1
|
Ramogida C, Price E. Transition and Post-Transition Radiometals for PET Imaging and Radiotherapy. Methods Mol Biol 2024; 2729:65-101. [PMID: 38006492 DOI: 10.1007/978-1-0716-3499-8_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2023]
Abstract
Radiometals are an exciting class of radionuclides because of the large number of metallic elements available that have medically useful isotopes. To properly harness radiometals, they must be securely bound by chelators, which must be carefully matched to the radiometal ion to maximize radiolabeling performance and the stability of the resulting complex. This chapter focuses on practical aspects of radiometallation chemistry including chelator selection, radiolabeling procedures and conditions, radiolysis prevention, purification, quality control, requisite equipment and reagents, and useful tips.
Collapse
Affiliation(s)
- Caterina Ramogida
- Department of Chemistry, Simon Fraser University, Burnaby, BC, Canada.
- Life Sciences Division, TRIUMF, Vancouver, BC, Canada.
| | - Eric Price
- Department of Chemistry, College of Arts and Science, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
2
|
Radiometals—Chemistry and radiolabeling. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00044-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
3
|
George KJH, Borjian S, Cross MC, Hicks JW, Schaffer P, Kovacs MS. Expanding the PET radioisotope universe utilizing solid targets on small medical cyclotrons. RSC Adv 2021; 11:31098-31123. [PMID: 35498914 PMCID: PMC9041346 DOI: 10.1039/d1ra04480j] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 08/25/2021] [Indexed: 12/17/2022] Open
Abstract
Molecular imaging with medical radioisotopes enables the minimally-invasive monitoring of aberrant biochemical, cellular and tissue-level processes in living subjects. The approach requires the administration of radiotracers composed of radioisotopes attached to bioactive molecules, the pairing of which considers several aspects of the radioisotope in addition to the biological behavior of the targeting molecule to which it is attached. With the advent of modern cellular and biochemical techniques, there has been a virtual explosion in potential disease recognition antigens as well as targeting moieties, which has subsequently opened new applications for a host of emerging radioisotopes with well-matched properties. Additionally, the global radioisotope production landscape has changed rapidly, with reactor-based production and its long-defined, large-scale centralized manufacturing and distribution paradigm shifting to include the manufacture and distribution of many radioisotopes via a worldwide fleet of cyclotrons now in operation. Cyclotron-based radioisotope production has become more prevalent given the commercial availability of instruments, coupled with the introduction of new target hardware, process automation and target manufacturing methods. These advances enable sustained, higher-power irradiation of solid targets that allow hospital-based radiopharmacies to produce a suite of radioisotopes that drive research, clinical trials, and ultimately clinical care. Over the years, several different radioisotopes have been investigated and/or selected for radiolabeling due to favorable decay characteristics (i.e. a suitable half-life, high probability of positron decay, etc.), well-elucidated chemistry, and a feasible production framework. However, longer-lived radioisotopes have surged in popularity given recent regulatory approvals and incorporation of radiopharmaceuticals into patient management within the medical community. This review focuses on the applications, nuclear properties, and production and purification methods for some of the most frequently used/emerging positron-emitting, solid-target-produced radioisotopes that can be manufactured using small-to-medium size cyclotrons (≤24 MeV).
Collapse
Affiliation(s)
- K J H George
- Lawson Health Research Institute 268 Grosvenor Street London ON N6A 4V2 Canada
- Medical Biophysics, Western University 1151 Richmond Street N. London ON N6A 5C1 Canada
| | - S Borjian
- ARTMS 301-4475 Wayburn Drive Burnaby BC V5G 4X4 Canada
| | - M C Cross
- ARTMS 301-4475 Wayburn Drive Burnaby BC V5G 4X4 Canada
| | - J W Hicks
- Lawson Health Research Institute 268 Grosvenor Street London ON N6A 4V2 Canada
- Medical Biophysics, Western University 1151 Richmond Street N. London ON N6A 5C1 Canada
| | - P Schaffer
- Life Sciences, TRIUMF 4004 Wesbrook Mall Vancouver BC V6T 2A3 Canada
- ARTMS 301-4475 Wayburn Drive Burnaby BC V5G 4X4 Canada
- Radiology, University of British Columbia 2775 Laurel St Vancouver BC V5Z 1M9 Canada
- Chemistry, Simon Fraser University 8888 University Dr Burnaby BC V5A 1S6 Canada
| | - M S Kovacs
- Lawson Health Research Institute 268 Grosvenor Street London ON N6A 4V2 Canada
- Medical Biophysics, Western University 1151 Richmond Street N. London ON N6A 5C1 Canada
- Medical Imaging, Western University 1151 Richmond Street N. London ON N6A 5C1 Canada
| |
Collapse
|
4
|
Sier VQ, van der Vorst JR, Quax PHA, de Vries MR, Zonoobi E, Vahrmeijer AL, Dekkers IA, de Geus-Oei LF, Smits AM, Cai W, Sier CFM, Goumans MJTH, Hawinkels LJAC. Endoglin/CD105-Based Imaging of Cancer and Cardiovascular Diseases: A Systematic Review. Int J Mol Sci 2021; 22:4804. [PMID: 33946583 PMCID: PMC8124553 DOI: 10.3390/ijms22094804] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 04/27/2021] [Accepted: 04/28/2021] [Indexed: 02/07/2023] Open
Abstract
Molecular imaging of pathologic lesions can improve efficient detection of cancer and cardiovascular diseases. A shared pathophysiological feature is angiogenesis, the formation of new blood vessels. Endoglin (CD105) is a coreceptor for ligands of the Transforming Growth Factor-β (TGF-β) family and is highly expressed on angiogenic endothelial cells. Therefore, endoglin-based imaging has been explored to visualize lesions of the aforementioned diseases. This systematic review highlights the progress in endoglin-based imaging of cancer, atherosclerosis, myocardial infarction, and aortic aneurysm, focusing on positron emission tomography (PET), single-photon emission computed tomography (SPECT), magnetic resonance imaging (MRI), near-infrared fluorescence (NIRF) imaging, and ultrasound imaging. PubMed was searched combining the following subjects and their respective synonyms or relevant subterms: "Endoglin", "Imaging/Image-guided surgery". In total, 59 papers were found eligible to be included: 58 reporting about preclinical animal or in vitro models and one ex vivo study in human organs. In addition to exact data extraction of imaging modality type, tumor or cardiovascular disease model, and tracer (class), outcomes were described via a narrative synthesis. Collectively, the data identify endoglin as a suitable target for intraoperative and diagnostic imaging of the neovasculature in tumors, whereas for cardiovascular diseases, the evidence remains scarce but promising.
