101
|
Fu H, Du B, Chen Z, Li Y. Radiolabeled Peptides for SPECT and PET Imaging in the Detection of Breast Cancer: Preclinical and Clinical Perspectives. Curr Med Chem 2021; 27:6987-7002. [PMID: 32003658 DOI: 10.2174/0929867327666200128110827] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 12/22/2019] [Accepted: 01/02/2020] [Indexed: 01/05/2023]
Abstract
Breast cancer is the most common cancer in women worldwide. Due to the heterogeneous nature of breast cancer, the optimal treatment and expected response for each patient may not necessarily be universal. Molecular imaging techniques could play an important role in the early detection and targeted therapy evaluation of breast cancer. This review focuses on the development of peptides labeled with SPECT and PET radionuclides for breast cancer imaging. We summarized the current status of radiolabeled peptides for different receptors in breast cancer. The characteristics of radionuclides and major techniques for peptide labeling are also briefly discussed.
Collapse
Affiliation(s)
- Hao Fu
- Medical College of Xiamen University, Xiamen University, Xiamen, China
| | - Bulin Du
- Department of Nuclear Medicine, The First Hospital of China Medical University, Shenyang 110001, China
| | - Zijun Chen
- Medical College of Xiamen University, Xiamen University, Xiamen, China
| | - Yesen Li
- Department of Nuclear Medicine & Minnan PET Center, Xiamen Cancer Hospital, The First Affiliated Hospital of Xiamen University, Teaching Hospital of Fujian Medical University, Xiamen, China
| |
Collapse
|
102
|
Vosoughi S, Salek N, Arani SS, Samani AB, Maragheh MG. Investigation of radiolabeling efficacy by enhancement of the chemical form of no carrier added 177Lu isolated by electro amalgamation process. Curr Radiopharm 2021; 15:56-62. [PMID: 33480353 DOI: 10.2174/1874471014666210122150134] [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: 09/28/2020] [Revised: 11/30/2020] [Accepted: 12/01/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Due to the suitable nuclear decay characteristics, 177Lu is an attractive radionuclide for various therapeutic applications. The non-carrier added form of 177Lu has drawn many attention because of its high specific activity needed in radiolabeling studies. There have been several separation methods for NCA 177Lu production. OBJECTIVES Among the various separation methods, the electro-amalgamation separation method has got a large potential for large scale production. Li presence is a significant problem in this separation method, which seriously affects the radiolabeling efficiency. METHOD In this study, Li was separated from the final product of electro-amalgamation separation by adding an ion-exchange chromatography column to the separation process. RESULTS NCA 177Lu was obtained by 84.09% ELM separation yield, 99.9% radionuclide purity and, 65 Ci/g specific activity. Then, 177Lu (177LuCl3 chemical form) was separated from Li using the ion exchange chromatography method by a separation yield of 94%. The obtained results of the radiolabeling efficacy studies showed that the radiochemical purity and radio-complex stability were significantly increased by separating of NCA 177Lu from Li. CONCLUSION This new separation setup consisting of two steps allows using 177Lu of such a favorable quality for labeling studies.
Collapse
Affiliation(s)
- Sara Vosoughi
- Radiation Application Research School, Nuclear Science and Technology Research Institute (NSTRI),Tehran. Iran
| | - Nafise Salek
- Nuclear Fuel Research School, Nuclear Science and Technology Research Institute (NSTRI). Iran
| | | | - Ali Bahrami Samani
- Nuclear Fuel Research School, Nuclear Science and Technology Research Institute (NSTRI). Iran
| | | |
Collapse
|
103
|
NAGAI Y. Production scheme for diagnostic-therapeutic radioisotopes by accelerator neutrons. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2021; 97:292-323. [PMID: 34121042 PMCID: PMC8403527 DOI: 10.2183/pjab.97.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 03/19/2021] [Indexed: 06/12/2023]
Abstract
Interest has been growing in the development of medical radioisotopes used for noninvasive nuclear medicine imaging of disease and cancer therapy. Especially the development of an alternative production scheme of 99Mo, the mother radioisotope of 99mTc used for imaging, is required, because the current supply chain of the reactor product 99Mo is fragile worldwide. We have proposed a new production scheme of 99Mo as well as therapeutic radioisotopes, such as 64Cu and 67Cu, using accelerator neutrons provided by the natC(d,n) reaction. Based on this scheme we have obtained high-quality 99mTc, 64Cu, and 67Cu suitable for clinical use by developing both production and separation methods of the radioisotopes. We proposed a new facility to constantly and reliably produce a wide variety of high-quality, carrier-free radioisotopes, including 99Mo, with accelerator neutrons. We report on the development of the proposed scheme and future prospects of the facility toward the domestic production of medical radioisotopes.
Collapse
Affiliation(s)
- Yasuki NAGAI
- Professor Emeritus, Osaka University, Suita, Osaka, Japan
- Professor Emeritus, Tokyo Institute of Technology, Tokyo, Japan
- QST-Associate, National Institute for Quantum and Radiological Science and Technology, Tokai, Ibaraki, Japan
- Research Professor, Tohoku University, Sendai, Miyagi, Japan
- Research Fellow, Chiyoda Technol Co., Tokyo, Japan
| |
Collapse
|
104
|
Analysis of lutetium-177 production at the WWR-K research reactor. Appl Radiat Isot 2020; 169:109561. [PMID: 33360502 DOI: 10.1016/j.apradiso.2020.109561] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 12/06/2020] [Accepted: 12/07/2020] [Indexed: 11/23/2022]
Abstract
Production of lutetium-177 using direct nuclear reaction 176Lu(n,γ)177Lu by WWR-K reactor neutrons on enriched LuCl3 (up to 82% of 176Lu) is described. Calculations were performed by MCNP6 transport code. Two different irradiation positions of the WWR-K research reactor were considered. Estimates of the maximum specific activity of the luthetium-177 are obtained for the reactor irradiation positions located: (a) in the reactor core centre, (b) in the core periphery. In these positions, thermal neutron flux is two times different. Experimental data was shown that k-factor is 1.5 for considered irradiation positions. The study shows that for the position located in the core center, the estimated maximum specific activity of lutetium-177 is 819 GBq/mg, is to be achieved after 15 days of irradiation. For the position located in the core periphery, specific activity of lutetium-177 is 561 GBq/mg, is to be achieved after 20 days of irradiation. Ratio of Lu-177m to Lu-177 specific activity is not more than 0.025 for both irradiation positions.
Collapse
|
105
|
Tan HY, Yeong CH, Wong YH, McKenzie M, Kasbollah A, Md Shah MN, Perkins AC. Neutron-activated theranostic radionuclides for nuclear medicine. Nucl Med Biol 2020; 90-91:55-68. [PMID: 33039974 DOI: 10.1016/j.nucmedbio.2020.09.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 09/08/2020] [Accepted: 09/22/2020] [Indexed: 12/14/2022]
Abstract
Theranostics in nuclear medicine refers to personalized patient management that involves targeted therapy and diagnostic imaging using a single or combination of radionuclide (s). The radionuclides emit both alpha (α) or beta (β-) particles and gamma (γ) rays which possess therapeutic and diagnostic capabilities, respectively. However, the production of these radionuclides often faces difficulties due to high cost, complexity of preparation methods and that the products are often sourced far from the healthcare facilities, hence losing activity due to radioactive decay during transportation. Subject to the availability of a nuclear reactor within an accessible distance from healthcare facilities, neutron activation is the most practical and cost-effective route to produce radionuclides suitable for theranostic purposes. Holmium-166 (166Ho), Lutetium-177 (177Lu), Rhenium-186 (186Re), Rhenium-188 (188Re) and Samarium-153 (153Sm) are some of the most promising neutron-activated radionuclides that are currently in clinical practice and undergoing clinical research for theranostic applications. The aim of this paper is to review the physical characteristics, current clinical applications and future prospects of these neutron activated radionuclides in theranostics. The production, physical properties, validated clinical applications and clinical studies for each neutron-activated radionuclide suitable for theranostic use in nuclear medicine are reviewed in this paper.
Collapse
Affiliation(s)
- Hun Yee Tan
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University, 47500 Subang Jaya, Selangor, Malaysia
| | - Chai Hong Yeong
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University, 47500 Subang Jaya, Selangor, Malaysia
| | - Yin How Wong
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University, 47500 Subang Jaya, Selangor, Malaysia
| | - Molly McKenzie
- School of Life Sciences, University of Dundee, DD1 4HN, United Kingdom
| | - Azahari Kasbollah
- Medical Technology Division, Malaysian Nuclear Agency, Bangi, 43000 Kajang, Selangor, Malaysia
| | - Mohamad Nazri Md Shah
- Department of Biomedical Imaging, University of Malaya Medical Centre, 59100 Kuala Lumpur, Malaysia
| | - Alan Christopher Perkins
- Radiological Sciences, School of Medicine, University of Nottingham, Nottingham NG7 2UH, United Kingdom.
| |
Collapse
|
106
|
Heesch A, Maurer J, Stickeler E, Beheshti M, Mottaghy FM, Morgenroth A. Development of Radiotracers for Breast Cancer-The Tumor Microenvironment as an Emerging Target. Cells 2020; 9:cells9102334. [PMID: 33096754 PMCID: PMC7590199 DOI: 10.3390/cells9102334] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/19/2020] [Accepted: 10/19/2020] [Indexed: 12/17/2022] Open
Abstract
Molecular imaging plays an increasingly important role in the diagnosis and treatment of different malignancies. Radiolabeled probes enable the visualization of the primary tumor as well as the metastases and have been also employed in targeted therapy and theranostic approaches. With breast cancer being the most common malignancy in women worldwide it is of special interest to develop novel targeted treatments. However, tumor microenvironment and escape mechanisms often limit their therapeutic potential. Addressing tumor stroma associated targets provides a promising option to inhibit tumor growth and angiogenesis and to disrupt tumor tissue architecture. This review describes recent developments on radiolabeled probes used in diagnosis and treatment of breast cancer especially in triple negative type with the focus on potential targets offered by the tumor microenvironment, like tumor associated macrophages, cancer associated fibroblasts, and endothelial cells.
Collapse
Affiliation(s)
- Amelie Heesch
- Department of Nuclear Medicine, University Hospital Aachen, RWTH Aachen University, 52074 Aachen, Germany; (A.H.); (M.B.); (F.M.M.)
| | - Jochen Maurer
- Department of Obstetrics and Gynecology, University Hospital Aachen (UKA), 52074 Aachen, Germany; (J.M.); (E.S.)
| | - Elmar Stickeler
- Department of Obstetrics and Gynecology, University Hospital Aachen (UKA), 52074 Aachen, Germany; (J.M.); (E.S.)
| | - Mohsen Beheshti
- Department of Nuclear Medicine, University Hospital Aachen, RWTH Aachen University, 52074 Aachen, Germany; (A.H.); (M.B.); (F.M.M.)
- Department of Nuclear Medicine, University Hospital Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Felix M. Mottaghy
- Department of Nuclear Medicine, University Hospital Aachen, RWTH Aachen University, 52074 Aachen, Germany; (A.H.); (M.B.); (F.M.M.)
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center (MUMC+), 6202 Maastricht, The Netherlands
| | - Agnieszka Morgenroth
- Department of Nuclear Medicine, University Hospital Aachen, RWTH Aachen University, 52074 Aachen, Germany; (A.H.); (M.B.); (F.M.M.)
- Correspondence:
| |
Collapse
|
107
|
Bhardwaj R, Wolterbeek HT, Denkova AG, Serra-Crespo P. Modelling of the 177mLu/ 177Lu radionuclide generator. Appl Radiat Isot 2020; 166:109261. [PMID: 32961525 DOI: 10.1016/j.apradiso.2020.109261] [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: 10/07/2019] [Revised: 04/24/2020] [Accepted: 06/01/2020] [Indexed: 10/23/2022]
Abstract
In order to determine the potential of 177mLu/177Lu radionuclide generator in 177Lu production it is important to establish the technical needs that can lead to a clinically acceptable 177Lu product quality. In this work, a model that includes all the processes and the parameters affecting the performance of the 177mLu/177Lu radionuclide generator has been developed. The model has been based on the use of a ligand to complex 177mLu ions, followed by the separation of the freed 177Lu ions. The dissociation kinetics of the Lu-ligand complex has been found to be the most crucial aspect governing the specific activity and 177mLu content of the produced 177Lu. The dissociation rate constants lower than 1*10-11 s-1 would be required to lead to onsite 177Lu production with specific activity close to theoretical maximum of 4.1 TBq 177Lu/mg Lu and with 177mLu content of less than 0.01%. Lastly, the calculations suggest that more than one patient dose per week can be supplied for a period of up to 7 months on starting with the 177mLu produced using 3 g Lu2O3 target with 60% 176Lu enrichment. The requirements of the starting 177mLu activity production needs to be adapted depending on the required patient doses, and the technical specifications of the involved 177mLu-177Lu separation process.
