1
|
Hou R, Liu N, Li F. Nanoradiopharmaceuticals: An Attractive Concept in Oncotherapy. ChemMedChem 2024; 19:e202400423. [PMID: 39140435 DOI: 10.1002/cmdc.202400423] [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: 06/03/2024] [Revised: 08/10/2024] [Accepted: 08/12/2024] [Indexed: 08/15/2024]
Abstract
Radiopharmaceuticals are of significant importance in the fields of tumor imaging and therapy. In recent decades, the increasing role of nanotechnology has led to the attractive concept of nanoradiopharmaceuticals. Consequently, it is imperative to provide a concise summary of the necessary guidelines to facilitate the translation of nanoradiopharmaceuticals. In this work, we have presented the contents of radiolabeling strategies and some applications of nanoradiopharmaceuticals. Such a framework can assist researchers in identifying more pertinent insights or making more informed decisions in the study of nanoradiopharmaceuticals.
Collapse
Affiliation(s)
- Ruitong Hou
- Key Laboratory of Radiation Physics and Technology of the Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University, Chengdu, 610064, PR China
| | - Ning Liu
- Key Laboratory of Radiation Physics and Technology of the Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University, Chengdu, 610064, PR China
| | - Feize Li
- Key Laboratory of Radiation Physics and Technology of the Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University, Chengdu, 610064, PR China
| |
Collapse
|
2
|
Ode Y, Pradipta AR, Ishiwata A, Nambu A, Ohnuki K, Mizuma H, Haba H, Tanaka K. Metallic radionuclide-labeled tetrameric 2,6-diisopropylphenyl azides for cancer treatment. Chem Commun (Camb) 2024; 60:3291-3294. [PMID: 38421438 DOI: 10.1039/d4cc00048j] [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: 03/02/2024]
Abstract
This study proposes a new method for radionuclide therapy that involves the use of oligomeric 2,6-diisopropylphenyl azides and a chelator to form stable complexes with metallic radionuclides. The technique works by taking advantage of the endogenous acrolein produced by cancer cells. The azides react with the acrolein to give a diazo derivative that immediately attaches to the nearest organelle, effectively anchoring the radionuclide within the tumor. Preliminary in vivo experiments were conducted on a human lung carcinoma xenograft model, demonstrating the feasibility of this approach for cancer treatment.
Collapse
Affiliation(s)
- Yudai Ode
- Department of Chemical Science and Engineering, School of Materials and Chemical Technology, Tokyo Institute of Technology, 2-12-1 Ookayama, Meguro, Tokyo 152-8552, Japan.
| | - Ambara R Pradipta
- Department of Chemical Science and Engineering, School of Materials and Chemical Technology, Tokyo Institute of Technology, 2-12-1 Ookayama, Meguro, Tokyo 152-8552, Japan.
| | - Akihiro Ishiwata
- Biofunctional Synthetic Chemistry Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Akihiro Nambu
- Nuclear Chemistry Group, RIKEN Nishina Center for Accelerator-Based Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Kazunobu Ohnuki
- Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, 6-5-1 Kashiwanoha, Kashiwa, Chiba 277-8577, Japan
| | - Hiroshi Mizuma
- Department of Functional Brain Imaging, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba-shi, Chiba 263-8555, Japan
- Laboratory for Pathophysiological and Health Science, RIKEN Center for Biosystems Dynamics Research, Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Hiromitsu Haba
- Nuclear Chemistry Group, RIKEN Nishina Center for Accelerator-Based Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Katsunori Tanaka
- Department of Chemical Science and Engineering, School of Materials and Chemical Technology, Tokyo Institute of Technology, 2-12-1 Ookayama, Meguro, Tokyo 152-8552, Japan.
- Biofunctional Synthetic Chemistry Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| |
Collapse
|
3
|
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
|
4
|
Tang Y, Liao Z, Li F, Liu W, Gao J, Li Y, Hu Y, Cai H, Ma H, Yang Y, Yang J, Liao J, Liu N. A novel theranostic probe [ 111In]In-DO3A-NHS-nimotuzumab in glioma xenograft. RADIOCHIM ACTA 2022. [DOI: 10.1515/ract-2021-1064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Abstract
Indium-111 (111In) has an appropriate half-life (T
1/2 = 67 h) and energy characteristics for cancer diagnosis via γ-ray imaging and cancer therapy with Auger electrons. The aim of our study is to evaluate the potential of [111In]In-DO3A-NHS-nimotuzumab as a theranostic agent for radioimmunoimaging (RII) and radioimmunotherapy (RIT) against human glioma xenografts in mice. We explored the chelators DO3A-NHS and DOTA-p-SCN-Bz to optimize 111In radiolabeling efficiency of nimotuzumab. The radiopharmaceuticals were purified by PD-10 mini-column and their in vitro stabilities were assessed. We investigated the biodistribution of [111In]In-DO3A-NHS-nimotuzumab as it had relatively superior labeling efficiency and stability in vitro. We conducted SPECT imaging on mice bearing glioma (U87MG) xenografts, which were injected with ∼3.7 MBq of [111In]In-DO3A-NHS-nimotuzumab. The in vivo radiotherapeutic effects of [111In]In-DO3A-NHS-nimotuzumab was analyzed via injecting a single 37 MBq dose, 2 × 18 MBq doses, or 2 × 37 MBq doses into mice bearing U87MG xenografts. The control groups were administered either 30 μg nimotuzumab or saline. The radiochemical yields of [111In]In-DO3A-NHS-nimotuzumab and [111In]In-DOTA-p-SCN-Bz-nimotuzumab were > 85% and > 75%, respectively. [111In]In-DO3A-NHS-nimotuzumab had > 95% radiochemical purity and was more stable in vitro than [111In]In-DOTA-p-SCN-Bz-nimotuzumab. Biodistribution study demonstrated that [111In]In-DO3A-NHS-nimotuzumab was highly stable in vivo. SPECT imaging disclosed that [111In]In-DO3A-NHS-nimotuzumab had excellent targeted tumor uptake and retained in tumors for 24 and 72 h. All [111In]In-DO3A-NHS-nimotuzumab treatments substantially inhibited tumor growth over the controls. The 2 × 37 MBq treatment was particularly efficacious, and presented with survival time prolonged by ≤66 days. In contrast, the survival time of the control group was only 30 days. In our study, we developed an optimized synthesis protocol for radiopharmaceutical 111In-DO3A-NHS-nimotuzumab and demonstrated that it is a promising theranostic agent. It could be highly efficacious in RII and RIT against EGFR-expressing glioma.