Collapse
Affiliation(s)
- Vincent Q. Sier
- Department of Surgery, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (V.Q.S.); (J.R.v.d.V.); (P.H.A.Q.); (M.R.d.V.); (E.Z.); (A.L.V.)
| | - Joost R. van der Vorst
- Department of Surgery, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (V.Q.S.); (J.R.v.d.V.); (P.H.A.Q.); (M.R.d.V.); (E.Z.); (A.L.V.)
| | - Paul H. A. Quax
- Department of Surgery, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (V.Q.S.); (J.R.v.d.V.); (P.H.A.Q.); (M.R.d.V.); (E.Z.); (A.L.V.)
| | - Margreet R. de Vries
- Department of Surgery, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (V.Q.S.); (J.R.v.d.V.); (P.H.A.Q.); (M.R.d.V.); (E.Z.); (A.L.V.)
| | - Elham Zonoobi
- Department of Surgery, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (V.Q.S.); (J.R.v.d.V.); (P.H.A.Q.); (M.R.d.V.); (E.Z.); (A.L.V.)
- Edinburgh Molecular Imaging Ltd. (EMI), Edinburgh EH16 4UX, UK
| | - Alexander L. Vahrmeijer
- Department of Surgery, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (V.Q.S.); (J.R.v.d.V.); (P.H.A.Q.); (M.R.d.V.); (E.Z.); (A.L.V.)
| | - Ilona A. Dekkers
- Department of Radiology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands;
| | - Lioe-Fee de Geus-Oei
- Department of Radiology, Section of Nuclear Medicine, Leiden University Medical Center, 2300 RC Leiden, The Netherlands;
- Biomedical Photonic Imaging Group, University of Twente, 7500 AE Enschede, The Netherlands
| | - Anke M. Smits
- Department of Cell & Chemical Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (A.M.S.); (M.J.T.H.G.)
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI 53705, USA;
| | - Cornelis F. M. Sier
- Department of Surgery, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (V.Q.S.); (J.R.v.d.V.); (P.H.A.Q.); (M.R.d.V.); (E.Z.); (A.L.V.)
- Percuros B.V., 2333 CL Leiden, The Netherlands
| | - Marie José T. H. Goumans
- Department of Cell & Chemical Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (A.M.S.); (M.J.T.H.G.)
| | - Lukas J. A. C. Hawinkels
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands;
| |
Collapse
|
5
|
Oroujeni M, Xu T, Gagnon K, Rinne SS, Weis J, Garousi J, Andersson KG, Löfblom J, Orlova A, Tolmachev V. The Use of a Non-Conventional Long-Lived Gallium Radioisotope 66Ga Improves Imaging Contrast of EGFR Expression in Malignant Tumours Using DFO-ZEGFR:2377 Affibody Molecule. Pharmaceutics 2021; 13:pharmaceutics13020292. [PMID: 33672373 PMCID: PMC7926986 DOI: 10.3390/pharmaceutics13020292] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/13/2021] [Accepted: 02/19/2021] [Indexed: 12/18/2022] Open
Abstract
Epidermal growth factor receptor (EGFR) is overexpressed in many malignancies. EGFR-targeted therapy extends survival of patients with disseminated cancers. Radionuclide molecular imaging of EGFR expression would make EGFR-directed treatment more personalized and therefore more efficient. A previous study demonstrated that affibody molecule [68Ga]Ga-DFO-ZEGFR:2377 permits specific positron-emission tomography (PET) imaging of EGFR expression in xenografts at 3 h after injection. We anticipated that imaging at 24 h after injection would provide higher contrast, but this is prevented by the short half-life of 68Ga (67.6 min). Here, we therefore tested the hypothesis that the use of the non-conventional long-lived positron emitter 66Ga (T1/2 = 9.49 h, β+ = 56.5%) would permit imaging with higher contrast. 66Ga was produced by the 66Zn(p,n)66Ga nuclear reaction and DFO-ZEGFR:2377 was efficiently labelled with 66Ga with preserved binding specificity in vitro and in vivo. At 24 h after injection, [66Ga]Ga-DFO-ZEGFR:2377 provided 3.9-fold higher tumor-to-blood ratio and 2.3-fold higher tumor-to-liver ratio than [68Ga]Ga-DFO-ZEGFR:2377 at 3 h after injection. At the same time point, [66Ga]Ga-DFO-ZEGFR:2377 provided 1.8-fold higher tumor-to-blood ratio, 3-fold higher tumor-to-liver ratio, 1.9-fold higher tumor-to-muscle ratio and 2.3-fold higher tumor-to-bone ratio than [89Zr]Zr-DFO-ZEGFR:2377. Biodistribution data were confirmed by whole body PET combined with magnetic resonance imaging (PET/MRI). The use of the positron emitter 66Ga for labelling of DFO-ZEGFR:2377 permits PET imaging of EGFR expression at 24 h after injection and improves imaging contrast.
Collapse
Affiliation(s)
- Maryam Oroujeni
- Department of Immunology, Genetics and Pathology, Uppsala University, 75185 Uppsala, Sweden; (M.O.); (T.X.); (J.G.)
| | - Tianqi Xu
- Department of Immunology, Genetics and Pathology, Uppsala University, 75185 Uppsala, Sweden; (M.O.); (T.X.); (J.G.)
| | - Katherine Gagnon
- GE Healthcare, GEMS PET Systems, 75015 Uppsala, Sweden;
- Department of Medicinal Chemistry, Uppsala University, 75183 Uppsala, Sweden; (S.S.R.); (A.O.)
| | - Sara S. Rinne
- Department of Medicinal Chemistry, Uppsala University, 75183 Uppsala, Sweden; (S.S.R.); (A.O.)
| | - Jan Weis
- Department of Medical Physics, Uppsala University Hospital, 75185 Uppsala, Sweden;
| | - Javad Garousi
- Department of Immunology, Genetics and Pathology, Uppsala University, 75185 Uppsala, Sweden; (M.O.); (T.X.); (J.G.)
| | - Ken G. Andersson
- Department of Protein Science, KTH Royal Institute of Technology, 10691 Stockholm, Sweden; (K.G.A.); (J.L.)
| | - John Löfblom
- Department of Protein Science, KTH Royal Institute of Technology, 10691 Stockholm, Sweden; (K.G.A.); (J.L.)
| | - Anna Orlova
- Department of Medicinal Chemistry, Uppsala University, 75183 Uppsala, Sweden; (S.S.R.); (A.O.)
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, 75185 Uppsala, Sweden; (M.O.); (T.X.); (J.G.)