Collapse
Affiliation(s)
- Rupali Bhardwaj
- Applied Radiation and Isotopes, Department of Radiation Science and Technology, Faculty of Applied Sciences, Delft University of Technology, Mekelweg 15, 2629 JB, Delft, the Netherlands; Catalysis Engineering, Department of Chemical Engineering, Faculty of Applied Sciences, Delft University of Technology, Van der Maasweg 9, 2629 HZ, Delft, the Netherlands
| | - Hubert T Wolterbeek
- Applied Radiation and Isotopes, Department of Radiation Science and Technology, Faculty of Applied Sciences, Delft University of Technology, Mekelweg 15, 2629 JB, Delft, the Netherlands
| | - Antonia G Denkova
- Applied Radiation and Isotopes, Department of Radiation Science and Technology, Faculty of Applied Sciences, Delft University of Technology, Mekelweg 15, 2629 JB, Delft, the Netherlands
| | - Pablo Serra-Crespo
- Applied Radiation and Isotopes, Department of Radiation Science and Technology, Faculty of Applied Sciences, Delft University of Technology, Mekelweg 15, 2629 JB, Delft, the Netherlands.
| |
Collapse
|
108
|
Kimberlin A, Nash KL. Dimerization of 2-Ethylhexylphosphonic Acid Mono-2-ethylhexyl Ester (HEH[EHP]) as Determined by NMR Spectrometry. SOLVENT EXTRACTION AND ION EXCHANGE 2020. [DOI: 10.1080/07366299.2020.1816610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
| | - Kenneth L. Nash
- Department of Chemistry, Washington State University, Pullman, WA, USA
| |
Collapse
|
109
|
Targeted Tumor Therapy with Radiolabeled DNA Intercalator: A Possibility? Preclinical Investigations with 177Lu-Acridine. BIOMED RESEARCH INTERNATIONAL 2020; 2020:9514357. [PMID: 32775454 PMCID: PMC7397433 DOI: 10.1155/2020/9514357] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/25/2020] [Accepted: 06/29/2020] [Indexed: 01/12/2023]
Abstract
Objective A DNA intercalating agent reversibly stacks between the adjacent base pairs of DNA and thus is expected to exhibit preferential localization in the tumorous lesions as tumors are associated with enhanced DNA replication. Therefore, radiolabeled DNA intercalators are supposed to have potential to be used in targeted tumor therapy. Working in this direction, an attempt was made to radiolabel 9-aminoacridine, a DNA intercalator, with 177Lu, one of the most useful therapeutic radionuclides, and study the potential of 177Lu-acridine in targeted tumor therapy. Experiments. 9-Aminoacridine was coupled with p-NCS-benzyl-DOTA to facilitate radiolabeling, and the conjugate was radiolabeled with 177Lu. Different reaction parameters were optimized in order to obtain 177Lu-acridine complex with maximum radiochemical purity. In vitro stability of the radiolabeled complex was studied in normal saline and human blood serum. Biological behavior of the radiolabeled agent was studied both in vitro and in vivo using the Raji cell line and fibrosarcoma tumor-bearing Swiss mice, respectively. Results 177Lu-acridine complex was obtained with ~100% radiochemical purity under the optimized reaction conditions involving incubation of 1.5 mg/mL of ligand with 177Lu (1 mCi, 37 MBq) at 100°C at pH ~5 for 45 minutes. The complex maintained a radiochemical purity of >85% in saline at 6 d and >70% in human serum at 2 d postpreparation. In vitro cellular study showed uptake of the radiotracer (5.3 ± 0.13%) in the Raji cells along with significant cytotoxicity (78.06 ± 2.31% after 6 d). Biodistribution study revealed considerable accumulation of the radiotracer in tumor 9.98 ± 0.13 %ID/g within 1 h postadministration and retention therein till 6 d postadministration 4.00 ± 0.16 %ID/g with encouraging tumor to nontarget organ uptake ratios. Conclusions The present study, although preliminary, indicates the potential of 177Lu-acridine and thus radiolabeled DNA intercalators in targeted tumor therapy. However, further detailed evaluation is required to explore the actual potential of such agents in targeted tumor therapy.
Collapse
|
110
|
Suman S, Priya R, Kameswaran M. Induction of different cellular arrest and molecular responses in low EGFR expressing A549 and high EGFR expressing A431 tumor cells treated with various doses of 177Lu-Nimotuzumab. Int J Radiat Biol 2020; 96:1144-1156. [PMID: 32657634 DOI: 10.1080/09553002.2020.1793012] [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] [Indexed: 01/22/2023]
Abstract
INTRODUCTION Radioimmunotherapy (RIT) is a major anti-cancer therapy in cancer management multimodalities. 177Lu-Nimotuzumab has been in the use for radioimmunotherapy of EGFR expressing tumors. This study focuses on understanding the differential cellular and molecular mechanisms of anti-tumor effects of 177Lu-Nimotuzumab on low EGFR expressing A549 and high EGFR expressing A431 tumor cells. MATERIALS AND METHODS Nimotuzumab labeled with 177Lu was characterized by SE-HPLC. Specificity of 177Lu-Nimotuzumab to EGFR expressed on A549 and A431 cells was confirmed by competitive assay using increasing amounts of unlabeled Nimotuzumab. Cellular responses of A549 (low EGFR) and A431 (high EGFR) in response to different doses of 177Lu-Nimotuzumab were determined by Viable count assay for cellular viability, cell-cycle analysis by DNA staining, apoptotic assay for cell death, and CFSE dilution assay for cellular proliferation capacity. The number of DNA DSBs formed was determined using γ-H2AX assay with PI staining. Transcription of genes involved in DNA damage response and repair (DRR) pathways was monitored by RT-qPCR. RESULTS 177Lu-Nimotuzumab characterized by SE-HPLC exhibited a radiochemical purity of 99.1 ± 0.6%. Cell binding competition studies with 177Lu-Nimotuzumab showed specific binding of 34.3 ± 1.7% with A431 cells and 18.4 ± 1.9% with A549 cells which decreased when co-incubated with unlabeled Nimotuzumab. Cytotoxicity and DNA damage (DNA DSBs) increased with an increase in the dose of 177Lu-Nimotuzumab. A549 displayed G2/M arrest while A431 showed G1 arrest. Apoptotic death was determined to be one of the modes of death of arrested A549 and A431 cells which increases with the increase in the dose of 177Lu-Nimotuzumab. Loss of proliferation capacity was higher in A431 showed by CFSE staining at different doses of 177Lu-Nimotuzumab. Transcription profile of most DRR genes in A431 and A549 showed a decrease in the transcription at 4 h followed by recovery at 16 h post-treatment. The degree of transcription of most DRR genes was similar, irrespective of 177Lu-Nimotuzumab dose. CONCLUSION 177Lu-Nimotuzumab induces different cellular arrest and molecular responses in low EGFR expressing A549 and high EGFR expressing A431 tumor cells. This study would enable the development of integrative novel treatment strategies for radioimmunotherapy in low and high EGFR expressing tumors by 177Lu-Nimotuzumab with therapeutic gains.
Collapse
Affiliation(s)
- ShishuKant Suman
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Mumbai, India
| | - Rashmi Priya
- Radiation Biology and Health Sciences Division, Bhabha Atomic Research Centre, Mumbai, India
| | - Mythili Kameswaran
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Mumbai, India
| |
Collapse
|
111
|
Pirooznia N, Abdi K, Beiki D, Emami F, Arab SS, Sabzevari O, Soltani-Gooshkhaneh S. 177Lu-labeled cyclic RGD peptide as an imaging and targeted radionuclide therapeutic agent in non-small cell lung cancer: Biological evaluation and preclinical study. Bioorg Chem 2020; 102:104100. [PMID: 32711088 DOI: 10.1016/j.bioorg.2020.104100] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 07/10/2020] [Accepted: 07/11/2020] [Indexed: 12/29/2022]
Abstract
Non-small cell lung carcinoma (NSCLC) is among the most lethal lung cancers responsible for 80-85% of death. αvβ3 integrin receptor subtype has been identified as a lung cancer biomarker since its expression correlates with tumor progression and metastasis. The extracellular domain of the receptor forms a binding site for RGD-based sequences. Therefore, specific targeting of αvβ3 integrin receptors by these short peptides can be an excellent candidate for cancer imaging and therapy. In this research, the radiolabeling of DOTA-E(cRGDfK)2 with 177Lu was efficiently implemented. The Log P value, in vivo, in vitro, metabolic stability, cellular uptake and specific binding of the radiopeptide was determined. The tumor targeting capacity and the therapeutic potential of the radiotracer was studied in A549 tumor-bearing mice. Imaging studies at different time intervals were performed by SPECT/CT. Radiochemical purity of more than 99% and Log P of -3.878 was obtained for 177Lu-labelled peptide. Radiotracer showed favorable in vivo, in vitro and metabolic stability. The radiopeptide dissociation constant (Kd) was 15.07 nM. Radiopeptide specific binding was more than 95%. Biodistribution studies showed high accumulation of the radiopeptide in tumor and rapid excretion by urinary route. Maximum tumor uptake was at 4 h post-injection. Following administration of this radiopeptide to mice, not only tumor growth was suppressed, but significant tumor shrinkage was also observed. In conclusion, this radiopeptide can be employed for staging, follow-up imaging and as peptide receptor radionuclide therapeutic agent allowing efficient therapy for NSCLC and other cancers overexpressing αvβ3 integrin receptors.
Collapse
Affiliation(s)
- Nazanin Pirooznia
- Department of Radiopharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Khosrou Abdi
- Department of Radiopharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| | - Davood Beiki
- Research Center for Nuclear Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Farshad Emami
- Nuclear Medicine and Molecular Imaging Department, Imam Reza International University, Razavi Hospital, Mashhad, Iran
| | - Seyed Shahriar Arab
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Omid Sabzevari
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Toxicology and Poisoning Research Centre, Tehran University of Medical Sciences, Tehran, Iran; Toxicology and Poisoning Research Centre, Tehran University of Medical Sciences, Tehran, Iran
| | - Samira Soltani-Gooshkhaneh
- Nuclear Medicine and Molecular Imaging Department, Imam Reza International University, Razavi Hospital, Mashhad, Iran
| |
Collapse
|
112
|
Sherman M, Levine R. Nuclear Medicine and Wall Street: An Evolving Relationship. J Nucl Med 2020; 60:20S-24S. [PMID: 31481585 DOI: 10.2967/jnumed.118.220798] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 04/19/2019] [Indexed: 11/16/2022] Open
Abstract
Until recently, it has been challenging to engage Wall Street and large pharmaceutical companies in radiopharmaceutical opportunities. The modest economic prospects of most diagnostic radiopharmaceuticals have not attracted keen interest from the broader business community, despite the rapid advancement of diagnostic imaging capabilities and their increasingly crucial role in the therapeutic process. Similarly, compelling science supporting select radiopharmaceutical therapies in oncology has been overshadowed by the unique challenges posed by this class of drugs and historical commercial failures that serve as sobering reminders of risk. Fortunately, a few notable successes in the targeted radioligand therapeutic space are changing this dynamic, fueling a new flow of investor capital into these technologies and inciting increased merger and acquisition activity that has yielded significant value creation for investors. If the nuclear medicine industry is able to continue to effectively manage historical challenges, then there is significant opportunity for a new and promising wave of radioligand therapies to significantly change the oncology treatment paradigm and elevate the profile of the entire nuclear medicine sector.