Collapse
Affiliation(s)
- Yu Tang
- Key Laboratory of Radiation Physics and Technology of Ministry of Education , Institute of Nuclear Science and Technology, Sichuan University , Chengdu 610064 , P. R. China
| | - Zhonghui Liao
- Key Laboratory of Radiation Physics and Technology of Ministry of Education , Institute of Nuclear Science and Technology, Sichuan University , Chengdu 610064 , P. R. China
| | - Feize Li
- Key Laboratory of Radiation Physics and Technology of Ministry of Education , Institute of Nuclear Science and Technology, Sichuan University , Chengdu 610064 , P. R. China
| | - Weihao Liu
- Key Laboratory of Radiation Physics and Technology of Ministry of Education , Institute of Nuclear Science and Technology, Sichuan University , Chengdu 610064 , P. R. China
| | - Jing Gao
- Key Laboratory of Radiation Physics and Technology of Ministry of Education , Institute of Nuclear Science and Technology, Sichuan University , Chengdu 610064 , P. R. China
| | - Yuhao Li
- Department of Nuclear Medicine, Laboratory of Clinical Nuclear Medicine , West China Hospital, Sichuan University , Chengdu 610041 , P. R. China
| | - Yingjiang Hu
- Key Laboratory of Radiation Physics and Technology of Ministry of Education , Institute of Nuclear Science and Technology, Sichuan University , Chengdu 610064 , P. R. China
| | - Huawei Cai
- Department of Nuclear Medicine, Laboratory of Clinical Nuclear Medicine , West China Hospital, Sichuan University , Chengdu 610041 , P. R. China
| | - Huan Ma
- Key Laboratory of Radiation Physics and Technology of Ministry of Education , Institute of Nuclear Science and Technology, Sichuan University , Chengdu 610064 , P. R. China
| | - Yuanyou Yang
- Key Laboratory of Radiation Physics and Technology of Ministry of Education , Institute of Nuclear Science and Technology, Sichuan University , Chengdu 610064 , P. R. China
| | - Jijun Yang
- Key Laboratory of Radiation Physics and Technology of Ministry of Education , Institute of Nuclear Science and Technology, Sichuan University , Chengdu 610064 , P. R. China
| | - Jiali Liao
- Key Laboratory of Radiation Physics and Technology of Ministry of Education , Institute of Nuclear Science and Technology, Sichuan University , Chengdu 610064 , P. R. China
| | - Ning Liu
- Key Laboratory of Radiation Physics and Technology of Ministry of Education , Institute of Nuclear Science and Technology, Sichuan University , Chengdu 610064 , P. R. China
| |
Collapse
|
5
|
Bartoli F, Eckelman WC, Boyd M, Mairs RJ, Erba PA. Principles of Molecular Targeting for Radionuclide Therapy. NUCLEAR ONCOLOGY 2022:41-93. [DOI: 10.1007/978-3-031-05494-5_32] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
6
|
Abstract
Abstract
Theragnostics in nuclear medicine constitute an essential element of precision medicine. This notion integrates radionuclide diagnostics procedures and radionuclide therapies using appropriate radiopharmaceutics and treatment targeting specific biological pathways or receptors. The term theragnostics should also include another aspect of treatment: not only whether a given radioisotopic drug can be used, but also in what dose it ought to be used. Theragnostic procedures also allow predicting the effects of treatment based on the assessment of specific receptor density or the metabolic profile of neoplastic cells. The future of theragnostics depends not only on the use of new radiopharmaceuticals, but also on new gamma cameras. Modern theragnostics already require unambiguous pharmacokinetic and pharmacodynamic measurements based on absolute values. Only dynamic studies provide such a possibility. The introduction of the dynamic total-body PET-CT will enable this type of measurements characterizing metabolic processes and receptor expression on the basis of Patlak plot.
Collapse
Affiliation(s)
- Leszek Królicki
- Department of Nuclear Medicine , Medical University of Warsaw , Warszawa , Poland
| | - Jolanta Kunikowska
- Department of Nuclear Medicine , Medical University of Warsaw , Warszawa , Poland
| |
Collapse
|
7
|
Bavelaar BM, Song L, Jackson MR, Able S, Tietz O, Skaripa-Koukelli I, Waghorn PA, Gill MR, Carlisle RC, Tarsounas M, Vallis KA. Oligonucleotide-Functionalized Gold Nanoparticles for Synchronous Telomerase Inhibition, Radiosensitization, and Delivery of Theranostic Radionuclides. Mol Pharm 2021; 18:3820-3831. [PMID: 34449222 PMCID: PMC8493550 DOI: 10.1021/acs.molpharmaceut.1c00442] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 08/07/2021] [Accepted: 08/11/2021] [Indexed: 12/13/2022]
Abstract
Telomerase represents an attractive target in oncology as it is expressed in cancer but not in normal tissues. The oligonucleotide inhibitors of telomerase represent a promising anticancer strategy, although poor cellular uptake can restrict their efficacy. In this study, gold nanoparticles (AuNPs) were used to enhance oligonucleotide uptake. "match" oligonucleotides complementary to the telomerase RNA template subunit (hTR) and "scramble" (control) oligonucleotides were conjugated to diethylenetriamine pentaacetate (DTPA) for 111In-labeling. AuNPs (15.5 nm) were decorated with a monofunctional layer of oligonucleotides (ON-AuNP) or a multifunctional layer of oligonucleotides, PEG(polethylene glycol)800-SH (to reduce AuNP aggregation) and the cell-penetrating peptide Tat (ON-AuNP-Tat). Match-AuNP enhanced the cellular uptake of radiolabeled oligonucleotides while retaining the ability to inhibit telomerase activity. The addition of Tat to AuNPs increased nuclear localization. 111In-Match-AuNP-Tat induced DNA double-strand breaks and caused a dose-dependent reduction in clonogenic survival of telomerase-positive cells but not telomerase-negative cells. hTR inhibition has been reported to sensitize cancer cells to ionizing radiation, and 111In-Match-AuNP-Tat therefore holds promise as a vector for delivery of radionuclides into cancer cells while simultaneously sensitizing them to the effects of the emitted radiation.
Collapse
Affiliation(s)
- Bas M. Bavelaar
- Oxford
Institute for Radiation Oncology, University
of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, U.K.
| | - Lei Song
- Oxford
Institute for Radiation Oncology, University
of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, U.K.
| | - Mark R. Jackson
- Institute
of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, University Avenue, Glasgow G12 8QQ, U.K.
| | - Sarah Able
- Oxford
Institute for Radiation Oncology, University
of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, U.K.
| | - Ole Tietz
- Oxford
Institute for Radiation Oncology, University
of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, U.K.
| | - Irini Skaripa-Koukelli
- Oxford
Institute for Radiation Oncology, University
of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, U.K.
| | - Philip A. Waghorn
- Charles
River Laboratories, Elphinstone Research Centre, Elphinstone, Tranent EH33 2NE, U.K.
| | - Martin R. Gill
- Oxford
Institute for Radiation Oncology, University
of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, U.K.
| | - Robert C. Carlisle
- Institute
of Biomedical Engineering, Department of Engineering Science, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, U.K.
| | - Madalena Tarsounas
- Oxford
Institute for Radiation Oncology, University
of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, U.K.
| | - Katherine A. Vallis
- Oxford
Institute for Radiation Oncology, University
of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, U.K.