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia
- Correspondence:
| |
Collapse
|
6
|
Rinne SS, Abouzayed A, Gagnon K, Tolmachev V, Orlova A. 66Ga-PET-imaging of GRPR-expression in prostate cancer: production and characterization of [ 66Ga]Ga-NOTA-PEG 2-RM26. Sci Rep 2021; 11:3631. [PMID: 33574368 PMCID: PMC7878787 DOI: 10.1038/s41598-021-82995-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 01/26/2021] [Indexed: 12/16/2022] Open
Abstract
Molecular imaging of the gastrin-releasing peptide receptor (GRPR) could improve patient management in prostate cancer. This study aimed to produce gallium-66 (T½ = 9.5 h) suitable for radiolabeling, and investigate the imaging properties of gallium-66 labeled GRPR-antagonist NOTA-PEG2-RM26 for later-time point PET-imaging of GRPR expression. Gallium-66 was cyclotron-produced using a liquid target, and enriched [66Zn]Zn(NO3)2. In vitro, [66Ga]Ga-NOTA-PEG2-RM26 was characterized in GRPR-expressing PC-3 prostate cancer cells. In vivo, specificity test and biodistribution studies were performed 3 h and 22 h pi in PC-3 xenografted mice. microPET/MR was performed 3 h and 22 h pi. Biodistribution of [66Ga]Ga-NOTA-PEG2-RM26 was compared with [68Ga]Ga-NOTA-PEG2-RM26 3 h pi. [66Ga]Ga-NOTA-PEG2-RM26 was successfully prepared with preserved binding specificity and high affinity towards GRPR. [66Ga]Ga-NOTA-PEG2-RM26 cleared rapidly from blood via kidneys. Tumor uptake was GRPR-specific and exceeded normal organ uptake. Normal tissue clearance was limited, resulting in no improvement of tumor-to-organ ratios with time. Tumors could be clearly visualized using microPET/MR. Gallium-66 was successfully produced and [66Ga]Ga-NOTA-PEG2-RM26 was able to clearly visualize GRPR-expression both shortly after injection and on the next day using PET. However, delayed imaging did not improve contrast for Ga-labeled NOTA-PEG2-RM26.
Collapse
Affiliation(s)
- Sara S Rinne
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Ayman Abouzayed
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | | | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Centrum for Oncotheranostics, National Research Tomsk Polytechnic University, Tomsk, Russia
| | - Anna Orlova
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden.
- Centrum for Oncotheranostics, National Research Tomsk Polytechnic University, Tomsk, Russia.
- Science for Life Laboratory, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
7
|
Sugo Y, Miyachi R, Maruyama YH, Ohira SI, Mori M, Ishioka NS, Toda K. Electrodialytic Handling of Radioactive Metal Ions for Preparation of Tracer Reagents. Anal Chem 2020; 92:14953-14958. [PMID: 32959650 DOI: 10.1021/acs.analchem.0c02456] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Radioactive metals are applied in biochemistry, medical diagnosis such as positron emission tomography (PET), and cancer therapy. However, the activity of radioisotopes exponentially decreases with time; therefore, rapid and reliable probe preparation methods are strongly recommended. In the present study, electrodialytic radioactive metal ion handling is studied for counter ion conversion and in-line probe synthesis. Presently, counter ion conversion and probe synthesis are achieved by evaporative dryness and solution mixing, respectively. Evaporative dryness is time-consuming and is a possible process that can lead to loss of radioactive metal ions. Mixing of solutions for synthesis makes dilution and undesirable effects of counter ion on the synthesis. An optimized electrodialytic flow device can transfer a radioisotope, 64Cu2+, with high recovery from HCl matrices to HNO3 (∼100%). Matrices can also be transferred into acetic acid and citric acid, even though the concentration of the metal ion is at the picomolar level. The ion transfer can also be achieved with simultaneous counter ion conversion, complex synthesis, and enrichment. When the ligand was dissolved in an acceptor solution, the transferred metal ions from the donor were well mixed and formed a complex with the ligand in-line. The efficiency of the synthesis was ∼100% for 1.0 pM 64Cu. A relatively larger donor-to-acceptor flow rate can enrich the metal ion in the acceptor solution continuously. The flow rate ratio of 10 (donor/acceptor) can achieve 10 times enrichment. The present electrodialytic ion handling method can treat ultra-trace radioisotopes in a closed system. With this method, rapid, effective, and safe radioisotope treatments were achieved.
Collapse
Affiliation(s)
- Yumi Sugo
- Department of Radiation-Applied Biology Research, Takasaki Advanced Radiation Research Institute, National Institutes for Quantum and Radiological Science and Technology, 1233 Watanuki, Takasaki 370-1292, Japan
| | - Ryoma Miyachi
- Department of Chemistry, Kumamoto University, 2-39-1 Kurokami, Chuo-ku, Kumamoto 860-8555, Japan
| | - Yo-Hei Maruyama
- Faculty of Science and Technology, Kochi University, 2-5-1, Akebono-cho, Kochi 780-8520, Japan
| | - Shin-Ichi Ohira
- Department of Chemistry, Kumamoto University, 2-39-1 Kurokami, Chuo-ku, Kumamoto 860-8555, Japan
| | - Masanobu Mori
- Faculty of Science and Technology, Kochi University, 2-5-1, Akebono-cho, Kochi 780-8520, Japan
| | - Noriko S Ishioka
- Department of Radiation-Applied Biology Research, Takasaki Advanced Radiation Research Institute, National Institutes for Quantum and Radiological Science and Technology, 1233 Watanuki, Takasaki 370-1292, Japan
| | - Kei Toda
- Department of Chemistry, Kumamoto University, 2-39-1 Kurokami, Chuo-ku, Kumamoto 860-8555, Japan
| |
Collapse
|
8
|
Dewulf J, Adhikari K, Vangestel C, Wyngaert TVD, Elvas F. Development of Antibody Immuno-PET/SPECT Radiopharmaceuticals for Imaging of Oncological Disorders-An Update. Cancers (Basel) 2020; 12:E1868. [PMID: 32664521 PMCID: PMC7408676 DOI: 10.3390/cancers12071868] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/06/2020] [Accepted: 07/10/2020] [Indexed: 01/12/2023] Open
Abstract
Positron emission tomography (PET) and single-photon emission computed tomography (SPECT) are molecular imaging strategies that typically use radioactively labeled ligands to selectively visualize molecular targets. The nanomolar sensitivity of PET and SPECT combined with the high specificity and affinity of monoclonal antibodies have shown great potential in oncology imaging. Over the past decades a wide range of radio-isotopes have been developed into immuno-SPECT/PET imaging agents, made possible by novel conjugation strategies (e.g., site-specific labeling, click chemistry) and optimization and development of novel radiochemistry procedures. In addition, new strategies such as pretargeting and the use of antibody fragments have entered the field of immuno-PET/SPECT expanding the range of imaging applications. Non-invasive imaging techniques revealing tumor antigen biodistribution, expression and heterogeneity have the potential to contribute to disease diagnosis, therapy selection, patient stratification and therapy response prediction achieving personalized treatments for each patient and therefore assisting in clinical decision making.
Collapse
Affiliation(s)
- Jonatan Dewulf
- Molecular Imaging Center Antwerp, Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, B-2610 Wilrijk, Belgium; (J.D.); (C.V.); (T.V.D.W.)
- Department of Nuclear Medicine, Antwerp University Hospital, Wilrijkstraat 10, B-2650 Edegem, Belgium
| | - Karuna Adhikari
- Faculty of Pharmaceutical Biomedical and Veterinary Sciences, Medicinal Chemistry, University of Antwerp, Universiteitsplein 1, B-2610 Wilrijk, Belgium;
| | - Christel Vangestel
- Molecular Imaging Center Antwerp, Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, B-2610 Wilrijk, Belgium; (J.D.); (C.V.); (T.V.D.W.)