Collapse
Affiliation(s)
| | - Rachel Levine
- Communications, Advanced Accelerator Applications, USA, Inc., Millburn, New Jersey
| |
Collapse
|
113
|
Estimation of 47Sc and 177Lu production rates from their natural targets in Kyoto University Research Reactor. J Radioanal Nucl Chem 2020. [DOI: 10.1007/s10967-020-07156-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
114
|
Investigation of the deuteron induced nuclear reaction cross sections on lutetium up to 50 MeV: review of production routes for 177Lu, 175Hf and 172Hf via charged particle activation. J Radioanal Nucl Chem 2020. [DOI: 10.1007/s10967-020-07182-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
AbstractIn a systematic study of light charged particle induced nuclear reactions we investigated the excitation functions of deuteron induced nuclear reactions on natural lutetium targets. Experimental excitation functions up to 50 MeV on high purity natLu were determined using the standard stacked foil activation technique. High resolution off-line gamma-ray spectrometry was applied to assess the activity of each foil. From the measured activity direct and/or cumulative elemental cross-section data for production of 171,172,173,175Hf, 171,172,173,174g,176m,177m,177gLu and 169Yb radioisotopes were determined. The experimental data were compared to results of the TALYS theoretical code taken from the TENDL databases and results of our calculations using the ALICE-IPPE-D and the EMPIRE-D codes. No earlier experimental data were found in the literature. Thick target yields for the investigated radionuclides were calculated from the measured excitation functions.
Collapse
|
115
|
Kostelnik TI, Wang X, Southcott L, Wagner HK, Kubeil M, Stephan H, Jaraquemada-Peláez MDG, Orvig C. Rapid Thermodynamically Stable Complex Formation of [ nat/111In]In 3+, [ nat/90Y]Y 3+, and [ nat/177Lu]Lu 3+ with H 6dappa. Inorg Chem 2020; 59:7238-7251. [PMID: 32337985 DOI: 10.1021/acs.inorgchem.0c00671] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
A phosphinate-bearing picolinic acid-based chelating ligand (H6dappa) was synthesized and characterized to assess its potential as a bifunctional chelator (BFC) for inorganic radiopharmaceuticals. Nuclear magnetic resonance (NMR) spectroscopy was employed to investigate the chelator coordination chemistry with a variety of nonradioactive trivalent metal ions (In3+, Lu3+, Y3+, Sc3+, La3+, Bi3+). Density functional theory (DFT) calculations explored the coordination environments of aforementioned metal complexes. The thermodynamic stability of H6dappa with four metal ions (In3+, Lu3+, Y3+, Sc3+) was deeply investigated via potentiometric and spectrophotometric (UV-vis) titrations, employing a combination of acidic in-batch, joint potentiometric/spectrophotometric, and ligand-ligand competition titrations; high stability constants and pM values were calculated for all four metal complexes. Radiolabeling conditions for three clinically relevant radiometal ions were optimized ([111In]In3+, [177Lu]Lu3+, [90Y]Y3+), and the serum stability of [111In][In(dappa)]3- was studied. Through concentration-, time-, temperature-, and pH-dependent labeling experiments, it was determined that H6dappa radiolabels most effectively at near-physiological pH for all radiometal ions. Furthermore, very rapid radiolabeling at ambient temperature was observed, as maximal radiolabeling was achieved in less than 1 min. Molar activities of 29.8 GBq/μmol and 28.2 GBq/μmol were achieved for [111In]In3+ and [177Lu]Lu3+, respectively. For H6dappa, high thermodynamic stability did not correlate with kinetic inertness-lability was observed in serum stability studies, suggesting that its metal complexes might not be suitable as a BFC in radiopharmaceuticals.
Collapse
Affiliation(s)
- Thomas I Kostelnik
- Medicinal Inorganic Chemistry Group, Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, British Columbia V6T 1Z1, Canada.,Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, British Columbia V6T 2A3, Canada
| | - Xiaozhu Wang
- Medicinal Inorganic Chemistry Group, Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, British Columbia V6T 1Z1, Canada
| | - Lily Southcott
- Medicinal Inorganic Chemistry Group, Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, British Columbia V6T 1Z1, Canada.,Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, British Columbia V6T 2A3, Canada
| | - Hannah K Wagner
- Medicinal Inorganic Chemistry Group, Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, British Columbia V6T 1Z1, Canada.,Anorganish-Chemisches Institut, Universität Heidelberg, Im Neuenheimer Feld 276, 69120 Heidelberg, Germany
| | - Manja Kubeil
- Institute of Radiopharmaceutical Cancer Research, Helmholtz - Zentrum Dresden - Rossendorf, Bautzner Landstrasse 400, 01328 Dresden, Germany
| | - Holger Stephan
- Institute of Radiopharmaceutical Cancer Research, Helmholtz - Zentrum Dresden - Rossendorf, Bautzner Landstrasse 400, 01328 Dresden, Germany
| | - María de Guadalupe Jaraquemada-Peláez
- Medicinal Inorganic Chemistry Group, Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, British Columbia V6T 1Z1, Canada
| | - Chris Orvig
- Medicinal Inorganic Chemistry Group, Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, British Columbia V6T 1Z1, Canada
| |
Collapse
|
116
|
Ramonaheng K, van Staden JA, du Raan H. Validation of a Monte Carlo modelled gamma camera for Lutetium-177 imaging. Appl Radiat Isot 2020; 163:109200. [PMID: 32561041 DOI: 10.1016/j.apradiso.2020.109200] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 02/11/2020] [Accepted: 04/08/2020] [Indexed: 10/24/2022]
Abstract
This study validated a model of the Siemens Symbia T16 dual-head SPECT/CT gamma camera created using the Monte Carlo program SIMIND for 177Lu. The validation was done by comparing experimental and simulated gamma camera performance criteria tests for the 177Lu 208 keV photopeak with a medium-energy collimator. Results showed good agreement between the experimental and simulated values. These results illustrated that SIMIND could emulate the Symbia T16 successfully and therefore, can be used with confidence to model 177Lu images.
Collapse
Affiliation(s)
- K Ramonaheng
- Department of Medical Physics, University of the Free State, PO Box 339, Bloemfontein, 9300, South Africa.
| | - J A van Staden
- Department of Medical Physics, University of the Free State, PO Box 339, Bloemfontein, 9300, South Africa
| | - H du Raan
- Department of Medical Physics, University of the Free State, PO Box 339, Bloemfontein, 9300, South Africa
| |
Collapse
|
117
|
Czerwińska M, Bilewicz A, Kruszewski M, Wegierek-Ciuk A, Lankoff A. Targeted Radionuclide Therapy of Prostate Cancer-From Basic Research to Clinical Perspectives. Molecules 2020; 25:E1743. [PMID: 32290196 PMCID: PMC7181060 DOI: 10.3390/molecules25071743] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 03/23/2020] [Accepted: 04/07/2020] [Indexed: 12/18/2022] Open
Abstract
Prostate cancer is the most commonly diagnosed malignancy in men and the second leading cause of cancer-related deaths in Western civilization. Although localized prostate cancer can be treated effectively in different ways, almost all patients progress to the incurable metastatic castration-resistant prostate cancer. Due to the significant mortality and morbidity rate associated with the progression of this disease, there is an urgent need for new and targeted treatments. In this review, we summarize the recent advances in research on identification of prostate tissue-specific antigens for targeted therapy, generation of highly specific and selective molecules targeting these antigens, availability of therapeutic radionuclides for widespread medical applications, and recent achievements in the development of new-generation small-molecule inhibitors and antibody-based strategies for targeted prostate cancer therapy with alpha-, beta-, and Auger electron-emitting radionuclides.
Collapse
Affiliation(s)
- Malwina Czerwińska
- Centre for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195 Warsaw, Poland; (M.C.); (M.K.)
| | - Aleksander Bilewicz
- Centre of Radiochemistry and Nuclear Chemistry, Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195 Warsaw, Poland;
| | - Marcin Kruszewski
- Centre for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195 Warsaw, Poland; (M.C.); (M.K.)
- Department of Molecular Biology and Translational Research, Institute of Rural Health, Jaczewskiego 2, 20-090 Lublin, Poland
| | - Aneta Wegierek-Ciuk
- Department of Medical Biology, Institute of Biology, Jan Kochanowski University, Uniwersytecka 7, 24-406 Kielce, Poland;
| | - Anna Lankoff
- Centre for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195 Warsaw, Poland; (M.C.); (M.K.)
- Department of Medical Biology, Institute of Biology, Jan Kochanowski University, Uniwersytecka 7, 24-406 Kielce, Poland;
| |
Collapse
|
118
|
Sharma R, Kameswaran M, Dash A. Comparative In Vitro Cytotoxicity Studies of 177Lu-CHX-A″-DTPA-Trastuzumab and 177Lu-CHX-A″-DTPA-F(ab') 2-Trastuzumab in HER2-Positive Cancer Cell Lines. Cancer Biother Radiopharm 2020; 35:177-189. [PMID: 32196365 DOI: 10.1089/cbr.2019.2882] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background: Human epidermal growth factor receptor 2 (HER2) is found to be amplified in ∼15%-20% of breast cancers. In this study, the authors report the synthesis and comparative in vitro therapeutic efficacy of 177Lu-CHX-A″-DTPA-trastuzumab and 177Lu-CHX-A″-DTPA-F(ab')2-trastuzumab to determine their potential as theranostic agents for patients with breast cancer. Materials and Methods: Bivalent F(ab')2-trastuzumab was produced by enzymatic digestion of trastuzumab, conjugated with p-SCN-Bn-CHX-A″-DTPA and subsequently radiolabeled with 177Lu. Cell viability, membrane toxicity assays, and apoptosis analysis were carried out with 177Lu-CHX-A″-DTPA-trastuzumab and 177Lu-CHX-A″-DTPA-F(ab')2-trastuzumab in HER2-positive ovarian (SK-OV-3) and breast cancer (SK-BR-3 and MDA-MB-453) cells. Results: In vitro cell binding studies showed ∼20%-25% binding of 177Lu-CHX-A″-DTPA-trastuzumab and 177Lu-CHX-A″-DTPA-F(ab')2-trastuzumab to SK-OV-3, SK-BR-3, and MDA-MB-453 cells. The cells exhibited similar degree of membrane integrity and cellular toxicity when treated with same amount (activity) of 177Lu-CHX-A″-DTPA-F(ab')2-trastuzumab and 177Lu-CHX-A″-DTPA-trastuzumab, and the toxicity was dose dependent. The mode of cell death was predominantly by apoptosis and necrosis with both the radioimmunoconjugates. Conclusions: The results indicated that the efficacy of both the radioimmunoconjugates, in terms of inducing cell death, was similar thereby ascertaining their potential as good therapeutic agents for patients with breast cancer.
Collapse
Affiliation(s)
- Rohit Sharma
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Mumbai, India
| | - Mythili Kameswaran
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Mumbai, India
| | - Ashutosh Dash
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Mumbai, India.,Homi Bhabha National Institute, Mumbai, India
| |
Collapse
|
119
|
Talip Z, Favaretto C, Geistlich S, van der Meulen NP. A Step-by-Step Guide for the Novel Radiometal Production for Medical Applications: Case Studies with 68Ga, 44Sc, 177Lu and 161Tb. Molecules 2020; 25:E966. [PMID: 32093425 PMCID: PMC7070971 DOI: 10.3390/molecules25040966] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 02/16/2020] [Accepted: 02/17/2020] [Indexed: 02/06/2023] Open
Abstract
The production of novel radionuclides is the first step towards the development of new effective radiopharmaceuticals, and the quality thereof directly affects the preclinical and clinical phases. In this review, novel radiometal production for medical applications is briefly elucidated. The production status of the imaging nuclide 44Sc and the therapeutic β--emitter nuclide 161Tb are compared to their more established counterparts, 68Ga and 177Lu according to their targetry, irradiation process, radiochemistry, and quality control aspects. The detailed discussion of these significant issues will help towards the future introduction of these promising radionuclides into drug manufacture for clinical application under Good Manufacturing Practice (GMP).