| |
Collapse
|
8
|
Jaymand M, Davatgaran Taghipour Y, Rezaei A, Derakhshankhah H, Foad Abazari M, Samadian H, Hamblin MR. Radiolabeled carbon-based nanostructures: New radiopharmaceuticals for cancer therapy? Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2021.213974] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
9
|
Askari Rizvi SF, Zhang H. Emerging trends of receptor-mediated tumor targeting peptides: A review with perspective from molecular imaging modalities. Eur J Med Chem 2021; 221:113538. [PMID: 34022717 DOI: 10.1016/j.ejmech.2021.113538] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 05/06/2021] [Accepted: 05/07/2021] [Indexed: 01/10/2023]
Abstract
Natural peptides extracted from natural components such are known to have a relatively short in-vivo half-life and can readily metabolize by endo- and exo-peptidases. Fortunately, synthetic peptides can be easily manipulated to increase in-vivo stability, membrane permeability and target specificity with some well-known natural families. Many natural as well as synthetic peptides target to their endogenous receptors for diagnosis and therapeutic applications. In order to detect these peptides externally, they must be modified with radionuclides compatible with single photon emission computed tomography (SPECT) or positron emission tomography (PET). Although, these techniques mainly rely on physiological changes and have profound diagnostic strength over anatomical modalities such as MRI and CT. However, both SPECT and PET observed to possess lack of anatomical reference frame which is a key weakness of these techniques, and unfortunately, cannot be available freely in most clinical centres especially in under-developing countries. Hence, it is need of the time to design and develop economic, patient friendly and versatile strategies to grapple with existing problems without any hazardous side effects. Optical molecular imaging (OMI) has emerged as a novel technique in field of medical science using fluorescent probes as imaging modality and has ability to couple with organic drugs, small molecules, chemotherapeutics, DNA, RNA, anticancer peptide and protein without adding chelators as necessary for radionuclides. Furthermore, this review focuses on difference in imaging modalities and provides ample knowledge about reliable, economic and patient friendly optical imaging technique rather radionuclide-based imaging techniques.
Collapse
Affiliation(s)
- Syed Faheem Askari Rizvi
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, 730000, Gansu, PR China
| | - Haixia Zhang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, 730000, Gansu, PR China.
| |
Collapse
|
10
|
Clinical Perspectives of Theranostics. Molecules 2021; 26:molecules26082232. [PMID: 33924345 PMCID: PMC8070270 DOI: 10.3390/molecules26082232] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/29/2021] [Accepted: 04/06/2021] [Indexed: 12/24/2022] Open
Abstract
Theranostics is a precision medicine which integrates diagnostic nuclear medicine and radionuclide therapy for various cancers throughout body using suitable tracers and treatment that target specific biological pathways or receptors. This review covers traditional theranostics for thyroid cancer and pheochromocytoma with radioiodine compounds. In addition, recent theranostics of radioimmunotherapy for non-Hodgkin lymphoma, and treatment of bone metastasis using bone seeking radiopharmaceuticals are described. Furthermore, new radiopharmaceuticals for prostatic cancer and pancreatic cancer have been added. Of particular, F-18 Fluoro-2-Deoxyglucose (FDG) Positron Emission Tomography (PET) is often used for treatment monitoring and estimating patient outcome. A recent clinical study highlighted the ability of alpha-radiotherapy with high linear energy transfer (LET) to overcome treatment resistance to beta--particle therapy. Theranostics will become an ever-increasing part of clinical nuclear medicine.
Collapse
|
11
|
Yoshinaga K, Abe T, Okamoto S, Uchiyama Y, Manabe O, Ito YM, Tamura N, Ito N, Yoshioka N, Washino K, Shinohara N, Tamaki N, Shiga T. Effects of Repeated 131I- Meta-Iodobenzylguanidine Radiotherapy on Tumor Size and Tumor Metabolic Activity in Patients with Metastatic Neuroendocrine Tumors. J Nucl Med 2020; 62:685-694. [PMID: 33067337 DOI: 10.2967/jnumed.120.250803] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 09/15/2020] [Indexed: 12/30/2022] Open
Abstract
131I-meta-iodobenzylguanidine (131I-MIBG) radiotherapy has shown some survival benefits in metastatic neuroendocrine tumors (NETs). European Association of Nuclear Medicine clinical guidelines for 131I-MIBG radiotherapy suggest a repeated treatment protocol, although none currently exists. The existing single-high-dose 131I-MIBG radiotherapy (444 MBq/kg) has been shown to have some benefits for patients with metastatic NETs. However, this protocol increases adverse effects and requires alternative therapeutic approaches. Therefore, the aim of this study was to evaluate the effects of repeated 131I-MIBG therapy on tumor size and tumor metabolic response in patients with metastatic NETs. Methods: Eleven patients with metastatic NETs (aged 49.2 ± 16.3 y) prospectively received repeated 5,550-MBq doses of 131I-MIBG therapy at 6-mo intervals. In total, 31 treatments were performed. The mean number of treatments was 2.8 ± 0.4, and the cumulative 131I-MIBG dose was 15,640.9 ± 2,245.1 MBq (286.01 MBq/kg). Tumor response was observed by CT and 18F-FDG PET or by 18F-FDG PET/CT before and 3-6 mo after the final 131I-MIBG treatment. Results: On the basis of the CT findings with RECIST, 3 patients showed a partial response and 6 patients showed stable disease. The remaining 2 patients showed progressive disease. Although there were 2 progressive-disease patients, analysis of all patients showed no increase in summed length diameter (median, 228.7 mm [interquartile range (IQR), 37.0-336.0 mm] to 171.0 mm [IQR, 38.0-270.0 mm]; P = 0.563). In tumor region-based analysis with partial-response and stable-disease patients (n = 9), 131I-MIBG therapy significantly reduced tumor diameter (79 lesions; median, 16 mm [IQR, 12-22 mm] to 11 mm [IQR, 6-16 mm]; P < 0.001). Among 5 patients with hypertension, there was a strong trend toward systolic blood pressure reduction (P = 0.058), and diastolic blood pressure was significantly reduced (P = 0.006). Conclusion: Eighty-two percent of metastatic NET patients effectively achieved inhibition of disease progression, with reduced tumor size and reduced metabolic activity, through repeated 131I-MIBG therapy. Therefore, this relatively short-term repeated 131I-MIBG treatment may have potential as one option in the therapeutic protocol for metastatic NETs. Larger prospective studies with control groups are warranted.