- Department of Nuclear Medicine, Antwerp University Hospital, Wilrijkstraat 10, B-2650 Edegem, Belgium
| | - Tim Van Den Wyngaert
- Molecular Imaging Center Antwerp, Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, B-2610 Wilrijk, Belgium; (J.D.); (C.V.); (T.V.D.W.)
- Department of Nuclear Medicine, Antwerp University Hospital, Wilrijkstraat 10, B-2650 Edegem, Belgium
| | - Filipe Elvas
- Molecular Imaging Center Antwerp, Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, B-2610 Wilrijk, Belgium; (J.D.); (C.V.); (T.V.D.W.)
- Department of Nuclear Medicine, Antwerp University Hospital, Wilrijkstraat 10, B-2650 Edegem, Belgium
| |
Collapse
|
9
|
Abstract
Immuno-positron emission tomography (immunoPET) is a paradigm-shifting molecular imaging modality combining the superior targeting specificity of monoclonal antibody (mAb) and the inherent sensitivity of PET technique. A variety of radionuclides and mAbs have been exploited to develop immunoPET probes, which has been driven by the development and optimization of radiochemistry and conjugation strategies. In addition, tumor-targeting vectors with a short circulation time (e.g., Nanobody) or with an enhanced binding affinity (e.g., bispecific antibody) are being used to design novel immunoPET probes. Accordingly, several immunoPET probes, such as 89Zr-Df-pertuzumab and 89Zr-atezolizumab, have been successfully translated for clinical use. By noninvasively and dynamically revealing the expression of heterogeneous tumor antigens, immunoPET imaging is gradually changing the theranostic landscape of several types of malignancies. ImmunoPET is the method of choice for imaging specific tumor markers, immune cells, immune checkpoints, and inflammatory processes. Furthermore, the integration of immunoPET imaging in antibody drug development is of substantial significance because it provides pivotal information regarding antibody targeting abilities and distribution profiles. Herein, we present the latest immunoPET imaging strategies and their preclinical and clinical applications. We also emphasize current conjugation strategies that can be leveraged to develop next-generation immunoPET probes. Lastly, we discuss practical considerations to tune the development and translation of immunoPET imaging strategies.
Collapse
Affiliation(s)
- Weijun Wei
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, 1111 Highland Avenue, Room 7137, Madison, Wisconsin 53705, United States
| | - Zachary T Rosenkrans
- Department of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Jianjun Liu
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Gang Huang
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Quan-Yong Luo
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, 1111 Highland Avenue, Room 7137, Madison, Wisconsin 53705, United States
- Department of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin 53705, United States
| |
Collapse
|
10
|
In vivo imaging of TGFβ signalling components using positron emission tomography. Drug Discov Today 2019; 24:2258-2272. [DOI: 10.1016/j.drudis.2019.08.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 08/01/2019] [Accepted: 08/28/2019] [Indexed: 12/21/2022]
|
11
|
Rodríguez-Villafuerte M, Hernández EM, Alva-Sánchez H, Martínez-Dávalos A, Ávila-Rodríguez MA. Positron range effects of 66Ga in small-animal PET imaging. Phys Med 2019; 67:50-57. [PMID: 31669670 DOI: 10.1016/j.ejmp.2019.10.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 09/25/2019] [Accepted: 10/09/2019] [Indexed: 11/18/2022] Open
Abstract
PURPOSE Gallium-66 is a non-conventional positron emitter that stands out not only for its high potential to label peptides, proteins and antibodies, but also because it can provide spatio-temporal information of relatively slow physiological processes in the body due to its conveniently long half-life of 9.5 h. However, 66Ga emits the most energetic positrons for PET imaging. The lack of information of the positron range effect on spatial resolution for this positron emitter is an issue, particularly in preclinical imaging. METHODS The line spread function (LSF) in tissue-equivalent materials with densities between 0.2 and 1.93 g/cm3 was obtained with 66Ga and 18F. A complementary study with the NEMA NU 4-2008 image quality phantom is also included. RESULTS High-energy positrons moving in lower density materials produce far-reaching activity distributions. The LSFs were characterized with Lorentzian-Gaussian fits, with spatial resolution (FWHM) in the 2.14-3.2 mm range, and long tails extending a few tens of mm depending on the material type and density. A narrowing of the LSF was observed for lung-equivalent materials, indicating the lack of enough material for the positron annihilation to take place. The NEMA NU 4-2008 image quality phantom produced blurred images, notoriously observed in the hot and cold cylinders used for evaluation of recovery coefficients (RC) and spill-over ratios (SOR), producing very low RC and very large SOR. CONCLUSIONS Quantitative PET imaging with the non-conventional 66Ga is hampered due to the large range of its high-energy positrons affecting both spatial resolution and activity concentration quantification.
Collapse
Affiliation(s)
| | - E M Hernández
- Instituto de Física, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - H Alva-Sánchez
- Instituto de Física, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - A Martínez-Dávalos
- Instituto de Física, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - M A Ávila-Rodríguez
- Unidad Radiofarmacia-Ciclotrón, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
12
|
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
|
13
|
Amor-Coarasa A, Kelly JM, Ponnala S, Nikolopoulou A, Williams C, Babich JW. 66Ga: A Novelty or a Valuable Preclinical Screening Tool for the Design of Targeted Radiopharmaceuticals? Molecules 2018; 23:molecules23102575. [PMID: 30304795 PMCID: PMC6222850 DOI: 10.3390/molecules23102575] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 10/01/2018] [Accepted: 10/05/2018] [Indexed: 11/30/2022] Open
Abstract
Emerging interest in extending the plasma half-life of small molecule radioligands warrants a consideration of the appropriate radionuclide for PET imaging at longer time points (>8 h). Among candidate positron-emitting radionuclides, 66Ga (t1/2 = 9.5 h, β+ = 57%) has suitable nuclear and chemical properties for the labeling and PET imaging of radioligands of this profile. We investigated the value of 66Ga to preclinical screening and the evaluation of albumin-binding PSMA-targeting small molecules. 66Ga was produced by irradiation of a natZn target. 66Ga3+ ions were separated from Zn2+ ions by an optimized UTEVA anion exchange column that retained 99.99987% of Zn2+ ions and allowed 90.2 ± 2.8% recovery of 66Ga3+. Three ligands were radiolabeled in 46.4 ± 20.5%; radiochemical yield and >90% radiochemical purity. Molar activity was 632 ± 380 MBq/µmol. Uptake in the tumor and kidneys at 1, 3, 6, and 24 h p.i. was determined by µPET/CT imaging and more completely predicted the distribution kinetics than uptake of the [68Ga]Ga-labeled ligands did. Although there are multiple challenges to the use of 66Ga for clinical PET imaging, it can be a valuable research tool for ligand screening and preclinical imaging beyond 24 h.