Collapse
Affiliation(s)
- Zeynep Talip
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232 Villigen-PSI, Switzerland
| | - Chiara Favaretto
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232 Villigen-PSI, Switzerland
- Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Susanne Geistlich
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232 Villigen-PSI, Switzerland
- Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Nicholas P. van der Meulen
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232 Villigen-PSI, Switzerland
- Laboratory of Radiochemistry, Paul Scherrer Institute, 5232 Villigen-PSI, Switzerland
| |
Collapse
|
120
|
Ebbers SC, Braat AJAT, Moelker A, Stokkel MPM, Lam MGEH, Barentsz MW. Intra-arterial versus standard intravenous administration of lutetium-177-DOTA-octreotate in patients with NET liver metastases: study protocol for a multicenter, randomized controlled trial (LUTIA trial). Trials 2020; 21:141. [PMID: 32024533 PMCID: PMC7003409 DOI: 10.1186/s13063-019-3888-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 11/05/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Lutetium-177-DOTA-octreotate (177Lu-DOTATATE) significantly increases survival and response rates in patients with grade I and grade II neuroendocrine tumors (NETs). However, survival and response rates are significantly lower in patients with bulky liver metastases. Increasing the tumor-absorbed dose in liver metastases may improve response to 177Lu-DOTATATE. The LUTIA (Lutetium Intra-Arterial) study aims to increase the tumor-absorbed dose in liver metastases by intra-arterial (IA) administration of 177Lu-DOTATATE, compared to conventional intravenous (IV) administration. METHODS A multicenter, within-patient randomized controlled trial (RCT) in 26 patients with progressive, liver-dominant, unresectable grade I or grade II NET will be conducted. Patients with bilobar bulky disease will be randomly allocated to receive IA treatment into either the left or the right hepatic artery. Using this approach, one liver lobe will be treated intra-arterially (first-pass effect), while the contralateral lobe will receive an intravenous treatment as a second-pass effect. The primary endpoint of this study is the difference in tumor-to-non-tumor ratio of 177Lu-DOTATATE uptake between the two liver lobes on post-treatment SPECT/CT (IA versus IV). Secondary endpoints include absorbed dose in both liver lobes, tumor response, dose-response relationship, toxicity, uptake in extrahepatic lesions, and renal uptake. DISCUSSION This multicenter, within-patient RCT will investigate whether IA administration of 177Lu-DOTATATE results in a higher activity concentration in liver metastases compared to IV administration. TRIAL REGISTRATION ClinicalTrials.gov, NCT03590119. Registered on 17 July 2018.
Collapse
Affiliation(s)
- Sander C Ebbers
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Arthur J A T Braat
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Adriaan Moelker
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Marcel P M Stokkel
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Marnix G E H Lam
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Maarten W Barentsz
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands.
| |
Collapse
|
121
|
Rousseau E, Lau J, Zhang Z, Zhang C, Kwon D, Uribe CF, Kuo HT, Zeisler J, Bratanovic I, Lin KS, Bénard F. Comparison of biological properties of [ 177 Lu]Lu-ProBOMB1 and [ 177 Lu]Lu-NeoBOMB1 for GRPR targeting. J Labelled Comp Radiopharm 2020; 63:56-64. [PMID: 31715025 DOI: 10.1002/jlcr.3815] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 10/15/2019] [Accepted: 11/04/2019] [Indexed: 11/06/2022]
Abstract
The gastrin-releasing peptide receptor (GRPR) is overexpressed in prostate cancer and other solid malignancies. Following up on our work on [68 Ga]Ga-ProBOMB1 that had better imaging characteristics than [68 Ga]Ga-NeoBOMB1, we investigated the effects of substituting 68 Ga for 177 Lu to determine if the resulting radiopharmaceuticals could be used with a therapeutic aim. We radiolabeled the bombesin antagonist ProBOMB1 (DOTA-pABzA-DIG-D-Phe-Gln-Trp-Ala-Val-Gly-His-Leu-ψ-Pro-NH2 ) with lutetium-177 and compared it with [177 Lu]Lu-NeoBOMB1 (obtained in 54.2 ± 16.5% isolated radiochemical yield with >96% radiochemical purity and 440.8 ± 165.1 GBq/μmol molar activity) for GRPR targeting. Lu-NeoBOMB1 had better binding affinity for GRPR than Lu-ProBOMB1 (Ki values: 2.26 ± 0.24 and 30.2 ± 3.23nM). [177 Lu]Lu-ProBOMB1 was obtained in 53.7 ± 5.4% decay-corrected radiochemical yield with 444.2 ± 193.2 GBq/μmol molar activity and >95% radiochemical purity. In PC-3 prostate cancer xenograft mice, tumor uptake of [177 Lu]Lu-ProBOMB1 was 3.38 ± 1.00, 1.32 ± 0.24, and 0.31 ± 0.04%ID/g at 1, 4, and 24 hours pi. However, the uptake in tumor was lower than [177 Lu]Lu-NeoBOMB1 at all time points. [177 Lu]Lu-ProBOMB1 was inferior to [177 Lu]Lu-NeoBOMB1, which had better therapeutic index for the organs receiving the highest doses.
Collapse
Affiliation(s)
- Etienne Rousseau
- Department of Molecular Oncology, BC Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Joseph Lau
- Department of Molecular Oncology, BC Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Zhengxing Zhang
- Department of Molecular Oncology, BC Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Chengcheng Zhang
- Department of Molecular Oncology, BC Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Daniel Kwon
- Department of Molecular Oncology, BC Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Carlos F Uribe
- Department of Molecular Oncology, BC Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Hsiou-Ting Kuo
- Department of Molecular Oncology, BC Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Jutta Zeisler
- Department of Molecular Oncology, BC Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Ivica Bratanovic
- Department of Molecular Oncology, BC Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Kuo-Shyan Lin
- Department of Molecular Oncology, BC Cancer Research Centre, Vancouver, British Columbia, Canada
- Department of Radiology, University of British Columbia, Vancouver, British Columbia, Canada
| | - François Bénard
- Department of Molecular Oncology, BC Cancer Research Centre, Vancouver, British Columbia, Canada
- Department of Radiology, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
122
|
Praena J, Garcia-Infantes F, Rivera R, Fernandez-Maza L, Arias de Saavedra F, Porras I. Radioisotope production at the IFMIF-DONES facility. EPJ WEB OF CONFERENCES 2020. [DOI: 10.1051/epjconf/202023923001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The International Fusion Materials Irradiation Facility - Demo Oriented NEutron Source (IFMIF-DONES) is a single-sited novel Research Infrastructure for testing, validation and qualification of the materials to be used in a fusion reactor. Recently, IFMIF-DONES has been declared of interest by ESFRI (European Strategy Forum on Research Infrastructures) and its European host city would be Granada (Spain). In spite the first and most important application of IFMIF-DONES related to fusion technology, the unprecedented neutron flux available could be exploited without modifying the routine operation of IFMIF-DONES. Thus, it is already planned an experimental hall for a complementary program with neutrons. Also, a complementary program on the use of the deuteron beam could help IFMIF-DONES to be more sustainable. In the present work, we study radioisotope production with deuterons of 177Lu. The results show the viability of IFMIF-DONES for such production in terms of the needs of a territory of small-medium size. Also the study suggests that new nuclear data at higher deuteron energies are mandatory for an accurate study in this field.
Collapse
|
123
|
Ermert J, Benešová M, Hugenberg V, Gupta V, Spahn I, Pietzsch HJ, Liolios C, Kopka K. Radiopharmaceutical Sciences. Clin Nucl Med 2020. [DOI: 10.1007/978-3-030-39457-8_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
124
|
Large-scale production of lutetium-177m for the 177mLu/ 177Lu radionuclide generator. Appl Radiat Isot 2019; 156:108986. [PMID: 31786419 DOI: 10.1016/j.apradiso.2019.108986] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 10/07/2019] [Accepted: 11/11/2019] [Indexed: 11/22/2022]
Abstract
In this work, 177mLu has been produced by irradiation of natural Lu2O3 targets at the BR2 reactor (Mol, Belgium) and the obtained data together with literature values have been used to theoretically investigate the production of 177mLu at different neutron fluxes, irradiation times and enrichment of 176Lu. The irradiation time (tmax) needed to reach the maximum 177mLu production has been found to change from 42, 12, 4 days with the increase in the thermal neutron flux from 2*1014, 8*1014, 2.5*1015 n cm-2 s-1, respectively while keeping the maximum 177mLu activity unaffected. The results of our calculations suggest that 0.11 TBq 177mLu with a specific activity of 0.3 TBq g-1 Lu can be produced in a short irradiation time of 4 days using 1g of 84.44% 176Lu enriched Lu2O3 and a thermal neutron flux of 2.5*1015 n cm-2 s-1.
Collapse
|
125
|
Preparation of [ 177Lu]Lu-DOTA-Ahx-Lys40-Exendin-4 for radiotherapy of insulinoma: a detailed insight into the radiochemical intricacies. Nucl Med Biol 2019; 78-79:31-40. [PMID: 31731177 DOI: 10.1016/j.nucmedbio.2019.11.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 10/11/2019] [Accepted: 11/07/2019] [Indexed: 01/05/2023]
Abstract
INTRODUCTION [177Lu]Lu-DOTA-Ahx-Lys40-Exendin-4 ([177Lu]Lu-DOTA-Exendin-4) is a potential agent for radiotherapy of insulinomas owing to its specificity towards GLP-1 (Glucagon like peptide-1) receptors over-expressed on such cancers. The objective of the present study is to optimize the various radiochemistry parameters for the consistent formulation of the agent with high radiolabeling yield using carrier added [177Lu]LuCl3 and also to evaluate its biological behaviour in small animal model. METHODS In order to optimize the radiolabeling parameters, DOTA-Exendin-4 was radiolabeled with [177Lu]LuCl3 in two different buffer systems (sodium acetate and HEPES) at three different temperatures (45, 65 and 95 °C) using three different ligand to metal ratios (3:1, 4:1 and 5:1). The radiolabeled peptide was characterized by both paper chromatography and HPLC. The effect of addition of three different radio-protectors on complexation yield was also studied. Bio-distribution studies were carried out in healthy Swiss mice to evaluate the pharmacokinetic behaviour of the radiolabeled peptide as well as to determine the in vivo specificity of the radiotracer towards GLP-1 receptors (blocking studies). Urine and kidney lysate of the animals were analyzed at various post-administration time-points in order to determine the in vivo stability of the radiolabeled peptide. RESULTS The [177Lu]Lu-DOTA-Exendin-4 complex could be prepared consistently with >95% radiolabeling yield using the optimized reaction conditions. Bio-distribution studies revealed early accumulation of [177Lu]Lu-DOTA-Exendin-4 in pancreas along with fast clearance via renal pathway. Significantly high accumulation of the radiotracer was observed in kidneys. Analyses of urine and kidney lysate of the animals revealed in vivo stability of [177Lu]Lu-DOTA-Exendin-4. Blocking studies showed displacement of significant amount of radiotracer from GLP-1 receptor-positive organs such as, pancreas and lungs (p <0.05) in presence of unlabeled peptide, indicating the specificity of the radiolabeled preparation towards GLP-1 receptors. CONCLUSIONS Present study shows that [177Lu]Lu-DOTA-Exendin-4 could be formulated for radiotherapeutic application with high radiochemical purity and adequate in vivo stability using [177Lu]LuCl3 produced via direct neutron irradiation. ADVANCES IN KNOWLEDGE AND IMPLICATIONS FOR PATIENT CARE Findings of the present study will be helpful in preparing the patient dose of [177Lu]Lu-labeled Exendin for radiotherapy of insulinoma using carrier added [177Lu]LuCl3, produced in a medium flux reactor, without the requirement of post-labeling purification.