Collapse
Affiliation(s)
- Keiichiro Yoshinaga
- Diagnostic and Therapeutic Nuclear Medicine, Quantum and Radiological Science and Technology, National Institute of Radiological Sciences, Chiba, Japan
| | - Takashige Abe
- Department of Urological Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Shozo Okamoto
- Department of Diagnostic Radiology, Hokkaido University Graduate School of Medicine, Sapporo, Japan .,Department of Radiology, Obihiro Kosei Hospital, Obihiro, Japan
| | - Yuko Uchiyama
- Department of Diagnostic Radiology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Osamu Manabe
- Department of Diagnostic Radiology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Yoichi M Ito
- Research Center for Medical and Health Data Science, Institute of Statistical Mathematics, Tokyo, Japan; and
| | - Naomi Tamura
- Research Center for Medical and Health Data Science, Institute of Statistical Mathematics, Tokyo, Japan; and
| | - Natsue Ito
- Diagnostic and Therapeutic Nuclear Medicine, Quantum and Radiological Science and Technology, National Institute of Radiological Sciences, Chiba, Japan
| | - Naho Yoshioka
- Diagnostic and Therapeutic Nuclear Medicine, Quantum and Radiological Science and Technology, National Institute of Radiological Sciences, Chiba, Japan
| | - Komei Washino
- Diagnostic and Therapeutic Nuclear Medicine, Quantum and Radiological Science and Technology, National Institute of Radiological Sciences, Chiba, Japan
| | - Nobuo Shinohara
- Department of Urological Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Nagara Tamaki
- Department of Radiology, Kyoto Prefectural Medical University, Kyoto, Japan
| | - Tohru Shiga
- Department of Diagnostic Radiology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| |
Collapse
|
12
|
Xie L, Hanyu M, Fujinaga M, Zhang Y, Hu K, Minegishi K, Jiang C, Kurosawa F, Morokoshi Y, Li HK, Hasegawa S, Nagatsu K, Zhang MR. 131I-IITM and 211At-AITM: Two Novel Small-Molecule Radiopharmaceuticals Targeting Oncoprotein Metabotropic Glutamate Receptor 1. J Nucl Med 2019; 61:242-248. [PMID: 31451486 DOI: 10.2967/jnumed.119.230946] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 07/31/2019] [Indexed: 11/16/2022] Open
Abstract
Targeted radionuclide therapy (TRT) targeting oncoproteins facilitates the delivery of therapeutic radionuclides to tumor tissues with high precision. Herein, we developed 2 new radiopharmaceuticals, 4-131I-iodo- and 4-211At-astato-N-[4-(6-(isopropylamino)pyridine-4-yl)-1,3-thiazol-2-yl]-N-methylbenzamide (131I-IITM and 211At-AITM), targeting the ectopic metabotropic glutamate receptor 1 (mGluR1) in melanomas for TRT studies. Methods: 131I-IITM and 211At-AITM were synthesized by reacting a stannyl precursor with 131I-NaI and 211At in the presence of an oxidizing agent. The therapeutic efficacy and safety of the 2 radiopharmaceuticals were investigated using mGluR1-expressing B16F10 melanoma cells and melanoma-bearing mice. Results: 131I-IITM and 211At-AITM were obtained with a radiochemical purity of greater than 99% and radiochemical yields of 42.7% ± 10.4% and 45.7% ± 6.5%, respectively, based on the total radioactivity of used radionuclides. 131I-IITM and 211At-AITM exhibited a maximum uptake of 4.66% ± 0.70 and 7.68% ± 0.71 percentage injected dose per gram (%ID/g) in the targeted melanomas, respectively, and were rapidly cleared from nontarget organs after intravenous injection. Both agents markedly inhibited melanoma growth compared with the controls (61.00% and 95.68%, respectively). In the melanoma model, considerably greater therapeutic efficacy with negligible toxicity was observed using 211At-AITM. Conclusion: The nontoxic radiopharmaceuticals 131I-IITM and 211At-AITM are useful high-precision TRT agents that can be used to target the oncoprotein mGluR1 for melanoma therapy.
Collapse
Affiliation(s)
- Lin Xie
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Masayuki Hanyu
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Masayuki Fujinaga
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Yiding Zhang
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Kuan Hu
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Katsuyuki Minegishi
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Cuiping Jiang
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Fuki Kurosawa
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan.,Department of Charged Particle Therapy Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan; and
| | - Yukie Morokoshi
- Department of Charged Particle Therapy Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan; and
| | - Huizi Keiko Li
- Department of Charged Particle Therapy Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan; and.,The Japan Society for the Promotion of Science, Tokyo, Japan
| | - Sumitaka Hasegawa
- Department of Charged Particle Therapy Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan; and
| | - Kotaro Nagatsu
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Ming-Rong Zhang
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| |
Collapse
|
13
|
Comparison between Targeted Radionuclide Therapy of Bone Metastases Based on β-Emitting and α-Emitting Radionuclides. J Med Imaging Radiat Sci 2019; 50:272-279. [DOI: 10.1016/j.jmir.2018.12.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 12/02/2018] [Accepted: 12/20/2018] [Indexed: 12/30/2022]
|
14
|
Sadremomtaz A, Masoumi M. An assessment of bone-seeking radionuclides for palliation of metastatic bone pain in a vertebral model. Ann Nucl Med 2019; 33:252-264. [PMID: 30659480 DOI: 10.1007/s12149-019-01329-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 12/28/2018] [Indexed: 11/28/2022]
Abstract
OBJECTIVE Bone-seeking radiopharmaceuticals have the main role in the treatment of painful bone metastases. The aim of this study was to dosimetrically compare radiopharmaceuticals in use for bone pain palliation therapy and bone scan. METHODS The MCNPX code was used to simulate the radiation transport in a vertebral phantom. Absorbed fractions were calculated for monoenergetic electrons, photons and alpha particles. S values were obtained for radionuclides 32P, 33P, 89Sr, 90Y, 99mTc, 117mSn, 153Sm, 166Ho, 169Er, 177Lu, 186Re, 188Re, 223Ra, 224Ra and their progenies for target regions including the active marrow and the bone endosteum. RESULTS The results demonstrated the dependence of dosimetric parameters on the source or target size, particle energy and location of the source. The electron emitters including 33P, 117mSn, 169Er and 177Lu and 223Ra as an α-emitter gave the lower absorbed dose to the active marrow. These radionuclides gave the highest values of the Relative Advantage Factor (RAF). CONCLUSIONS According to the results, 33P, 117mSn, 169Er, 177Lu and 223Ra have fewer side effects on the active marrow than other investigated radionuclides. Therefore, these radionuclides may be a better choice for use in palliative radiotherapy.
Collapse
Affiliation(s)
| | - Mahboubeh Masoumi
- Physics Department, Faculty of Science, University of Guilan, Rasht, Iran
| |
Collapse
|
15
|
Al-Salahi R, Moustapha ME, Abuelizz HA, Alharthi AI, Alburikan KA, Ibrahim IT, Marzouk M, Motaleb MA. Radioiodination and biodistribution of newly synthesized 3-benzyl-2-([3-methoxybenzyl]thio)benzo[ g]quinazolin-4-(3 H)-one in tumor bearing mice. Saudi Pharm J 2018; 26:1120-1126. [PMID: 30532632 PMCID: PMC6260473 DOI: 10.1016/j.jsps.2018.06.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 06/05/2018] [Indexed: 12/22/2022] Open
Abstract
3-Benzyl-2-((3-methoxybenzyl)thio)benzo[g]quinazolin-4(3H)-one was previously synthesized and proved by physicochemical analyses (HRMS, 1H and 13C NMR). The target compound was examined for its radioactivity and the results showed that benzo[g]quinazoline was successfully labeled with radioactive iodine using NBS via an electrophilic substitution reaction. The reaction parameters that affected the labeling yield such as concentration, pH and time were studied to optimize the labeling conditions. The radiochemical yield was 91.2 ± 1.22% and the in vitro studies showed that the target compound was stable for up to 24 h. The thyroid was among the other organs in which the uptake of 125I-benzoquinazoline has increased significantly over the time up to 4.1%. The tumor uptake was 6.95%. Radiochemical and metabolic stability of the benzoquinazoline in vivo/in vitro and biodistribution studies provide some insights about the requirements for developing more potent radiopharmaceutical for targeting the tumor cells.