Collapse
Affiliation(s)
- Alejandro Amor-Coarasa
- Division of Radiopharmaceutical Sciences and MI3, Department of Radiology, Weill Cornell Medicine, New York, NY 10065, USA.
| | - James M Kelly
- Division of Radiopharmaceutical Sciences and MI3, Department of Radiology, Weill Cornell Medicine, New York, NY 10065, USA.
| | - Shashikanth Ponnala
- Division of Radiopharmaceutical Sciences and MI3, Department of Radiology, Weill Cornell Medicine, New York, NY 10065, USA.
| | - Anastasia Nikolopoulou
- Division of Radiopharmaceutical Sciences and MI3, Department of Radiology, Weill Cornell Medicine, New York, NY 10065, USA.
- Citigroup Biomedical Imaging Center, Weill Cornell Medicine, New York, NY 10065, USA.
| | - Clarence Williams
- Division of Radiopharmaceutical Sciences and MI3, Department of Radiology, Weill Cornell Medicine, New York, NY 10065, USA.
| | - John W Babich
- Division of Radiopharmaceutical Sciences and MI3, Department of Radiology, Weill Cornell Medicine, New York, NY 10065, USA.
- Citigroup Biomedical Imaging Center, Weill Cornell Medicine, New York, NY 10065, USA.
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|
14
|
Richard M, Ariztia J, Lamandé-Langle S, Pellegrini Moïse N. Sugar γ-Amino Acids as Building Blocks for the Synthesis of Cyclic Neoglycopeptides. ChemistrySelect 2018. [DOI: 10.1002/slct.201802146] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
| | - Julen Ariztia
- Université de Lorraine, CNRS, L2CM; F-5400 Nancy France
| | | | | |
Collapse
|
15
|
Ehlerding EB, Ferreira CA, Aluicio-Sarduy E, Jiang D, Lee HJ, Theuer CP, Engle JW, Cai W. 86/90Y-Based Theranostics Targeting Angiogenesis in a Murine Breast Cancer Model. Mol Pharm 2018; 15:2606-2613. [PMID: 29787283 PMCID: PMC6028311 DOI: 10.1021/acs.molpharmaceut.8b00133] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Angiogenesis is widely recognized as one of the hallmarks of cancer. Therefore, imaging and therapeutic agents targeted to angiogenic vessels may be widely applicable in many types of cancer. To this end, the theranostic isotope pair, 86Y and 90Y, were used to create a pair of agents for targeted imaging and therapy of neovasculature in murine breast cancer models using a chimeric anti-CD105 antibody, TRC105. Serial positron emission tomography imaging with 86Y-DTPA-TRC105 demonstrated high uptake in 4T1 tumors, peaking at 9.6 ± 0.3%ID/g, verified through ex vivo studies. Additionally, promising results were obtained in therapeutic studies with 90Y-DTPA-TRC105, wherein significantly ( p < 0.05) decreased tumor volumes were observed for the targeted treatment group over all control groups near the end of the study. Dosimetric extrapolation and tissue histological analysis corroborated trends found in vivo. Overall, this study demonstrated the potential of the pair 86/90Y for theranostics, enabling personalized treatments for cancer.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/chemistry
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal/therapeutic use
- Antineoplastic Agents/chemistry
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Cell Line, Tumor/transplantation
- Drug Screening Assays, Antitumor
- Female
- Humans
- Immunoconjugates/chemistry
- Immunoconjugates/pharmacology
- Immunoconjugates/therapeutic use
- Mammary Neoplasms, Experimental/diagnostic imaging
- Mammary Neoplasms, Experimental/pathology
- Mammary Neoplasms, Experimental/radiotherapy
- Mice
- Mice, Inbred BALB C
- Neovascularization, Pathologic/diagnostic imaging
- Neovascularization, Pathologic/drug therapy
- Positron-Emission Tomography/methods
- Radioimmunotherapy/methods
- Theranostic Nanomedicine/methods
- Tissue Distribution
- Treatment Outcome
- Yttrium Radioisotopes/chemistry
- Yttrium Radioisotopes/pharmacology
- Yttrium Radioisotopes/therapeutic use
Collapse
Affiliation(s)
| | - Carolina A Ferreira
- Department of Biomedical Engineering , Univesity of Wisconsin-Madison , Madison , Wisconsin 53706 , United States
| | | | | | | | - Charles P Theuer
- TRACON Pharmaceuticals, Inc. , San Diego , California 92122 , United States
| | | | - Weibo Cai
- Department of Biomedical Engineering , Univesity of Wisconsin-Madison , Madison , Wisconsin 53706 , United States
- Carbone Comprehensive Cancer Center , University of Wisconsin-Madison , Madison , Wisconsin 53792 , United States
| |
Collapse
|
16
|
Aluicio-Sarduy E, Ellison PA, Barnhart TE, Cai W, Nickles RJ, Engle JW. PET radiometals for antibody labeling. J Labelled Comp Radiopharm 2018; 61:636-651. [PMID: 29341227 PMCID: PMC6050152 DOI: 10.1002/jlcr.3607] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 12/29/2017] [Accepted: 01/05/2018] [Indexed: 02/06/2023]
Abstract
Recent advances in molecular characterization of tumors have made possible the emergence of new types of cancer therapies where traditional cytotoxic drugs and nonspecific chemotherapy can be complemented with targeted molecular therapies. One of the main revolutionary treatments is the use of monoclonal antibodies (mAbs) that selectively target the disseminated tumor cells while sparing normal tissues. mAbs and related therapeutics can be efficiently radiolabeled with a wide range of radionuclides to facilitate preclinical and clinical studies. Non-invasive molecular imaging techniques, such as Positron Emission Tomography (PET), using radiolabeled mAbs provide useful information on the whole-body distribution of the biomolecules, which may enable patient stratification, diagnosis, selection of targeted therapies, evaluation of treatment response, and prediction of dose limiting tissue and adverse effects. In addition, when mAbs are labeled with therapeutic radionuclides, the combination of immunological and radiobiological cytotoxicity may result in enhanced treatment efficacy. The pharmacokinetic profile of antibodies demands the use of long half-life isotopes for longitudinal scrutiny of mAb biodistribution and precludes the use of well-stablished short half-life isotopes. Herein, we review the most promising PET radiometals with chemical and physical characteristics that make the appealing for mAb labeling, highlighting those with theranostic radioisotopes.