Collapse
|
126
|
Yoshii Y, Matsumoto H, Yoshimoto M, Oe Y, Zhang MR, Nagatsu K, Sugyo A, Tsuji AB, Higashi T. 64Cu-Intraperitoneal Radioimmunotherapy: A Novel Approach for Adjuvant Treatment in a Clinically Relevant Preclinical Model of Pancreatic Cancer. J Nucl Med 2019; 60:1437-1443. [PMID: 30850497 PMCID: PMC6785796 DOI: 10.2967/jnumed.118.225045] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Accepted: 02/20/2019] [Indexed: 01/11/2023] Open
Abstract
Pancreatic cancer (PC) has a very poor prognosis. Surgery is the primary treatment for patients with resectable PC; however, local recurrence, hepatic metastasis, and peritoneal dissemination often occur even after extensive surgery. Adjuvant chemotherapy, typically with gemcitabine, has been used clinically but with only a modest survival benefit. To achieve a better outcome, we investigated the efficacy of 64Cu-intraperitoneal radioimmunotherapy (ipRIT) with 64Cu-labeled antiepidermal growth factor receptor antibody cetuximab as an adjuvant treatment after PC surgery using an orthotopic xenografted mouse model. Methods: The efficacy of adjuvant 64Cu-ipRIT was investigated in a human PC mouse model harboring orthotopic xenografts of xPA-1-DC cells. To reproduce the clinical situation, PC xenografts were surgically resected when pancreatic tumors were readily visible but not metastatic tumors. Increasing doses of 64Cu-cetuximab were intraperitoneally injected, and the mice were monitored for toxicity to determine the safe therapeutic dose. For adjuvant 64Cu-ipRIT, the day after tumor resection, the mice were intraperitoneally administered 22.2 MBq of 64Cu-PCTA-cetuximab and the survival was compared with that in surgery-only controls. For comparison, adjuvant chemotherapy with gemcitabine was also examined using the same model. Results: The mouse model not only developed primary tumors in the pancreas but also subsequently reproduced local recurrence, hepatic metastasis, and peritoneal dissemination after surgery, which is similar to the manifestations that occur with human PC. Adjuvant 64Cu-ipRIT with 64Cu-labeled cetuximab after surgery effectively suppressed local recurrence, hepatic metastasis, and peritoneal dissemination in this model. Significant improvement of the survival with minimal toxicity was achieved by adjuvant 64Cu-ipRIT compared with that in control mice that underwent surgery only. Adjuvant chemotherapy with gemcitabine nominally prolonged the survival, but the effect was not statistically significant. Conclusion:64Cu-ipRIT with cetuximab can be an effective adjuvant therapy after PC surgery.
Collapse
Affiliation(s)
- Yukie Yoshii
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | | | - Mitsuyoshi Yoshimoto
- Division of Functional Imaging, National Cancer Center Hospital East, Chiba, Japan
| | - Yoko Oe
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Ming-Rong Zhang
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Kotaro Nagatsu
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Aya Sugyo
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Atsushi B Tsuji
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Tatsuya Higashi
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| |
Collapse
|
127
|
Kuznetsov RA, Bobrovskaya KS, Svetukhin VV, Fomin AN, Zhukov AV. Production of Lutetium-177: Process Aspects. RADIOCHEMISTRY 2019. [DOI: 10.1134/s1066362219040015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
128
|
Mikolajczak R, van der Meulen NP, Lapi SE. Radiometals for imaging and theranostics, current production, and future perspectives. J Labelled Comp Radiopharm 2019; 62:615-634. [PMID: 31137083 DOI: 10.1002/jlcr.3770] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 04/10/2019] [Accepted: 05/15/2019] [Indexed: 02/06/2023]
Abstract
The aim of this review is to make the reader familiar with currently available radiometals, their production modes, capacities, and quality concerns related to their medical use, as well as new emerging radiometals and irradiation technologies from the perspective of their diagnostic and theranostic applications. Production methods of 177 Lu serve as an example of various issues related to the production yield, specific activity, radionuclidic and chemical purity, and production economy. Other radiometals that are currently used or explored for potential medical applications, with particular focus on their theranostic value, are discussed. Using radiometals for diagnostic imaging and therapy is on the rise. The high demand for radiometals for medical use prompts investigations towards using alternative irradiation reactions, while using existing nuclear reactors and accelerator facilities. This review discusses these production capacities and what is necessary to cover the growing demand for theranostic nuclides.
Collapse
Affiliation(s)
- Renata Mikolajczak
- Radioisotope Centre POLATOM, National Centre for Nuclear Research, Otwock, Poland
| | | | - Suzanne E Lapi
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
129
|
Song IH, Jeong MS, Hong HJ, Shin JI, Park YS, Woo SK, Moon BS, Kim KI, Lee YJ, Kang JH, Lee TS. Development of a Theranostic Convergence Bioradiopharmaceutical for Immuno-PET Based Radioimmunotherapy of L1CAM in Cholangiocarcinoma Model. Clin Cancer Res 2019; 25:6148-6159. [PMID: 31337646 DOI: 10.1158/1078-0432.ccr-19-1157] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 05/31/2019] [Accepted: 07/18/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE Cholangiocarcinoma is a malignancy of bile duct with a poor prognosis. Conventional chemotherapy and radiotherapy are generally ineffective, and surgical resection is the only curative treatment for cholangiocarcinoma. L1-cell adhesion molecule (L1CAM) has been known as a novel prognostic marker and therapeutic target for cholangiocarcinoma. This study aimed to evaluate the feasibility of immuno-PET imaging-based radioimmunotherapy using radiolabeled anti-L1CAM antibody in cholangiocarcinoma xenograft model. EXPERIMENTAL DESIGN We prepared a theranostic convergence bioradiopharmaceutical using chimeric anti-L1CAM antibody (cA10-A3) conjugated with 1,4,7-triazacyclononane-1,4,7-triacetic acid (NOTA) chelator and labeled with 64Cu or 177Lu and evaluated the immuno-PET or SPECT/CT imaging and biodistribution with 64Cu-/177Lu-cA10-A3 in various cholangiocarcinoma xenograft models. Therapeutic efficacy and response monitoring were performed by 177Lu-cA10-A3 and 18F-FDG-PET, respectively, and immunohistochemistry was done by TUNEL and Ki-67. RESULTS Radiolabeled cA10-A3 antibodies specifically recognized L1CAM in vitro, clearly visualized cholangiocarcinoma tumors in immuno-PET and SPECT/CT imaging, and differentiated the L1CAM expression level in cholangiocarcinoma xenograft models. 177Lu-cA10-A3 (12.95 MBq/100 μg) showed statistically significant reduction in tumor volumes (P < 0.05) and decreased glucose metabolism (P < 0.01). IHC analysis revealed 177Lu-cA10-A3 treatment increased TUNEL-positive and decreased Ki-67-positive cells, compared with saline, cA10-A3, or 177Lu-isotype. CONCLUSIONS Anti-L1CAM immuno-PET imaging using 64Cu-cA10-A3 could be translated into the clinic for characterizing the pharmacokinetics and selecting appropriate patients for radioimmunotherapy. Radioimmunotherapy using 177Lu-cA10-A3 may provide survival benefit in L1CAM-expressing cholangiocarcinoma tumor. Theranostic convergence bioradiopharmaceutical strategy would be applied as imaging biomarker-based personalized medicine in L1CAM-expressing patients with cholangiocarcinoma.
Collapse
Affiliation(s)
- In Ho Song
- Division of RI Application, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea.,Department of Biomedical Laboratory Science, Yonsei University, Wonju, South Korea
| | - Mun Sik Jeong
- Department of Systems Immunology, Kangwon National University, Chuncheon, South Korea
| | - Hyo Jeong Hong
- Department of Systems Immunology, Kangwon National University, Chuncheon, South Korea.,Scripps Korea Antibody Institute, Chuncheon, South Korea
| | - Jong Il Shin
- Division of RI Application, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Yong Serk Park
- Department of Biomedical Laboratory Science, Yonsei University, Wonju, South Korea
| | - Sang-Keun Woo
- Division of RI Application, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Byung Seok Moon
- Department of Nuclear Medicine, Ewha Womans University Seoul Hospital, Ewha Womans University School of Medicine, Seoul, South Korea
| | - Kwang Il Kim
- Division of RI Application, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Yong Jin Lee
- Division of RI Application, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Joo Hyun Kang
- Division of RI Application, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Tae Sup Lee
- Division of RI Application, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea.
| |
Collapse
|
130
|
Gracheva N, Müller C, Talip Z, Heinitz S, Köster U, Zeevaart JR, Vögele A, Schibli R, van der Meulen NP. Production and characterization of no-carrier-added 161Tb as an alternative to the clinically-applied 177Lu for radionuclide therapy. EJNMMI Radiopharm Chem 2019; 4:12. [PMID: 31659528 PMCID: PMC6620226 DOI: 10.1186/s41181-019-0063-6] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 06/11/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND 161Tb is an interesting radionuclide for cancer treatment, showing similar decay characteristics and chemical behavior to clinically-employed 177Lu. The therapeutic effect of 161Tb, however, may be enhanced due to the co-emission of a larger number of conversion and Auger electrons as compared to 177Lu. The aim of this study was to produce 161Tb from enriched 160Gd targets in quantity and quality sufficient for first application in patients. METHODS No-carrier-added 161Tb was produced by neutron irradiation of enriched 160Gd targets at nuclear research reactors. The 161Tb purification method was developed with the use of cation exchange (Sykam resin) and extraction chromatography (LN3 resin), respectively. The resultant product (161TbCl3) was characterized and the 161Tb purity compared with commercial 177LuCl3. The purity of the final product (161TbCl3) was analyzed by means of γ-ray spectrometry (radionuclidic purity) and radio TLC (radiochemical purity). The radiolabeling yield of 161Tb-DOTA was assessed over a two-week period post processing in order to observe the quality change of the obtained 161Tb towards future clinical application. To understand how the possible drug products (peptides radiolabeled with 161Tb) vary with time, stability of the clinically-applied somatostatin analogue DOTATOC, radiolabeled with 161Tb, was investigated over a 24-h period. The radiolytic stability experiments were compared to those performed with 177Lu-DOTATOC in order to investigate the possible influence of conversion and Auger electrons of 161Tb on peptide disintegration. RESULTS Irradiations of enriched 160Gd targets yielded 6-20 GBq 161Tb. The final product was obtained at an activity concentration of 11-21 MBq/μL with ≥99% radionuclidic and radiochemical purity. The DOTA chelator was radiolabeled with 161Tb or 177Lu at the molar activity deemed useful for clinical application, even at the two-week time point after end of chemical separation. DOTATOC, radiolabeled with either 161Tb or 177Lu, was stable over 24 h in the presence of a stabilizer. CONCLUSIONS In this study, it was shown that 161Tb can be produced in high activities using different irradiation facilities. The developed method for 161Tb separation from the target material yielded 161TbCl3 in quality suitable for high-specific radiolabeling, relevant for future clinical application.
Collapse
Affiliation(s)
- Nadezda Gracheva
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232 Villigen-PSI, Switzerland
| | - Cristina Müller
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232 Villigen-PSI, Switzerland
| | - Zeynep Talip
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232 Villigen-PSI, Switzerland
| | - Stephan Heinitz
- Laboratory of Radiochemistry, Paul Scherrer Institute, 5232 Villigen-PSI, Switzerland
| | - Ulli Köster
- Institut Laue-Langevin, 38042 Grenoble, France
| | - Jan Rijn Zeevaart
- Radiochemistry, South African Nuclear Energy Corporation (Necsa), Brits, 0242 South Africa
| | - Alexander Vögele
- Laboratory of Radiochemistry, Paul Scherrer Institute, 5232 Villigen-PSI, Switzerland
| | - Roger Schibli
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232 Villigen-PSI, Switzerland
- Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Nicholas P. van der Meulen
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, 5232 Villigen-PSI, Switzerland
- Laboratory of Radiochemistry, Paul Scherrer Institute, 5232 Villigen-PSI, Switzerland
| |
Collapse
|
131
|
Pandey U, Subramanian S, Shaikh S, Gamre N, Kumar S, Dash A. Synthesis and Preliminary Biological Evaluation of 177Lu-Labeled Polyhydroxamic Acid Microparticles Toward Therapy of Hepatocellular Carcinoma. Cancer Biother Radiopharm 2019; 34:306-315. [PMID: 31188652 DOI: 10.1089/cbr.2018.2747] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background: Transarterial radioembolization (TARE) represents an effective targeted therapeutic option for hepatocellular carcinoma (HCC), a cancer with high mortality and poor prognosis. The aim of this study was the preparation and preliminary biological evaluation of 177Lu-labeled polyhydroxamic acid (PHA) microparticles toward possible use in the therapy of HCC. Materials and Methods: PHA microparticles were synthesized starting from polyacrylamide. They were characterized by Fourier-transform infrared spectroscopy (FT-IR), visual color test, and laser diffraction particle size analysis. Experimental variables such as reaction pH, amount of PHA microparticles, carrier Lu content, and incubation time were optimized for maximum uptake of 177Lu on PHA microparticles. Stability of 177Lu-PHA microparticles was tested in the presence of competing Fe(III) ions in solution. In vitro stability of 177Lu-PHA microparticles was evaluated in 0.05 M sodium phosphate solution (pH 7.5), saline, and serum. Bioevaluation studies were performed in normal Wistar rats by intrahepatic artery injection of the 177Lu-PHA microparticles. Results: Successful synthesis of PHA microparticles could be confirmed from the results of FT-IR analysis and visual color test. Laser diffraction-based particle size analysis confirmed median particle size to be 54 μm, suitable for TARE. Under the optimized conditions, >99% loading of 177Lu on PHA microparticles could be achieved. Even in the presence of high concentration of Fe(III) ions, 177Lu binding to PHA microparticles was stable. 177Lu-PHA microparticles exhibited excellent in vitro stability in sodium phosphate solution, saline, and serum up to 5 d at 37°C. In the bioevaluation studies performed in normal Wistar rats, 92.8% ± 3.1% of 177Lu-PHA microparticles were retained in the liver at 96 h postinjection without any significant leakage to other organs. Conclusion: This preliminary study demonstrates the potential of synthesized PHA microparticles as carriers of therapeutic radioisotopes such as 177Lu for treatment of HCC.