Collapse
Affiliation(s)
- Rashad Al-Salahi
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Moustapha E. Moustapha
- Department of Chemistry, College of Science and Humanities, Prince Sattam bin Abdulaziz University, P.O. Box 83, Al Kharj 11942, Saudi Arabia
| | - Hatem A. Abuelizz
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Abdulrahman I. Alharthi
- Department of Chemistry, College of Science and Humanities, Prince Sattam bin Abdulaziz University, P.O. Box 83, Al Kharj 11942, Saudi Arabia
| | - Khalid A. Alburikan
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Ismail T. Ibrahim
- Labeled Compound Department, Hot Laboratories Centre, Atomic Energy Authority, P.O. Box 13759, Cairo, Egypt
| | - Mohamed Marzouk
- Department of Chemistry, College of Science and Humanities, Prince Sattam bin Abdulaziz University, P.O. Box 83, Al Kharj 11942, Saudi Arabia
- Chemistry of Natural Products Group, Center of Excellence for Advanced Sciences, National Research Centre, Dokki, Cairo 12622, Egypt
| | - Mohamed A. Motaleb
- Labeled Compound Department, Hot Laboratories Centre, Atomic Energy Authority, P.O. Box 13759, Cairo, Egypt
| |
Collapse
|
16
|
Bavelaar BM, Lee BQ, Gill MR, Falzone N, Vallis KA. Subcellular Targeting of Theranostic Radionuclides. Front Pharmacol 2018; 9:996. [PMID: 30233374 PMCID: PMC6131480 DOI: 10.3389/fphar.2018.00996] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 08/13/2018] [Indexed: 12/16/2022] Open
Abstract
The last decade has seen rapid growth in the use of theranostic radionuclides for the treatment and imaging of a wide range of cancers. Radionuclide therapy and imaging rely on a radiolabeled vector to specifically target cancer cells. Radionuclides that emit β particles have thus far dominated the field of targeted radionuclide therapy (TRT), mainly because the longer range (μm-mm track length) of these particles offsets the heterogeneous expression of the molecular target. Shorter range (nm-μm track length) α- and Auger electron (AE)-emitting radionuclides on the other hand provide high ionization densities at the site of decay which could overcome much of the toxicity associated with β-emitters. Given that there is a growing body of evidence that other sensitive sites besides the DNA, such as the cell membrane and mitochondria, could be critical targets in TRT, improved techniques in detecting the subcellular distribution of these radionuclides are necessary, especially since many β-emitting radionuclides also emit AE. The successful development of TRT agents capable of homing to targets with subcellular precision demands the parallel development of quantitative assays for evaluation of spatial distribution of radionuclides in the nm-μm range. In this review, the status of research directed at subcellular targeting of radionuclide theranostics and the methods for imaging and quantification of radionuclide localization at the nanoscale are described.
Collapse
Affiliation(s)
| | | | | | | | - Katherine A. Vallis
- CR-UK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
17
|
Martins CD, Kramer-Marek G, Oyen WJG. Radioimmunotherapy for delivery of cytotoxic radioisotopes: current status and challenges. Expert Opin Drug Deliv 2017; 15:185-196. [PMID: 28893110 DOI: 10.1080/17425247.2018.1378180] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Radioimmunotherapy (RIT) with monoclonal antibodies and their fragments labelled with radionuclides emitting α -particles, β-particles or Auger electrons have been used for many years in the development of anticancer strategies. While RIT has resulted in approved radiopharmaceuticals for the treatment of hematological malignancies, its use in solid tumors still remains challenging. AREAS COVERED In this review, we discuss the exciting progress towards elucidating the potential of current and novel radioimmunoconjugates and address the challenges for translation into clinical practice. EXPERT OPINION There are still technical and logistical challenges associated with the use of RIT in routine clinical practice, including development of novel and more specific targeting moieties, broader access α to α-emitters and better tailoring of pre-targeting approaches. Moreover, improved understanding of the heterogeneous nature of solid tumors and the critical role of tumor microenvironments will help to optimize clinical response to RIT by delivering sufficient radiation doses to even more radioresistant tumor cells.
Collapse
Affiliation(s)
- Carlos Daniel Martins
- a Division of Radiotherapy and Imaging , The Institute of Cancer Research , London , UK
| | - Gabriela Kramer-Marek
- a Division of Radiotherapy and Imaging , The Institute of Cancer Research , London , UK
| | - Wim J G Oyen
- a Division of Radiotherapy and Imaging , The Institute of Cancer Research , London , UK.,b The Royal Marsden NHS Foundation Trust , Department of Nuclear Medicine , London , UK
| |
Collapse
|
18
|
Ge H, Riss PJ, Mirabello V, Calatayud DG, Flower SE, Arrowsmith RL, Fryer TD, Hong Y, Sawiak S, Jacobs RM, Botchway SW, Tyrrell RM, James TD, Fossey JS, Dilworth JR, Aigbirhio FI, Pascu SI. Behavior of Supramolecular Assemblies of Radiometal-Filled and Fluorescent Carbon Nanocapsules In Vitro and In Vivo. Chem 2017. [DOI: 10.1016/j.chempr.2017.06.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
19
|
Gill MR, Falzone N, Du Y, Vallis KA. Targeted radionuclide therapy in combined-modality regimens. Lancet Oncol 2017; 18:e414-e423. [PMID: 28677577 DOI: 10.1016/s1470-2045(17)30379-0] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 03/27/2017] [Accepted: 04/10/2017] [Indexed: 12/12/2022]
Abstract
Targeted radionuclide therapy (TRT) is a branch of cancer medicine concerned with the use of radioisotopes, radiolabelled molecules, nanoparticles, or microparticles that either naturally accumulate in or are designed to target tumours. TRT combines the specificity of molecular and sometimes physical targeting with the potent cytotoxicity of ionising radiation. Targeting vectors for TRT include antibodies, antibody fragments, proteins, peptides, and small molecules. The diversity of available carrier molecules, together with the large panel of suitable radioisotopes with unique physicochemical properties, allows vector-radionuclide pairings to be matched to the molecular, pathological, and physical characteristics of a tumour. Some pairings are designed for dual therapeutic and diagnostic applications. Use of TRT is increasing with the adoption into practice of radium-223 dichloride for the treatment of bone metastases and with the ongoing clinical development of, among others, 177Lu-dodecanetetraacetic acid tyrosine-3-octreotate (DOTATATE) for the treatment of neuroendocrine tumours and 90Y-microspheres for the treatment of hepatic tumours. The increasing use of TRT raises the question of how best to integrate TRT into multimodality protocols. Achievements in this area and the future prospects of TRT are evaluated in this Review.