Collapse
Affiliation(s)
| | - Paul A. Ellison
- University of Wisconsin-Madison, Department of Medical Physics, Madison, Wisconsin, USA
| | - Todd E. Barnhart
- University of Wisconsin-Madison, Department of Medical Physics, Madison, Wisconsin, USA
| | - Weibo Cai
- University of Wisconsin-Madison, Department of Medical Physics, Madison, Wisconsin, USA
- University of Wisconsin-Madison, Department of Radiology, Madison, Wisconsin, USA
- University of Wisconsin-Madison Carbone Cancer Center, Carbon Cancer Center, Madison, Wisconsin, USA
| | - Robert Jerry Nickles
- University of Wisconsin-Madison, Department of Medical Physics, Madison, Wisconsin, USA
| | - Jonathan W. Engle
- University of Wisconsin-Madison, Department of Medical Physics, Madison, Wisconsin, USA
- University of Wisconsin-Madison, Department of Radiology, Madison, Wisconsin, USA
| |
Collapse
|
17
|
Zhuo H, Zheng B, Liu J, Huang Y, Wang H, Zheng D, Mao N, Meng J, Zhou S, Zhong L, Zhao Y. Efficient targeted tumor imaging and secreted endostatin gene delivery by anti-CD105 immunoliposomes. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:42. [PMID: 29499713 PMCID: PMC5833054 DOI: 10.1186/s13046-018-0712-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 02/15/2018] [Indexed: 12/18/2022]
Abstract
BACKGROUND Anti-CD105 mAb-conjugated immunoliposomes, loaded with secreted mouse endostatin gene, were developed for targeted tumor imaging and antiangiogenic gene therapy. METHODS The liposomes were investigated for size, zeta-potential, lipid content, antibody binding ability, and pcDNA loading capacity. The ability of immunoliposomes to target tumor-derived endothelial cells and perform gene transfer in vitro was measured and their basic biocompatibility was evaluated. A nude mouse/breast cancer xenograft model was used to examine the tumor internalization of fluorescent-labeled liposomes and the clinical potential of immnuoliposomes loaded with pcDNA3.1-CSF1-endostatin. RESULTS Loaded immunoliposomes were homogenously distributed with a well-defined spherical shape and bilayer, diameter of 122 ± 11 nm, and zeta potential + 1.40 mV. No significant differences were observed in body weight, liver index, oxidative stress, or liver and kidney function in mice after liposomes exposure. The addition of CD105 mAb to liposomes conferred the ability to target tumor-derived endothelial cells in vitro and in vivo. Systemic intravenous administration of fluorescent immunoliposomes in the xenograft model resulted in selective and efficient internalization in tumor vasculature. Treatment of mice with pcDNA3.1-CSF1-endostatin-loaded immunoliposomes suppressed tumor growth by 71%. CONCLUSIONS These data demonstrate the advantages of using anti-CD105 mAb-conjugated immunoliposomes to enhance tumor targeting, imaging, and gene transfer applications.
Collapse
Affiliation(s)
- Huiqin Zhuo
- Department of Gastrointestinal Surgery, Institute of Gastrointestinal Oncology, Zhongshan Hospital, Xiamen University, Xiamen, Fujian, 361004, China
| | - Baoshi Zheng
- National Center for International Research of Biological Targeting Diagnosis and Therapy/Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research/Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China.,Department of Cardiothoracic Surgery, First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Jianming Liu
- The Third Xiangya Hospital, Central South University, Changsha, 410083, China
| | - Yong Huang
- National Center for International Research of Biological Targeting Diagnosis and Therapy/Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research/Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Huiling Wang
- National Center for International Research of Biological Targeting Diagnosis and Therapy/Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research/Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Duo Zheng
- Department of Basic Medicine, Shenzhen Key Laboratory of Translational Medicine of Tumor, School of Medicine, Shenzhen University, Shenzhen, Guangdong, 518000, China
| | - Naiquan Mao
- National Center for International Research of Biological Targeting Diagnosis and Therapy/Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research/Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Jinyu Meng
- Biomedical Polymers Laboratory, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Sufang Zhou
- National Center for International Research of Biological Targeting Diagnosis and Therapy/Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research/Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Liping Zhong
- National Center for International Research of Biological Targeting Diagnosis and Therapy/Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research/Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China.
| | - Yongxiang Zhao
- National Center for International Research of Biological Targeting Diagnosis and Therapy/Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research/Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China.
| |
Collapse
|
18
|
Zhan Y, Shi S, Ehlerding EB, Graves SA, Goel S, Engle JW, Liang J, Tian J, Cai W. Radiolabeled, Antibody-Conjugated Manganese Oxide Nanoparticles for Tumor Vasculature Targeted Positron Emission Tomography and Magnetic Resonance Imaging. ACS APPLIED MATERIALS & INTERFACES 2017; 9:38304-38312. [PMID: 29028311 PMCID: PMC5680099 DOI: 10.1021/acsami.7b12216] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Manganese oxide nanoparticles (Mn3O4 NPs) have attracted a great deal of attention in the field of biomedical imaging because of their ability to create an enhanced imaging signal in MRI as novel potent T1 contrast agents. In this study, we present tumor vasculature-targeted imaging in mice using Mn3O4 NPs through conjugation to the anti-CD105 antibody TRC105 and radionuclide copper-64 (64Cu, t1/2: 12.7 h). The Mn3O4 conjugated NPs, 64Cu-NOTA-Mn3O4@PEG-TRC105, exhibited sufficient stability in vitro and in vivo. Serial positron emission tomography (PET) and magnetic resonance imaging (MRI) studies evaluated the pharmacokinetics and demonstrated targeting of 64Cu-NOTA-Mn3O4@PEG-TRC105 to 4T1 murine breast tumors in vivo, compared to 64Cu-NOTA-Mn3O4@PEG. The specificity of 64Cu-NOTA-Mn3O4@PEG-TRC105 for the vascular marker CD105 was confirmed through in vivo, in vitro, and ex vivo experiments. Since Mn3O4 conjugated NPs exhibited desirable properties for T1 enhanced imaging and low toxicity, the tumor-specific Mn3O4 conjugated NPs reported in this study may serve as promising multifunctional nanoplatforms for precise cancer imaging and diagnosis.