Collapse
Affiliation(s)
- Usha Pandey
- 1 Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Mumbai, India.,2 Homi Bhabha National Institute, Mumbai, India
| | - Suresh Subramanian
- 1 Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Mumbai, India.,2 Homi Bhabha National Institute, Mumbai, India
| | - Samina Shaikh
- 2 Homi Bhabha National Institute, Mumbai, India.,3 Analytical Chemistry Division, Bhabha Atomic Research Centre, Mumbai, India
| | - Naresh Gamre
- 1 Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Mumbai, India
| | - Sanjukta Kumar
- 2 Homi Bhabha National Institute, Mumbai, India.,3 Analytical Chemistry Division, Bhabha Atomic Research Centre, Mumbai, India
| | - Ashutosh Dash
- 1 Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Mumbai, India.,2 Homi Bhabha National Institute, Mumbai, India
| |
Collapse
|
132
|
Jeon J. Review of Therapeutic Applications of Radiolabeled Functional Nanomaterials. Int J Mol Sci 2019; 20:E2323. [PMID: 31083402 PMCID: PMC6539387 DOI: 10.3390/ijms20092323] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/08/2019] [Accepted: 05/09/2019] [Indexed: 01/10/2023] Open
Abstract
In the last two decades, various nanomaterials have attracted increasing attention in medical science owing to their unique physical and chemical characteristics. Incorporating radionuclides into conventionally used nanomaterials can confer useful additional properties compared to the original material. Therefore, various radionuclides have been used to synthesize functional nanomaterials for biomedical applications. In particular, several α- or β-emitter-labeled organic and inorganic nanoparticles have been extensively investigated for efficient and targeted cancer treatment. This article reviews recent progress in cancer therapy using radiolabeled nanomaterials including inorganic, polymeric, and carbon-based materials and liposomes. We first provide an overview of radiolabeling methods for preparing anticancer agents that have been investigated recently in preclinical studies. Next, we discuss the therapeutic applications and effectiveness of α- or β-emitter-incorporated nanomaterials in animal models and the emerging possibilities of these nanomaterials in cancer therapy.
Collapse
Affiliation(s)
- Jongho Jeon
- Department of Applied Chemistry, School of Applied Chemical Engineering, Kyungpook National University, Daegu 41566, Korea.
| |
Collapse
|
133
|
Preparation of 177Lu-Trastuzumab injection for treatment of breast cancer. Appl Radiat Isot 2019; 148:184-190. [PMID: 30974402 DOI: 10.1016/j.apradiso.2019.04.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 03/28/2019] [Accepted: 04/02/2019] [Indexed: 11/21/2022]
Abstract
The objective of this study was the facile preparation of 177Lu-CHX-A''-DTPA-Trastuzumab injection for breast cancer therapy. Trastuzumab conjugated with CHX-A''-DTPA-NCS was radiolabeled with 177Lu in >95% radiochemical purity. In vitro studies in SKBR3 and MDA-MB-453 cells confirmed specificity of 177Lu-CHX-A''-DTPA-Trastuzumab to HER2 positive cells. The radioimmunoconjugate showed good immunoreactivity, in vitro stability in saline and Kd of 1.01 ± 0.13 nM in SKBR3 cells. Clearance of 177Lu-CHX-A''-DTPA-Trastuzumab in Swiss mice was predominantly through the hepatobiliary route with minimal bone uptake.
Collapse
|
134
|
Mishiro K, Hanaoka H, Yamaguchi A, Ogawa K. Radiotheranostics with radiolanthanides: Design, development strategies, and medical applications. Coord Chem Rev 2019. [DOI: 10.1016/j.ccr.2018.12.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
135
|
Radiochemical processing of nuclear-reactor-produced radiolanthanides for medical applications. Coord Chem Rev 2019. [DOI: 10.1016/j.ccr.2018.11.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
136
|
Pandey U, Kameswaran M, Gamre N, Dash A. Preparation of 177 Lu-labeled Nimotuzumab for radioimmunotherapy of EGFR-positive cancers: Comparison of DOTA and CHX-A″-DTPA as bifunctional chelators. J Labelled Comp Radiopharm 2019; 62:158-165. [PMID: 30663095 DOI: 10.1002/jlcr.3707] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 01/11/2019] [Indexed: 11/05/2022]
Abstract
This study was aimed at evaluating the role of bifunctional chelators DOTA-NCS and CHX-A″-DTPA-NCS used for conjugating 177 Lu with Nimotuzumab on the radiochemical yields, purity, in vitro stability, and specificity of the radioimmunoconjugates to EGFR. Two immunoconjugates were prepared wherein Nimotuzumab was conjugated with the acyclic ligand p-NCS-Bn-CHX-A″-DTPA and macrocyclic ligand p-NCS-Bn-DOTA. These were radiolabeled with 177 Lu, purified on PD-10 column, and characterized by SE-HPLC. In vitro stability was determined up to 4 days post preparation. Specificity of the radioimmunoconjugates was ascertained by in vitro studies in A431 cells while the biodistribution patterns were studied in normal Swiss mice up to 96 hours post injection. Four to five molecules of CHX-A″-DTPA/DOTA were attached to one molecule of Nimotuzumab. Radiochemical purity of both 177 Lu-CHX-A″-DTPA-Nimotuzumab and 177 Lu-DOTA-Nimotuzumab was determined to be greater than 98%. Both the radioimmunoconjugates exhibited good in vitro stability at 37°C up to 4 days post preparation in saline, and their clearance was largely by the hepatobiliary route. The DOTA- and CHX-A″-DTPA-based radioimmunoconjugates could be prepared with good radiochemical purity, in vitro stability, and specificity to EGFR. Further studies in EGFR-positive cancers would pave way for them for use in the clinics.
Collapse
Affiliation(s)
- Usha Pandey
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Mumbai, India.,Bhabha Atomic Research Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Mythili Kameswaran
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Mumbai, India
| | - Naresh Gamre
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Mumbai, India
| | - Ashutosh Dash
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Mumbai, India.,Bhabha Atomic Research Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| |
Collapse
|
137
|
Lau J, Jacobson O, Niu G, Lin KS, Bénard F, Chen X. Bench to Bedside: Albumin Binders for Improved Cancer Radioligand Therapies. Bioconjug Chem 2019; 30:487-502. [PMID: 30616340 DOI: 10.1021/acs.bioconjchem.8b00919] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Radioligand therapy (RLT) relies on the use of pharmacophores to selectively deliver ionization energy to cancers to exert its tumoricidal effects. Cancer cells that are not directly targeted by a radioconjugate remain susceptible to RLT because of the crossfire effect. This is significant given the inter- and intra-heterogeneity of tumors. In recent years, reversible albumin binders have been used as simple "add-ons" for radiopharmaceuticals to modify pharmacokinetics and to enhance therapeutic efficacy. In this Review, we discuss recent advances in albumin binder platforms used in RLT, with an emphasis on 4-( p-iodophenyl)butyric acid and Evans blue derivatives. We focus on four biological systems pertinent to oncology that utilize this class of compounds: folate receptor, integrin αvβ3, somatostatin receptor, and prostate-specific membrane antigen. Finally, we offer our perspectives on albumin binders for RLT, highlighting future areas of research that will help propel the technology further for clinical use.
Collapse
Affiliation(s)
- Joseph Lau
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB) , National Institutes of Health (NIH) , Bethesda , Maryland 20892 , United States
| | - Orit Jacobson
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB) , National Institutes of Health (NIH) , Bethesda , Maryland 20892 , United States
| | - Gang Niu
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB) , National Institutes of Health (NIH) , Bethesda , Maryland 20892 , United States
| | - Kuo-Shyan Lin
- Department of Molecular Oncology , BC Cancer , Vancouver , British Columbia V5Z 1L3 , Canada
| | - François Bénard
- Department of Molecular Oncology , BC Cancer , Vancouver , British Columbia V5Z 1L3 , Canada
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB) , National Institutes of Health (NIH) , Bethesda , Maryland 20892 , United States
| |
Collapse
|
138
|
Deblonde GJP, Lohrey TD, Abergel RJ. Inducing selectivity and chirality in group IV metal coordination with high-denticity hydroxypyridinones. Dalton Trans 2019; 48:8238-8247. [PMID: 31094380 DOI: 10.1039/c9dt01031a] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The solution- and solid-state interactions between the octadentate siderophore mimic 3,4,3-LI(1,2-HOPO) (343HOPO) and group IV metal ions were investigated using high-resolution mass spectrometry, liquid chromatography, UV-visible spectrophotometry, metal-competition batch titrations, and single crystal X-ray diffraction. 343HOPO forms a neutral 1 : 1 complex, [HfIV343HOPO], that exhibits extreme stability in aqueous solution, with a log β110 value reaching 42.3. These results affirm the remarkable charge-based selectivity of 343HOPO for octacoordinated tetravalent cations with a Hf(iv) complex 1021 more stable than its Lu(iii) analogue. Moreover, [HfIV343HOPO] and its Zr(iv) counterpart show exceptional robustness, with the ligand remaining bound to the cation over a very broad pH range: from pH ∼ 11 to acidic conditions as strong as 10 M HCl. In stark contrast, Ti(iv)-343HOPO species are far less stable and undergo hydrolysis at pH as low as ∼6, likely due to the mismatch between the preferred hexacoordinated Ti(iv) ion and octadentate 343HOPO ligand. The extreme charge-based and denticity-driven selectivity of 343HOPO, now observed across the periodic table, paves the way for new selective sequestration systems for radionuclides including medical 44Ti, 89Zr or 177Lu/Hf isotopes, toxic polonium (Po) contaminants, as well as rutherfordium (Rf) research isotopes. Furthermore, despite the lack of a chiral center in 343HOPO, its complexes with metal ions are chiral and appear to form a single set of enantiomers.
Collapse
Affiliation(s)
- Gauthier J-P Deblonde
- Chemical Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | | | | |
Collapse
|
139
|
Sahoo S, Shrivastava V, Selvam TP, Bakshi AK, Kumar R, Rama P, Datta D, Chinnaesakki S, Saxena SK, Kumar Y, Dash A. Dosimetry of indigenously developed 177Lu patch source for surface brachytherapy-Experimental and Monte Carlo methods. JOURNAL OF RADIOLOGICAL PROTECTION : OFFICIAL JOURNAL OF THE SOCIETY FOR RADIOLOGICAL PROTECTION 2019; 39:54-70. [PMID: 30523912 DOI: 10.1088/1361-6498/aaeeb6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
This paper describes the evaluation of dosimetry characteristics of an in-house developed 177Lu skin patch source for treatment of non-melanoma skin cancer. A 177Lu skin patch source based on Nafion-115 membrane backbone containing 3.46 ± 0.01 mCi of activity was used. Activity measurement of the patch source was based on gamma ray spectrometry using a HPGe detector. The efficiencies of the HPGe detector were fitted using an orthogonal polynomial function. The absorbed dose rate to water at 5 μm depth in water was determined using an extrapolation chamber, EBT3 Gafchromic film and compared with Monte Carlo methods. The correction factors such as Bragg-Gray stopping power ratio of water-to-air and chamber wall material being different from water, needed to be applied on measurements for establishing the dose rate at 5 μm depth, were calculated using the Monte Carlo method. Absorbed dose rate at 5 μm depth in water (surface dose rate) measured using an extrapolation chamber and EBT3 Gafchromic film were 9.9 ± 0.7 and 8.2 ± 0.1 Gy h-1 mCi-1 respectively for the source activity of 3.46 ± 0.01 mCi. The surface dose rate calculated using the Monte Carlo method was 8.7 ± 0.2 Gy h-1 mCi-1, which agrees reasonably well with measurement. The measured dose rate per mCi offers scope for ascertaining treatment time required to deliver the dose for propitious therapeutic outcome. Additionally, on-axis depth dose and lateral dose profiles at 5 μm and 1 mm depth in water phantom were also calculated using the Monte Carlo method.