Collapse
Affiliation(s)
- Martin R Gill
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Nadia Falzone
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Yong Du
- The Royal Marsden Hospital NHS Foundation Trust, Sutton, Surrey, UK
| | - Katherine A Vallis
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK.
| |
Collapse
|
20
|
Liberal FDCG, Tavares AAS, Tavares JMRS. Computational modeling of radiobiological effects in bone metastases for different radionuclides. Int J Radiat Biol 2017; 93:627-636. [PMID: 28276897 DOI: 10.1080/09553002.2017.1294274] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
PURPOSE Computational simulation is a simple and practical way to study and to compare a variety of radioisotopes for different medical applications, including the palliative treatment of bone metastases. This study aimed to evaluate and compare cellular effects modelled for different radioisotopes currently in use or under research for treatment of bone metastases using computational methods. METHODS Computational models were used to estimate the radiation-induced cellular effects (Virtual Cell Radiobiology algorithm) post-irradiation with selected particles emitted by Strontium-89 (89Sr), Samarium-153 (153Sm), Lutetium-177 (177Lu), and Radium-223 (223Ra). RESULTS Cellular kinetics post-irradiation using 89Sr β- particles, 153Sm β- particles, 177Lu β- particles and 223Ra α particles showed that the cell response was dose- and radionuclide-dependent. 177Lu beta minus particles and, in particular, 223Ra alpha particles, yielded the lowest survival fraction of all investigated particles. CONCLUSIONS 223Ra alpha particles induced the highest cell death of all investigated particles on metastatic prostate cells in comparison to irradiation with β- radionuclides, two of the most frequently used radionuclides in the palliative treatment of bone metastases in clinical routine practice. Moreover, the data obtained suggest that the used computational methods might provide some perception about cellular effects following irradiation with different radionuclides.
Collapse
Affiliation(s)
- Francisco D C Guerra Liberal
- a Faculdade de Engenharia , Instituto de Ciência e Inovação em Engenharia Mecânica e Engenharia Industrial, Universidade do Porto , Porto , Portugal
| | - Adriana Alexandre S Tavares
- a Faculdade de Engenharia , Instituto de Ciência e Inovação em Engenharia Mecânica e Engenharia Industrial, Universidade do Porto , Porto , Portugal
| | - João Manuel R S Tavares
- a Faculdade de Engenharia , Instituto de Ciência e Inovação em Engenharia Mecânica e Engenharia Industrial, Universidade do Porto , Porto , Portugal
| |
Collapse
|
21
|
Royle G, Falzone N, Chakalova R, Vallis K, Myhra S. Internalization of Auger electron-emitting isotopes into cancer cells: a method for spatial distribution determination of equivalent source terms. Int J Radiat Biol 2016; 92:633-640. [PMID: 27603222 DOI: 10.1080/09553002.2016.1233369] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 08/26/2016] [Accepted: 09/01/2016] [Indexed: 12/23/2022]
Abstract
PURPOSE A challenge for single-cell dosimetry of internalized Auger electron-emitting (AE) radiopharmaceuticals remains how best to elucidate their spatial distribution. To this end, a method, photoresist autoradiography (PAR), was previously developed to identify the lateral spatial distribution of AE-emitting radionuclides internalized in single cancer cells. In this paper, we present a simple mathematical model based on the radius and depth of radiation-induced patterns in photoresist material to identify the location in the z-plane of an 111In source capable of generating the pattern. MATERIALS AND METHODS SQ20B cells, derived from a head and neck squamous cell carcinoma, were exposed to 111In-labeled epidermal growth factor (EGF) (8 MBq/μg). The integrated electron fluence after four half-lives from the internalized radionuclide-containing construct was detected by a photoresist layer that was placed in close proximity to the cells. The resultant latent patterns were chemically developed and analyzed by atomic force microscopy (AFM). The features in the patterns were matched to locations of electrons emitted from simulated point sources, thereby determining the likely locations of internalized radionuclides. RESULTS The modeling procedure was validated using simple patterns. The model relates the depth and radius (in the x-y plane) of a pattern to the location and fluence of the source giving rise to the pattern. This point source modeling method provided a good fit to experimental data and can be expanded to analyze more complex patterns. CONCLUSIONS We have demonstrated the utility of the modelling technique to identify the location of internalized AE-emitting radionuclides. This methodology now needs to be extended to predict the source positions in more complex PAR patterns.
Collapse
Affiliation(s)
- Georgina Royle
- a CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology , University of Oxford , Oxford , UK
| | - Nadia Falzone
- a CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology , University of Oxford , Oxford , UK
- b Department of Biomedical Science , Tshwane University of Technology , Pretoria , South Africa
| | - Radka Chakalova
- c Department of Materials , Begbroke Science Park, University of Oxford , Oxford , UK
| | - Katherine Vallis
- a CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology , University of Oxford , Oxford , UK
| | - Sverre Myhra
- c Department of Materials , Begbroke Science Park, University of Oxford , Oxford , UK
| |
Collapse
|
22
|
Berg NG, Pearce BL, Snyder PJ, Rohrbaugh N, Nolan MW, Adhikari P, Khan SA, Ivanisevic A. Interfaces with Tunable Mechanical and Radiosensitizing Properties. ACS APPLIED MATERIALS & INTERFACES 2016; 8:21956-21961. [PMID: 26882455 DOI: 10.1021/acsami.5b11639] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
We report the fabrication of a composite containing nanostructured GaOOH and Matrigel with tunable radiosensitizing and stiffness properties. Composite characterization was done with microscopy and rheology. The utility of the interface was tested in vitro using fibroblasts. Cell viability and reactive oxygen species assays quantified the effects of radiation dosages and GaOOH concentrations. Fibroblasts' viability decreased with increasing concentration of GaOOH and composite stiffness. During ionizing radiation experiments the presence of the scintillating GaOOH triggered a different cellular response. Reactive oxygen species data demonstrated that one can reduce the amount of radiation needed to modulate the behavior of cells on interfaces with different stiffness containing a radiosensitizing material.