Collapse
Affiliation(s)
- Yonghua Zhan
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi’an, Shaanxi, 710071, China
| | - Sixiang Shi
- Department of Radiology, University of Wisconsin–Madison, WI, 53705,USA
| | - Emily B. Ehlerding
- Department of Medical Physics, University of Wisconsin-Madison, WI, 53705,USA
| | - Stephen A. Graves
- Department of Medical Physics, University of Wisconsin-Madison, WI, 53705,USA
| | - Shreya Goel
- Department of Radiology, University of Wisconsin–Madison, WI, 53705,USA
| | - Jonathan W. Engle
- Department of Medical Physics, University of Wisconsin-Madison, WI, 53705,USA
| | - Jimin Liang
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi’an, Shaanxi, 710071, China
- Department of Medical Physics, University of Wisconsin-Madison, WI, 53705,USA
| | - Jie Tian
- Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
- Corresponding Authors: (W. Cai); (J. Tian)
| | - Weibo Cai
- Department of Radiology, University of Wisconsin–Madison, WI, 53705,USA
- University of Wisconsin Carbone Cancer Center, Madison, WI, 53705, USA
- Corresponding Authors: (W. Cai); (J. Tian)
| |
Collapse
|
19
|
Chen F, Goel S, Hernandez R, Graves SA, Shi S, Nickles RJ, Cai W. Dynamic Positron Emission Tomography Imaging of Renal Clearable Gold Nanoparticles. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2016; 12:2775-82. [PMID: 27062146 PMCID: PMC4874869 DOI: 10.1002/smll.201600194] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 03/09/2016] [Indexed: 05/19/2023]
Abstract
Optical imaging has been the primary imaging modality for nearly all of the renal clearable nanoparticles since 2007. Due to the tissue depth penetration limitation, providing accurate organ kinetics non-invasively has long been a huge challenge. Although a more quantitative imaging technique has been developed by labeling nanoparticles with single-photon emission computed tomography (SPECT) isotopes, the low temporal resolution of SPECT still limits its potential for visualizing the rapid dynamic process of renal clearable nanoparticles in vivo. The dynamic positron emission tomography (PET) imaging of renal clearable gold (Au) nanoparticles by labeling them with copper-64 ((64) Cu) to form (64) Cu-NOTA-Au-GSH is reported. Systematic nanoparticle synthesis and characterizations are performed to demonstrate the efficient renal clearance of as-prepared nanoparticles. A rapid renal clearance of (64) Cu-NOTA-Au-GSH is observed (>75%ID at 24 h post-injection) with its elimination half-life calculated to be less than 6 min, over 130 times shorter than previously reported similar nanoparticles. Dynamic PET imaging not only addresses the current challenges in accurately and non-invasively acquiring the organ kinetics, but also potentially provides a highly useful tool for studying renal clearance mechanism of other ultra-small nanoparticles, as well as the diagnosis of kidney diseases in the near future.
Collapse
Affiliation(s)
- Feng Chen
- Department of Radiology, University of Wisconsin – Madison, WI, USA
| | - Shreya Goel
- Materials Science Program, University of Wisconsin - Madison, WI, USA
| | - Reinier Hernandez
- Department of Medical Physics, University of Wisconsin - Madison, WI, USA
| | - Stephen A. Graves
- Department of Medical Physics, University of Wisconsin - Madison, WI, USA
| | - Sixiang Shi
- Materials Science Program, University of Wisconsin - Madison, WI, USA
| | - Robert J. Nickles
- Department of Medical Physics, University of Wisconsin - Madison, WI, USA
| | - Weibo Cai
- Fax: (+1) 608-265-0614, , Homepage: http://mi.wisc.edu
| |
Collapse
|
20
|
Graves SA, Hernandez R, Fonslet J, England CG, Valdovinos HF, Ellison PA, Barnhart TE, Elema DR, Theuer CP, Cai W, Nickles RJ, Severin GW. Novel Preparation Methods of (52)Mn for ImmunoPET Imaging. Bioconjug Chem 2015; 26:2118-24. [PMID: 26317429 DOI: 10.1021/acs.bioconjchem.5b00414] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
(52)Mn (t1/2 = 5.59 d, β(+) = 29.6%, Eβave = 0.24 MeV) shows promise in positron emission tomography (PET) and in dual-modality manganese-enhanced magnetic resonance imaging (MEMRI) applications including neural tractography, stem cell tracking, and biological toxicity studies. The extension to bioconjugate application requires high-specific-activity (52)Mn in a state suitable for macromolecule labeling. To that end a (52)Mn production, purification, and labeling system is presented, and its applicability in preclinical, macromolecule PET is shown using the conjugate (52)Mn-DOTA-TRC105. (52)Mn is produced by 60 μA, 16 MeV proton irradiation of natural chromium metal pressed into a silver disc support. Radiochemical separation proceeds by strong anion exchange chromatography of the dissolved Cr target, employing a semiorganic mobile phase, 97:3 (v:v) ethanol:HCl (11 M, aqueous). The method is 62 ± 14% efficient (n = 7) in (52)Mn recovery, leading to a separation factor from Cr of (1.6 ± 1.0) × 10(6) (n = 4), and an average effective specific activity of 0.8 GBq/μmol (n = 4) in titration against DOTA. (52)Mn-DOTA-TRC105 conjugation and labeling demonstrate the potential for chelation applications. In vivo images acquired using PET/CT in mice bearing 4T1 xenograft tumors are presented. Peak tumor uptake is 18.7 ± 2.7%ID/g at 24 h post injection and ex vivo (52)Mn biodistribution validates the in vivo PET data. Free (52)Mn(2+) (as chloride or acetate) is used as a control in additional mice to evaluate the nontargeted biodistribution in the tumor model.
Collapse
Affiliation(s)
| | | | - Jesper Fonslet
- Technical University of Denmark , Center for Nuclear Technologies, 4000 Roskilde, Denmark
| | | | | | | | | | - Dennis R Elema
- Technical University of Denmark , Center for Nuclear Technologies, 4000 Roskilde, Denmark
| | - Charles P Theuer
- TRACON Pharmaceuticals , San Diego, California 92122, United States
| | | | | | - Gregory W Severin
- Technical University of Denmark , Center for Nuclear Technologies, 4000 Roskilde, Denmark
| |
Collapse
|
21
|
Vectors for the delivery of radiopharmaceuticals in cancer therapeutics. Ther Deliv 2015; 5:893-912. [PMID: 25337647 DOI: 10.4155/tde.14.57] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Internal radiation using radiopharmaceuticals promises efficient cancer therapeutics. The specificity and selectivity required for screening and pinpointing tumor cells for cell-kill has been made possible by targeted ligands based on 'magic bullet' and tracer principle- theories nearing a century. Overexpression of certain receptors has been exploited using biomolecules for targeting. The pragmatic analysis, however, is not as promising compared with the theoretical knowledge of available gamut of vectors and targets. The complex interplay of in vitro and in vivo parameters, and the effect of radionuclides involve a systematic assessment of radiopharmaceuticals as diagnostic and therapeutic agent. This review presents different vectors with their pros and cons, present status and recent design variations followed by a future perspective based on novel approaches.