Collapse
Affiliation(s)
- Sridhar Sahoo
- Radiological Physics & Advisory Division, Bhabha Atomic Research Centre, Mumbai-400 085, India. Homi Bhabha National Institute, Mumbai-400 094, India
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
140
|
Kelly JM, Amor-Coarasa A, Ponnala S, Nikolopoulou A, Williams C, DiMagno SG, Babich JW. Albumin-Binding PSMA Ligands: Implications for Expanding the Therapeutic Window. J Nucl Med 2018; 60:656-663. [PMID: 30552199 DOI: 10.2967/jnumed.118.221150] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 11/30/2018] [Indexed: 11/16/2022] Open
Abstract
Despite significant gains in the treatment of metastatic castration-resistant prostate cancer by radioligands targeting prostate-specific membrane antigen (PSMA), 30% of patients never respond to therapy. One possible explanation is insufficient dose delivery to the tumor because of suboptimal pharmacokinetics. We have recently described RPS-063, a trifunctional ligand targeting PSMA with high uptake in LNCaP xenograft tumors but also in kidneys. We aimed to use structural modifications to increase the tumor-to-kidney ratio through increased albumin binding and tumor uptake and reduction of kidney activity. Methods: Four structurally related trifunctional PSMA-targeting small molecules were prepared by either varying the albumin-binding group or inserting a polyethylene glycol 8 linker into a common structure. The compounds were ranked by PSMA affinity and albumin affinity and were radiolabeled with 68Ga and 177Lu. Tissue kinetics were determined in male BALB/C nu/nu mice bearing LNCaP xenograft tumors. Results: Each of the compounds binds PSMA with a half-maximal inhibitory concentration of no more than 10 nM. The albumin-binding group had a minimal effect on PSMA affinity but changed albumin affinity by an order of magnitude. However, the addition of a polyethylene glycol 8 spacer weakened affinity for albumin in each case. Increased affinity for albumin corresponded with delayed blood clearance and modified uptake kinetics in the tumor and kidney. Uptake of 177Lu-RPS-072 (34.9 ± 2.4 %ID/g) and 177Lu-RPS-077 (27.4 ± 0.6 %ID/g) increased up to 24 h after injection, and washout by 96 h was not significant. As a result, the area under the curve (AUC) in the tumor was in the following order: 177Lu-RPS-072 > 177Lu-RPS-077 > 177Lu-RPS-063 > 177Lu-RPS-071. Increased linker length corresponded to more rapid clearance from kidneys. Consequently, the ratio of tumor AUC and kidney AUC was 4.7 ± 0.3 for 177Lu-RPS-072. Conclusion: The tumor AUC and tumor-to-kidney ratio of 177Lu-RPS-072 are significantly enhanced compared with any small molecule investigated in a LNCaP xenograft model to date. In comparison to other PSMA-targeting radioligands that have been evaluated in a PC3-PIP model, activity in kidneys is reduced and activity in tumors compares favorably when the different PSMA expression levels in LNCaP and PC3-PIP cells are considered. RPS-072 therefore exhibits an increased therapeutic index, shows the potential to increase the dose delivered to tumors, and is a highly promising candidate for targeted radioligand therapy.
Collapse
Affiliation(s)
- James M Kelly
- Molecular Imaging Innovations Institute (MI3), Department of Radiology, Weill Cornell Medicine, New York, New York.,Division of Radiopharmaceutical Science, Department of Radiology, Weill Cornell Medicine, New York, New York
| | - Alejandro Amor-Coarasa
- Molecular Imaging Innovations Institute (MI3), Department of Radiology, Weill Cornell Medicine, New York, New York.,Division of Radiopharmaceutical Science, Department of Radiology, Weill Cornell Medicine, New York, New York
| | - Shashikanth Ponnala
- Molecular Imaging Innovations Institute (MI3), Department of Radiology, Weill Cornell Medicine, New York, New York.,Division of Radiopharmaceutical Science, Department of Radiology, Weill Cornell Medicine, New York, New York
| | - Anastasia Nikolopoulou
- Division of Radiopharmaceutical Science, Department of Radiology, Weill Cornell Medicine, New York, New York.,Citigroup Biomedical Imaging Center, Weill Cornell Medicine, New York, New York
| | - Clarence Williams
- Molecular Imaging Innovations Institute (MI3), Department of Radiology, Weill Cornell Medicine, New York, New York.,Division of Radiopharmaceutical Science, Department of Radiology, Weill Cornell Medicine, New York, New York
| | - Stephen G DiMagno
- College of Pharmacy, University of Illinois-Chicago, Chicago, Illinois; and
| | - John W Babich
- Molecular Imaging Innovations Institute (MI3), Department of Radiology, Weill Cornell Medicine, New York, New York .,Division of Radiopharmaceutical Science, Department of Radiology, Weill Cornell Medicine, New York, New York.,Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, New York
| |
Collapse
|
141
|
Basaco T, Pektor S, Bermudez JM, Meneses N, Heller M, Galván JA, Boligán KF, Schürch S, von Gunten S, Türler A, Miederer M. Evaluation of Radiolabeled Girentuximab In Vitro and In Vivo. Pharmaceuticals (Basel) 2018; 11:E132. [PMID: 30487460 PMCID: PMC6316122 DOI: 10.3390/ph11040132] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 11/21/2018] [Accepted: 11/26/2018] [Indexed: 11/16/2022] Open
Abstract
Girentuximab (cG250) targets carbonic anhydrase IX (CAIX), a protein which is expressed on the surface of most renal cancer cells (RCCs). cG250 labeled with 177Lu has been used in clinical trials for radioimmunotherapy (RIT) of RCCs. In this work, an extensive characterization of the immunoconjugates allowed optimization of the labeling conditions with 177Lu while maintaining immunoreactivity of cG250, which was then investigated in in vitro and in vivo experiments. cG250 was conjugated with S-2-(4-isothiocyanatobenzyl)-1,4,7,10-tetraazacyclododecane tetraacetic acid (DOTA(SCN)) by using incubation times between 30 and 90 min and characterized by mass spectrometry. Immunoconjugates with five to ten DOTA(SCN) molecules per cG250 molecule were obtained. Conjugates with ratios less than six DOTA(SCN)/cG250 had higher in vitro antigen affinity, both pre- and postlabeling with 177Lu. Radiochemical stability increased, in the presence of sodium ascorbate, which prevents radiolysis. The immunoreactivity of the radiolabeled cG250 tested by specific binding to SK-RC-52 cells decreased when the DOTA content per conjugate increased. The in vivo tumor uptake was < 10% ID/g and independent of the total amount of protein in the range between 5 and 100 µg cG250 per animal. Low tumor uptake was found to be due to significant necrotic areas and heterogeneous CAIX expression. In addition, low vascularity indicated relatively poor accessibility of the CAIX target.
Collapse
Affiliation(s)
- Tais Basaco
- Department of Chemistry and Biochemistry, University of Bern, 3012 Bern, Switzerland.
- Laboratory of Radiochemistry, Paul Scherrer Institute (PSI), 5232 Villigen PSI, Switzerland.
| | - Stefanie Pektor
- Clinic for Nuclear Medicine, University Medical Center Mainz, 55131 Mainz, Germany.
| | - Josue M Bermudez
- Department of Chemistry and Biochemistry, University of Bern, 3012 Bern, Switzerland.
| | - Niurka Meneses
- Department of Chemistry and Biochemistry, University of Bern, 3012 Bern, Switzerland.
| | - Manfred Heller
- Department for Biomedical Research (DBMR), University of Bern, 3010 Bern, Switzerland.
| | - José A Galván
- Institute of Pathology, University of Bern, 3010 Bern, Switzerland.
| | - Kayluz F Boligán
- Institute of Pharmacology (PKI), University of Bern, 3010 Bern, Switzerland.
| | - Stefan Schürch
- Department of Chemistry and Biochemistry, University of Bern, 3012 Bern, Switzerland.
| | - Stephan von Gunten
- Institute of Pharmacology (PKI), University of Bern, 3010 Bern, Switzerland.
| | - Andreas Türler
- Department of Chemistry and Biochemistry, University of Bern, 3012 Bern, Switzerland.
| | - Matthias Miederer
- Clinic for Nuclear Medicine, University Medical Center Mainz, 55131 Mainz, Germany.
| |
Collapse
|
142
|
Kameswaran M, Pandey U, Gamre N, Shinto A, Subramanian S, Sarma HD, Kamleshwaran KK, Dash A. Ready-to-use 177Lu-Rituximab injection for Non-Hodgkin’s Lymphoma: Formulation and preliminary clinical study. J Radioanal Nucl Chem 2018. [DOI: 10.1007/s10967-018-6042-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
143
|
Abstract
Radiometals possess an exceptional breadth of decay properties and have been applied to medicine with great success for several decades. The majority of current clinical use involves diagnostic procedures, which use either positron-emission tomography (PET) or single-photon imaging to detect anatomic abnormalities that are difficult to visualize using conventional imaging techniques (e.g., MRI and X-ray). The potential of therapeutic radiometals has more recently been realized and relies on ionizing radiation to induce irreversible DNA damage, resulting in cell death. In both cases, radiopharmaceutical development has been largely geared toward the field of oncology; thus, selective tumor targeting is often essential for efficacious drug use. To this end, the rational design of four-component radiopharmaceuticals has become popularized. This Review introduces fundamental concepts of drug design and applications, with particular emphasis on bifunctional chelators (BFCs), which ensure secure consolidation of the radiometal and targeting vector and are integral for optimal drug performance. Also presented are detailed accounts of production, chelation chemistry, and biological use of selected main group and rare earth radiometals.
Collapse
Affiliation(s)
- Thomas I Kostelnik
- Medicinal Inorganic Chemistry Group, Department of Chemistry , University of British Columbia , Vancouver , British Columbia V6T 1Z1 , Canada
| | - Chris Orvig
- Medicinal Inorganic Chemistry Group, Department of Chemistry , University of British Columbia , Vancouver , British Columbia V6T 1Z1 , Canada
| |
Collapse
|
144
|
Park BN, Lee SJ, Roh JH, Lee KH, An YS, Yoon JK. Radiolabeled Anti-Adenosine Triphosphate Synthase Monoclonal Antibody as a Theragnostic Agent Targeting Angiogenesis. Mol Imaging 2018; 16:1536012117737399. [PMID: 29239276 PMCID: PMC5734570 DOI: 10.1177/1536012117737399] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
INTRODUCTION The potential of a radioiodine-labeled, anti-adenosine triphosphate synthase monoclonal antibody (ATPS mAb) as a theragnostic agent for simultaneous cancer imaging and treatment was evaluated. METHODS Adenosine triphosphate synthase monoclonal antibody was labeled with radioiodine, then radiotracer uptake was measured in 6 different cancer cell lines. In vivo biodistribution was evaluated 24 and 48 hours after intravenous injection of 125I-ATPS mAb into MKN-45 tumor-bearing mice (n = 3). For radioimmunotherapy, 18.5 MBq 131I-ATPS mAb (n = 7), isotype immunoglobulin G (IgG) (n = 6), and vehicle (n = 6) were injected into MKN-45 tumor-bearing mice for 4 weeks, and tumor volume and percentage of tumor growth inhibition (TGI) were compared each week. RESULTS MKN-45 cells showed the highest in vitro cellular binding after 4 hours (0.00324 ± 0.00013%/μg), which was significantly inhibited by unlabeled ATPS mAb at concentrations of greater than 0.4 μM. The in vitro retention rate of 125I-ATPS mAb in MKN-45 cells was 64.1% ± 1.0% at 60 minutes. The highest tumor uptake of 125I-ATPS mAb in MKN-45 tumor-bearing mice was achieved 24 hours after injection (6.26% ± 0.47% injected dose [ID]/g), whereas tumor to muscle and tumor to blood ratios peaked at 48 hours. The 24-hour tumor uptake decreased to 3.43% ± 0.85% ID/g by blocking with unlabeled ATPS mAb. After 4 weeks of treatment, mice receiving 131I-ATPS mAb had significantly smaller tumors (679.4 ± 232.3 mm3) compared with control (1687.6 ± 420.4 mm3, P = .0431) and IgG-treated mice (2870.2 ± 484.1 mm3, P = .0010). The percentage of TGI of 131I-ATPS mAb was greater than 50% during the entire study period (range: 53.7%-75.9%). CONCLUSION The specific binding and antitumor effects of radioiodinated ATPS mAb were confirmed in in vitro and in vivo models of stomach cancer.
