intégrée Nantes-Angers)-UMR 1307, Université de Nantes, ERL 6001, 44000 Nantes, France
| | - Mickaël Bourgeois
- Nuclear Medicine Department, Nantes University Hospital, 44000 Nantes, France
- Inserm, CNRS, CRCI2NA (Centre de Recherche en Cancérologie et Immunologie Intégrée Nantes-Angers)-UMR 1307, Université de Nantes, ERL 6001, 44000 Nantes, France
- Groupement d'Intérêt Public ARRONAX, 1 Rue Aronnax, 44817 Saint Herblain, France
| |
Collapse
|
49
|
Merola E, Grana CM. Peptide Receptor Radionuclide Therapy (PRRT): Innovations and Improvements. Cancers (Basel) 2023; 15:2975. [PMID: 37296936 PMCID: PMC10251822 DOI: 10.3390/cancers15112975] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 05/27/2023] [Accepted: 05/28/2023] [Indexed: 06/12/2023] Open
Abstract
Neuroendocrine neoplasms (NENs) are tumors originating from neuroendocrine cells distributed throughout the human body. With an increasing incidence over the past few decades, they represent a highly heterogeneous group of neoplasms, mostly expressing somatostatin receptors (SSTRs) on their cell surface. Peptide receptor radionuclide therapy (PRRT) has emerged as a crucial strategy for treating advanced, unresectable neuroendocrine tumors by administering radiolabeled somatostatin analogs intravenously to target SSTRs. This article will focus on the multidisciplinary theranostic approach, treatment effectiveness (such as response rates and symptom relief), patient outcomes, and toxicity profile of PRRT for NEN patients. We will review the most significant studies, such as the phase III NETTER-1 trial, and discuss promising new radiopharmaceuticals, including alpha-emitting radionuclide-labeled somatostatin analogs and SSTR antagonists.
Collapse
Affiliation(s)
- Elettra Merola
- Gastroenterology Unit, G.B. Grassi Hospital (ASL Roma 3), Lido di Ostia, 00122 Rome, Italy
| | - Chiara Maria Grana
- Radiometabolic Therapy Unit, Division of Nuclear Medicine, IRCCS European Institute of Oncology, 20141 Milan, Italy;
| |
Collapse
|
50
|
Abstract
Neuroendocrine neoplasms (NENs) are tumors originating from neuroendocrine cells distributed throughout the human body. With an increasing incidence over the past few decades, they represent a highly heterogeneous group of neoplasms, mostly expressing somatostatin receptors (SSTRs) on their cell surface. Peptide receptor radionuclide therapy (PRRT) has emerged as a crucial strategy for treating advanced, unresectable neuroendocrine tumors by administering radiolabeled somatostatin analogs intravenously to target SSTRs. This article will focus on the multidisciplinary theranostic approach, treatment effectiveness (such as response rates and symptom relief), patient outcomes, and toxicity profile of PRRT for NEN patients. We will review the most significant studies, such as the phase III NETTER-1 trial, and discuss promising new radiopharmaceuticals, including alpha-emitting radionuclide-labeled somatostatin analogs and SSTR antagonists.
Collapse
Affiliation(s)
- Elettra Merola
- Gastroenterology Unit, G.B. Grassi Hospital (ASL Roma 3), Lido di Ostia, 00122 Rome, Italy
| | - Chiara Maria Grana
- Radiometabolic Therapy Unit, Division of Nuclear Medicine, IRCCS European Institute of Oncology, 20141 Milan, Italy
| |
Collapse
|