Collapse
Affiliation(s)
| | | | | | | | - Michael W Nolan
- Department of Clinical Sciences (College of Veterinary Medicine) and Comparative Medicine Institute, North Carolina State University , Raleigh, North Carolina 27606, United States
| | | | | | | |
Collapse
|
23
|
Palliative treatment of metastatic bone pain with radiopharmaceuticals: A perspective beyond Strontium-89 and Samarium-153. Appl Radiat Isot 2016; 110:87-99. [DOI: 10.1016/j.apradiso.2016.01.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 12/04/2015] [Accepted: 01/04/2016] [Indexed: 11/22/2022]
|
24
|
Büyüktimkin B, Stewart J, Tabanor K, Kiptoo P, Siahaan TJ. Protein and Peptide Conjugates for Targeting Therapeutics and Diagnostics to Specific Cells. Drug Deliv 2016. [DOI: 10.1002/9781118833322.ch20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
25
|
Violet JA, Farrugia G, Skene C, White J, Lobachevsky P, Martin R. Triple targeting of Auger emitters using octreotate conjugated to a DNA-binding ligand and a nuclear localizing signal. Int J Radiat Biol 2016; 92:707-715. [DOI: 10.3109/09553002.2016.1157278] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- John A. Violet
- Division of Radiation Oncology and Cancer Imaging, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - Gabriella Farrugia
- Molecular Radiation Biology Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Colin Skene
- School of Chemistry and Bio21 Institute, University of Melbourne, Melbourne, Victoria, Australia
| | - Jonathan White
- School of Chemistry and Bio21 Institute, University of Melbourne, Melbourne, Victoria, Australia
| | - Pavel Lobachevsky
- Molecular Radiation Biology Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Roger Martin
- Molecular Radiation Biology Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
26
|
Gudkov SV, Shilyagina NY, Vodeneev VA, Zvyagin AV. Targeted Radionuclide Therapy of Human Tumors. Int J Mol Sci 2015; 17:E33. [PMID: 26729091 PMCID: PMC4730279 DOI: 10.3390/ijms17010033] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2015] [Revised: 12/07/2015] [Accepted: 12/22/2015] [Indexed: 12/12/2022] Open
Abstract
Targeted radionuclide therapy is one of the most intensively developing directions of nuclear medicine. Unlike conventional external beam therapy, the targeted radionuclide therapy causes less collateral damage to normal tissues and allows targeted drug delivery to a clinically diagnosed neoplastic malformations, as well as metastasized cells and cellular clusters, thus providing systemic therapy of cancer. The methods of targeted radionuclide therapy are based on the use of molecular carriers of radionuclides with high affinity to antigens on the surface of tumor cells. The potential of targeted radionuclide therapy has markedly grown nowadays due to the expanded knowledge base in cancer biology, bioengineering, and radiochemistry. In this review, progress in the radionuclide therapy of hematological malignancies and approaches for treatment of solid tumors is addressed.
Collapse
Affiliation(s)
- Sergey V Gudkov
- Laboratory of Optical Theranostics, Lobachevsky Nizhny Novgorod State University, Gagarin Ave. 23, Nizhny Novgorod 603950, Russia.
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya St, 3, Pushchino, Moscow 142290, Russia.
- Prokhorov Institute of General Physics, Russian Academy of Sciences, Vavilova St, 38, Moscow 119991, Russia.
| | - Natalya Yu Shilyagina
- Laboratory of Optical Theranostics, Lobachevsky Nizhny Novgorod State University, Gagarin Ave. 23, Nizhny Novgorod 603950, Russia.
| | - Vladimir A Vodeneev
- Laboratory of Optical Theranostics, Lobachevsky Nizhny Novgorod State University, Gagarin Ave. 23, Nizhny Novgorod 603950, Russia.
| | - Andrei V Zvyagin
- Laboratory of Optical Theranostics, Lobachevsky Nizhny Novgorod State University, Gagarin Ave. 23, Nizhny Novgorod 603950, Russia.
- ARC Centre of Excellence for Nanoscale BioPhotonics (CNBP), Macquarie University, Sydney 2109, Australia.
| |
Collapse
|
27
|
Li JJ, Lan KL, Chang SF, Chen YF, Tsai WC, Chiang PH, Lin MH, Fischer WB, Shih YS, Yen SH, Liu RS, Tsay YG, Wang HE, Chang CA. Development and Characterization of the Recombinant Human VEGF-EGF Dual-Targeting Fusion Protein as a Drug Delivery System. Bioconjug Chem 2015; 26:2481-96. [DOI: 10.1021/acs.bioconjchem.5b00509] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Jia-Je Li
- Program
in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, Taiwan, 112
| | | | - Shun-Fu Chang
- Department
of Medical Research and Development, Chang Gung Memorial Hospital-Chiayi Branch, Chiayi 613, Taiwan
| | | | | | | | | | | | | | | | | | | | - Hsin-Ell Wang
- Program
in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, Taiwan, 112
| | | |
Collapse
|
28
|
Guerra Liberal FDC, Tavares AAS, Tavares JMRS. Comparative analysis of 11 different radioisotopes for palliative treatment of bone metastases by computational methods. Med Phys 2015; 41:114101. [PMID: 25370676 DOI: 10.1118/1.4897240] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
PURPOSE Throughout the years, the palliative treatment of bone metastases using bone seeking radiotracers has been part of the therapeutic resources used in oncology, but the choice of which bone seeking agent to use is not consensual across sites and limited data are available comparing the characteristics of each radioisotope. Computational simulation is a simple and practical method to study and to compare a variety of radioisotopes for different medical applications, including the palliative treatment of bone metastases. This study aims to evaluate and compare 11 different radioisotopes currently in use or under research for the palliative treatment of bone metastases using computational methods. METHODS Computational models were used to estimate the percentage of deoxyribonucleic acid (DNA) damage (fast Monte Carlo damage algorithm), the probability of correct DNA repair (Monte Carlo excision repair algorithm), and the radiation-induced cellular effects (virtual cell radiobiology algorithm) post-irradiation with selected particles emitted by phosphorus-32 ((32)P), strontium-89 ((89)Sr), yttrium-90 ((90)Y ), tin-117 ((117m)Sn), samarium-153 ((153)Sm), holmium-166 ((166)Ho), thulium-170 ((170)Tm), lutetium-177 ((177)Lu), rhenium-186 ((186)Re), rhenium-188 ((188)Re), and radium-223 ((223)Ra). RESULTS (223)Ra alpha particles, (177)Lu beta minus particles, and (170)Tm beta minus particles induced the highest cell death of all investigated particles and radioisotopes. The cell survival fraction measured post-irradiation with beta minus particles emitted by (89)Sr and (153)Sm, two of the most frequently used radionuclides in the palliative treatment of bone metastases in clinical routine practice, was higher than (177)Lu beta minus particles and (223)Ra alpha particles. CONCLUSIONS (223)Ra and (177)Lu hold the highest potential for palliative treatment of bone metastases of all radioisotopes compared in this study. Data reported here may prompt future in vitro and in vivo experiments comparing different radionuclides for palliative treatment of bone metastases, raise the need for the careful rethinking of the current widespread clinical use of (89)Sr and (153)Sm, and perhaps strengthen the use of (223)Ra and (177)Lu in the palliative treatment of bone metastases.