Collapse
Affiliation(s)
- Bok-Nam Park
- 1 Department of Nuclear Medicine and Molecular Imaging, Ajou University School of Medicine, Suwon, South Korea
| | - Su Jin Lee
- 1 Department of Nuclear Medicine and Molecular Imaging, Ajou University School of Medicine, Suwon, South Korea
| | - Jung Hyun Roh
- 1 Department of Nuclear Medicine and Molecular Imaging, Ajou University School of Medicine, Suwon, South Korea
| | - Kyung-Han Lee
- 2 Department of Nuclear Medicine and Molecular Imaging, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Young-Sil An
- 1 Department of Nuclear Medicine and Molecular Imaging, Ajou University School of Medicine, Suwon, South Korea
| | - Joon-Kee Yoon
- 1 Department of Nuclear Medicine and Molecular Imaging, Ajou University School of Medicine, Suwon, South Korea
| |
Collapse
|
145
|
|
146
|
177Lu-DOTA-coupled minigastrin peptides: promising theranostic agents in neuroendocrine cancers. Mol Biol Rep 2018; 45:1759-1767. [DOI: 10.1007/s11033-018-4319-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 08/16/2018] [Indexed: 02/08/2023]
|
147
|
Wevrett J, Fenwick A, Scuffham J, Johansson L, Gear J, Schlögl S, Segbers M, Sjögreen-Gleisner K, Solný P, Lassmann M, Tipping J, Nisbet A. Inter-comparison of quantitative imaging of lutetium-177 ( 177Lu) in European hospitals. EJNMMI Phys 2018; 5:17. [PMID: 30069805 PMCID: PMC6070453 DOI: 10.1186/s40658-018-0213-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 04/13/2018] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND This inter-comparison exercise was performed to demonstrate the variability of quantitative SPECT/CT imaging for lutetium-177 (177Lu) in current clinical practice. Our aim was to assess the feasibility of using international inter-comparison exercises as a means to ensure consistency between clinical sites whilst enabling the sites to use their own choice of quantitative imaging protocols, specific to their systems. Dual-compartment concentric spherical sources of accurately known activity concentrations were prepared and sent to seven European clinical sites. The site staff were not aware of the true volumes or activity within the sources-they performed SPECT/CT imaging of the source, positioned within a water-filled phantom, using their own choice of parameters and reported their estimate of the activities within the source. RESULTS The volumes reported by the participants for the inner section of the source were all within 29% of the true value and within 60% of the true value for the outer section. The activities reported by the participants for the inner section of the source were all within 20% of the true value, whilst those reported for the outer section were up to 83% different to the true value. CONCLUSIONS A variety of calibration and segmentation methods were used by the participants for this exercise which demonstrated the variability of quantitative imaging across clinical sites. This paper presents a method to assess consistency between sites using different calibration and segmentation methods.
Collapse
Affiliation(s)
- Jill Wevrett
- University of Surrey, Guildford, UK.
- National Physical Laboratory, Teddington, UK.
- Royal Surrey County Hospital NHS Foundation Trust, Guildford, UK.
| | | | - James Scuffham
- University of Surrey, Guildford, UK
- Royal Surrey County Hospital NHS Foundation Trust, Guildford, UK
| | | | | | | | - Marcel Segbers
- Erasmus University Medical Centre, Rotterdam, Netherlands
| | | | - Pavel Solný
- Motol University Hospital, Prague, Czech Republic
| | | | - Jill Tipping
- The Christie NHS Foundation Trust, Manchester, UK
| | - Andrew Nisbet
- University of Surrey, Guildford, UK
- Royal Surrey County Hospital NHS Foundation Trust, Guildford, UK
| |
Collapse
|
148
|
Dadakhanov JA, Lebedev NA, Velichkov AI, Karaivanov DV, Baimukhanova AE, Temerbulatova NT, Filosofov DV. 172Hf → 172Lu Radionuclide Generator Based on a Reverse-Tandem Separation Scheme. RADIOCHEMISTRY 2018. [DOI: 10.1134/s1066362218040112] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
149
|
Yoshii Y, Yoshimoto M, Matsumoto H, Tashima H, Iwao Y, Takuwa H, Yoshida E, Wakizaka H, Yamaya T, Zhang MR, Sugyo A, Hanadate S, Tsuji AB, Higashi T. Integrated treatment using intraperitoneal radioimmunotherapy and positron emission tomography-guided surgery with 64Cu-labeled cetuximab to treat early- and late-phase peritoneal dissemination in human gastrointestinal cancer xenografts. Oncotarget 2018; 9:28935-28950. [PMID: 29989003 PMCID: PMC6034757 DOI: 10.18632/oncotarget.25649] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 06/03/2018] [Indexed: 02/03/2023] Open
Abstract
Peritoneal dissemination is a common cause of death from gastrointestinal cancers and is difficult to treat using current therapeutic options, particularly late-phase disease. Here, we investigated the feasibility of integrated therapy using 64Cu-intraperitoneal radioimmunotherapy (ipRIT), alone or in combination with positron emission tomography (PET)-guided surgery using a theranostic agent (64Cu-labeled anti-epidermal growth factor receptor antibody cetuximab) to treat early- and late-phase peritoneal dissemination in mouse models. In this study, we utilized the OpenPET system, which has open space for conducting surgery while monitoring objects at high resolution in real time, as a novel approach to make PET-guided surgery feasible. 64Cu-ipRIT with cetuximab inhibited tumor growth and prolonged survival with little toxicity in mice with early-phase peritoneal dissemination of small lesions. For late-phase peritoneal dissemination, a combination of 64Cu-ipRIT for down-staging and subsequent OpenPET-guided surgery for resecting large tumor masses effectively prolonged survival. OpenPET clearly detected tumors (≥3 mm in size) behind other organs in the peritoneal cavity and was useful for confirming the presence or absence of residual tumors during an operation. These findings suggest that integrated 64Cu therapy can serve as a novel treatment strategy for peritoneal dissemination.
Collapse
Affiliation(s)
- Yukie Yoshii
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Mitsuyoshi Yoshimoto
- Division of Functional Imaging, National Cancer Center Hospital East, Chiba, Japan
| | | | - Hideaki Tashima
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Yuma Iwao
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Hiroyuki Takuwa
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Eiji Yoshida
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Hidekatsu Wakizaka
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Taiga Yamaya
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Ming-Rong Zhang
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Aya Sugyo
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Sayaka Hanadate
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Atsushi B Tsuji
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Tatsuya Higashi
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| |
Collapse
|
150
|
Aghevlian S, Lu Y, Winnik MA, Hedley DW, Reilly RM. Panitumumab Modified with Metal-Chelating Polymers (MCP) Complexed to 111In and 177Lu-An EGFR-Targeted Theranostic for Pancreatic Cancer. Mol Pharm 2018; 15:1150-1159. [PMID: 29314858 DOI: 10.1021/acs.molpharmaceut.7b01000] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
A metal-chelating polymer (MCP) with a polyglutamide (PGlu) backbone presenting on average 13 DOTA (tetraazacyclododecane-1,4,7,10-tetraacetic acid) chelators for complexing 111In or 177Lu and 10 polyethylene glycol (PEG) chains to minimize liver and spleen uptake was conjugated to antiepidermal growth factor receptor (EGFR) monoclonal antibody (mAb), panitumumab. Because panitumumab-MCP may be dual-labeled with 111In and 177Lu for SPECT, or radioimmunotherapy (RIT) exploiting the Auger electrons or β-particle emissions, respectively, we propose that panitumumab-MCP could be a useful theranostic agent for EGFR-positive PnCa. Bioconjugation was achieved by reaction of a hydrazine nicotinamide (HyNIC) group on the MCP with an aryl aromatic aldehyde introduced into panitumumab by reaction with succinimidyl-4-formylbenzamide (S-4FB). The conjugation reaction was monitored by measurement of the chromophoric bis-aryl hydrazone bond formed (ε350 nm = 24 500 M-1 cm-1) to achieve two MCPs/panitumumab. Labeling of panitumumab-MCP with 111In or 177Lu demonstrated that masses as small as 0.1 μg were labeled to >90% labeling efficiency (L.E.) and a specific activity (SA) of >70 MBq/μg. Panitumumab-DOTA incorporating two DOTA per mAb was labeled with 111In or 177Lu to a maximum SA of 65 MBq/μg and 46 MBq/μg, respectively. Panitumumab-MCP-177Lu exhibited saturable binding to EGFR-overexpressing MDA-MB-468 human breast cancer cells. The Kd for binding of panitumumab-MCP-177Lu to EGFR (2.2 ± 0.6 nmol/L) was not significantly different than panitumumab-DOTA-177Lu (1.0 ± 0.4 nmol/L). 111In and 177Lu were stably complexed to panitumumab-MCP. Panitumumab-MCP-111In exhibited similar whole body retention (55-60%) as panitumumab-DOTA-111In in NOD-scid mice up to 72 h postinjection (p.i.) and equivalent excretion of radioactivity into the urine and feces. The uptake of panitumumab-MCP-111In in most normal tissues in NOD-scid mice with EGFR-positive PANC-1 human pancreatic cancer (PnCa) xenografts at 72 h p.i. was not significantly different than panitumumab-DOTA-111In, except for the liver which was 3-fold greater for panitumumab-MCP-111In. Tumor uptake of panitumumab-MCP-111In (6.9 ± 1.3%ID/g) was not significantly different than panitumumab-DOTA-11In (6.6 ± 3.3%ID/g). Tumor uptake of panitumumab-MCP-111In and panitumumab-DOTA-111In were reduced by preadministration of excess panitumumab, suggesting EGFR-mediated uptake. Tumor uptake of nonspecific IgG-MCP (5.4 ± 0.3%ID/g) was unexpectedly similar to panitumumab-MCP-111In. An increased hydrodynamic radius of IgG when conjugated to an MCP may encourage tumor uptake via the enhanced permeability and retention (EPR) effect. Tumor uptake of panitumumab-DOTA-111In was 3.5-fold significantly higher than IgG-DOTA-111In. PANC-1 tumors were imaged by microSPECT/CT at 72 h p.i. of panitumumab-MCP-111In or panitumumab-DOTA-111In. Tumors were not visualized with preadministration of excess panitumumab to block EGFR, or with nonspecific IgG radioimmunoconjugates. We conclude that linking panitumumab to an MCP enabled higher SA labeling with 111In and 177Lu than DOTA-conjugated panitumumab, with preserved EGFR binding in vitro and comparable tumor localization in vivo in mice with s.c. PANC-1 human PnCa xenografts. Normal tissue distribution was similar except for the liver which was higher for the polymer radioimmunoconjugates.
Collapse
Affiliation(s)
- Sadaf Aghevlian
- Department of Pharmaceutical Sciences , University of Toronto , 144 College Street , Toronto , ON M5S 3M2 , Canada
| | - Yijie Lu
- Department of Chemistry , University of Toronto , 80 St. George Street , Toronto , ON M5S 3H6 , Canada
| | - Mitchell A Winnik
- Department of Chemistry , University of Toronto , 80 St. George Street , Toronto , ON M5S 3H6 , Canada
| | - David W Hedley
- Department of Medical Oncology, Princess Margaret Cancer Centre , 610 University Avenue , Toronto , ON M5G 2M9 , Canada
| | - Raymond M Reilly
- Department of Pharmaceutical Sciences , University of Toronto , 144 College Street , Toronto , ON M5S 3M2 , Canada.,Department of Medical Imaging , University of Toronto , 263 McCaul Street , Toronto , ON M5T 1W7 , Canada.,Toronto General Research Institute and Joint Department of Medical Imaging , University Health Network , 200 Elizabeth Street , Toronto , ON M5G 2C4 , Canada
| |
Collapse
|