Collapse
Affiliation(s)
- Francisco D C Guerra Liberal
- Instituto de Engenharia Mecânica e Gestão Industrial, Faculdade de Engenharia, Universidade do Porto, Rua Dr. Roberto Frias s/n, Porto 4200-465, Portugal
| | - Adriana Alexandre S Tavares
- Instituto de Engenharia Mecânica e Gestão Industrial, Faculdade de Engenharia, Universidade do Porto, Rua Dr. Roberto Frias s/n, Porto 4200-465, Portugal
| | - João Manuel R S Tavares
- Instituto de Engenharia Mecânica e Gestão Industrial, Faculdade de Engenharia, Universidade do Porto, Rua Dr. Roberto Frias s/n, Porto 4200-465, Portugal
| |
Collapse
|
29
|
Duatti A. Molecular imaging with endogenous and exogenous ligands: The instance of antibodies, peptides, iodide and cupric ions. Nucl Med Biol 2015; 42:215-8. [DOI: 10.1016/j.nucmedbio.2014.11.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Revised: 11/14/2014] [Accepted: 11/15/2014] [Indexed: 02/05/2023]
|
30
|
Rosenkranz AA, Ulasov AV, Slastnikova TA, Khramtsov YV, Sobolev AS. Use of intracellular transport processes for targeted drug delivery into a specified cellular compartment. BIOCHEMISTRY (MOSCOW) 2014; 79:928-46. [DOI: 10.1134/s0006297914090090] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
31
|
Hillyar CRT, Cornelissen B, Vallis KA. Uptake, internalization and nuclear translocation of radioimmunotherapeutic agents. Ther Deliv 2014; 5:319-35. [PMID: 24592956 DOI: 10.4155/tde.14.6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2024] Open
Abstract
Radioimmunotherapy (RIT) agents that incorporate short-range particle-emitting radionuclides exploit the high linear energy transfer of α-particles and Auger electrons. Both are densely ionizing, generate complex DNA double-strand breaks and so are profoundly cytotoxic. Internalizing RIT agents enter tumor cells through receptor-mediated endocytosis and by incorporation of cell-penetrating peptides. Once internalized, some RIT agents mediate escape from endosomes and/or translocate to the nucleus. In the classical nuclear import pathway, α/β-importins recognize nuclear localization sequences in RIT agents. Translocation through nuclear pores enables RIT agents to bind to nuclear targets induced by, for example, cellular stress, growth factors or anticancer therapy, such as γH2AX or p27(KIP-1). This review discusses RIT agents designed to exploit the mechanisms underlying these complex processes and compares them with noninternalizing RIT agents.
Collapse
Affiliation(s)
- Christopher R T Hillyar
- Cancer Research UK/Medical Research Council Gray Institute for Radiation Oncology & Biology, Department of Oncology, University of Oxford, OX3 7DQ, UK
| | | | | |
Collapse
|
32
|
Elgqvist J, Frost S, Pouget JP, Albertsson P. The potential and hurdles of targeted alpha therapy - clinical trials and beyond. Front Oncol 2014; 3:324. [PMID: 24459634 PMCID: PMC3890691 DOI: 10.3389/fonc.2013.00324] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Accepted: 12/19/2013] [Indexed: 01/23/2023] Open
Abstract
This article presents a general discussion on what has been achieved so far and on the possible future developments of targeted alpha (α)-particle therapy (TAT). Clinical applications and potential benefits of TAT are addressed as well as the drawbacks, such as the limited availability of relevant radionuclides. Alpha-particles have a particular advantage in targeted therapy because of their high potency and specificity. These features are due to their densely ionizing track structure and short path length. The most important consequence, and the major difference compared with the more widely used β−-particle emitters, is that single targeted cancer cells can be killed by self-irradiation with α-particles. Several clinical trials on TAT have been reported, completed, or are on-going: four using 213Bi, two with 211At, two with 225Ac, and one with 212Pb/212Bi. Important and conceptual proof-of-principle of the therapeutic advantages of α-particle therapy has come from clinical studies with 223Ra-dichloride therapy, showing clear benefits in castration-resistant prostate cancer.
Collapse
Affiliation(s)
- Jörgen Elgqvist
- IRCM, Institut de Recherche en Cancérologie de Montpellier , Montpellier , France ; INSERM, U896 , Montpellier , France ; Université Montpellier 1 , Montpellier , France ; Institut Régional de Cancérologie de Montpellier , Montpellier , France
| | - Sofia Frost
- Fred Hutchinson Cancer Research Center , Seattle, WA , USA
| | - Jean-Pierre Pouget
- IRCM, Institut de Recherche en Cancérologie de Montpellier , Montpellier , France ; INSERM, U896 , Montpellier , France ; Université Montpellier 1 , Montpellier , France ; Institut Régional de Cancérologie de Montpellier , Montpellier , France
| | - Per Albertsson
- Department of Oncology, University of Gothenburg , Gothenburg , Sweden
| |
Collapse
|
33
|
Yoshinaga K, Oriuchi N, Wakabayashi H, Tomiyama Y, Jinguji M, Higuchi T, Kayano D, Fukuoka M, Inaki A, Toratani A, Okamoto S, Shiga T, Ito YM, Nakajo M, Nakajo M, Kinuya S. Effects and safety of ¹³¹I-metaiodobenzylguanidine (MIBG) radiotherapy in malignant neuroendocrine tumors: results from a multicenter observational registry. Endocr J 2014; 61:1171-80. [PMID: 25214026 DOI: 10.1507/endocrj.ej14-0211] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Effective treatments for malignant neuroendocrine tumors are under development. While iodine-131 metaiodobenzylguanidine (¹³¹I-MIBG) radiotherapy has been used in the treatment of malignant neuroendocrine tumors, there are few studies evaluating its therapeutic effects and safety in a multicenter cohort. In the current study, we sought to evaluate the effects and safety of ¹³¹I-MIBG therapy for conditions including malignant pheochromocytoma and paraganglioma within a multicenter cohort. Forty-eight malignant neuroendocrine tumors (37 pheochromocytoma and 11 paraganglioma) from four centers underwent clinical ¹³¹I-MIBG radiotherapy. The tumor responses were observed before and 3 to 6 months after the ¹³¹I-MIBG radiotherapy in accordance with RECIST criteria. We also evaluated the data for any adverse effects. The four centers performed a total of 87 ¹³¹I-MIBG treatments on 48 patients between January 2000 and March 2009. Of the treatments, 65 were evaluable using RECIST criteria. One partial response (PR), 40 stable disease (SD), and 9 progressive disease (PD) in malignant pheochromocytoma were observed after each treatment. Fourteen SD and one PD-were observed in paraganglioma. Patients with normal hypertension (systolic blood pressure (BP) > 130 mmHg) showed significantly reduced systolic BP after the initial follow-up (n=10, 138.1±8.2 to 129.5±13.5 mmHg, P=0.03). In adult neuroendocrine tumors with a treatment-basis analysis, there were side effects following 41 treatments (47.1%) and most of them (90.2%) were minor. In this multicenter registry, PR or SD was achieved in 84.6% of the treatment occasions in adult neuroendocrine tumors through ¹³¹I-MIBG radiotherapy. This indicated that most of the ¹³¹I-MIBG radiotherapy was performed safely without significant side effects.
Collapse
Affiliation(s)
- Keiichiro Yoshinaga
- Department of Molecular Imaging, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|