Collapse
|
22
|
Zhao Q, Zhang Y, Guo J, Li J. A novel molecular probe 131I-K237 targeting tumor angiogenesis in human prostate cancer xenografts. Mol Med Rep 2015; 12:1363-7. [PMID: 25815588 DOI: 10.3892/mmr.2015.3504] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2014] [Accepted: 01/21/2015] [Indexed: 11/06/2022] Open
Abstract
Specific molecular probes are essential for the early diagnosis of prostate cancer. In addition, peptides have been shown to have numerous uses as diagnostic and therapeutic molecular probes. The K237 peptide binds to the vascular endothelial growth factor receptor with high affinity and specificity, and was predicted to have potential use as a probe in tumor angiogenesis. The overall aim of the present study was to assess the diagnostic potential of 131I‑K237 as a molecular probe for prostate cancer. The K237 peptide was radiolabeled with 131I using an Iodogen method. The radiolabeling efficiency and radiochemical purity were found to be 73.7 ± 3.2 and 96.7 ± 0.6%, respectively, which were determined using thin layer chromatography and high performance liquid chromatography in vitro. Cellular uptake and competition binding experiments were used to identify the affinity of 131I‑K237 to LNCaP prostate cancer cells. The binding ratio of 131I‑K237 to LNCaP cells in the experimental group was 95.8 ± 1.5%, whereas the binding ratios in the 5 kBq Na131I, 10 kBq Na131I, 15 kBq Na131I and PBS groups were 8.2 ± 0.4, 8.3 ± 0.2, 8.5 ± 0.2 and 0.0%, respectively. In addition, the binding ratio of 131I‑K237 to LNCaP significantly decreased with the increased dose of unlabeled K237. A total of 40 male BALB/c mice with LNCaP xenografts were used for biodistribution and single photon emission computed tomography imaging analysis. An image was obtained and tumors were visible from 2 h post injection of 131I‑K237. In conclusion, the results of the present study showed that 131I‑K237 had a high affinity for LNCaP cells and may be considered as a candidate diagnostic molecular probe for prostate cancer.
Collapse
Affiliation(s)
- Qian Zhao
- Department of Nuclear Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Yu Zhang
- Department of Nuclear Medicine, Affiliated Hospital of Luzhou Medical College, Luzhou, Sichuan 646000, P.R. China
| | - Jun Guo
- Department of Nuclear Medicine, Postgraduate College of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Juan Li
- Department of Nuclear Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| |
Collapse
|
23
|
Hong H, Wang F, Zhang Y, Graves SA, Eddine SBZ, Yang Y, Theuer CP, Nickles RJ, Wang X, Cai W. Red fluorescent zinc oxide nanoparticle: a novel platform for cancer targeting. ACS APPLIED MATERIALS & INTERFACES 2015; 7:3373-81. [PMID: 25607242 PMCID: PMC4326560 DOI: 10.1021/am508440j] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Multifunctional zinc oxide (ZnO) nanoparticles (NPs) with well-integrated multimodality imaging capacities have generated increasing research interest in the past decade. However, limited progress has been made in developing ZnO NP-based multimodality tumor-imaging agents. Here we developed novel red fluorescent ZnO NPs and described the successful conjugation of 64Cu (t1/2=12.7 h) and TRC105, a chimeric monoclonal antibody against CD105, to these ZnO NPs via well-developed surface engineering procedures. The produced dual-modality ZnO NPs were readily applicable for positron emission tomography (PET) imaging and fluorescence imaging of the tumor vasculature. Their pharmacokinetics and tumor-targeting efficacy/specificity in mice bearing murine breast 4T1 tumor were thoroughly investigated. ZnO NPs with dual-modality imaging properties can serve as an attractive candidate for future cancer theranostics.
Collapse
Affiliation(s)
- Hao Hong
- Department of Radiology, University of Wisconsin - Madison, Wisconsin 53705-2275, United States
| | - Fei Wang
- Department of Materials Science and Engineering, University of Wisconsin - Madison, Wisconsin 53705-2275, United States
| | - Yin Zhang
- Department of Medical Physics, University of Wisconsin - Madison, Wisconsin 53705-2275, United States
| | - Stephen A. Graves
- Department of Medical Physics, University of Wisconsin - Madison, Wisconsin 53705-2275, United States
| | - Savo Bou Zein Eddine
- Department of Radiology, University of Wisconsin - Madison, Wisconsin 53705-2275, United States
- Faculty of Medicine and Medical Center, American University of Beirut, Beirut 11072020, Lebanon
| | - Yunan Yang
- Department of Radiology, University of Wisconsin - Madison, Wisconsin 53705-2275, United States
| | | | - Robert J. Nickles
- Department of Medical Physics, University of Wisconsin - Madison, Wisconsin 53705-2275, United States
| | - Xudong Wang
- Department of Materials Science and Engineering, University of Wisconsin - Madison, Wisconsin 53705-2275, United States
| | - Weibo Cai
- Department of Radiology, University of Wisconsin - Madison, Wisconsin 53705-2275, United States
- Department of Medical Physics, University of Wisconsin - Madison, Wisconsin 53705-2275, United States
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin 53705-2275, United States
| |
Collapse
|
24
|
Guo J, Hong H, Chen G, Shi S, Nayak T, Theuer CP, Barnhart TE, Cai W, Gong S. Theranostic unimolecular micelles based on brush-shaped amphiphilic block copolymers for tumor-targeted drug delivery and positron emission tomography imaging. ACS APPLIED MATERIALS & INTERFACES 2014; 6:21769-79. [PMID: 24628452 PMCID: PMC4163544 DOI: 10.1021/am5002585] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 03/04/2014] [Indexed: 05/24/2023]
Abstract
Brush-shaped amphiphilic block copolymers were conjugated with a monoclonal antibody against CD105 (i.e., TRC105) and a macrocyclic chelator for (64)Cu-labeling to generate multifunctional theranostic unimolecular micelles. The backbone of the brush-shaped amphiphilic block copolymer was poly(2-hydroxyethyl methacrylate) (PHEMA) and the side chains were poly(L-lactide)-poly(ethylene glycol) (PLLA-PEG). The doxorubicin (DOX)-loaded unimolecular micelles showed a pH-dependent drug release profile and a uniform size distribution. A significantly higher cellular uptake of TRC105-conjugated micelles was observed in CD105-positive human umbilical vein endothelial cells (HUVEC) than nontargeted micelles due to CD105-mediated endocytosis. In contrast, similar and extremely low cellular uptake of both targeted and nontargeted micelles was observed in MCF-7 human breast cancer cells (CD105-negative). The difference between the in vivo tumor accumulation of (64)Cu-labeled TRC105-conjugated micelles and that of nontargeted micelles was studied in 4T1 murine breast tumor-bearing mice, by serial positron emission tomography (PET) imaging and validated by biodistribution studies. These multifunctional unimolecular micelles offer pH-responsive drug release, noninvasive PET imaging capability, together with both passive and active tumor-targeting abilities, thus making them a desirable nanoplatform for cancer theranostics.
Collapse
Affiliation(s)
- Jintang Guo
- School
of Chemical Engineering, Tianjin University, Tianjin 300072, China
- Department
of Biomedical Engineering, University of
Wisconsin−Madison, Madison, Wisconsin 53706, United States
- Wisconsin
Institutes for Discovery, University of
Wisconsin−Madison, Madison, Wisconsin 53715, United States
| | - Hao Hong
- Departments
of Radiology and Medical Physics, University
of Wisconsin−Madison, Madison, Wisconsin 53705, United States
| | - Guojun Chen
- Wisconsin
Institutes for Discovery, University of
Wisconsin−Madison, Madison, Wisconsin 53715, United States
- Materials
Science Program, University of Wisconsin−Madison, Madison, Wisconsin53706, United States
| | - Sixiang Shi
- Materials
Science Program, University of Wisconsin−Madison, Madison, Wisconsin53706, United States
| | - Tapas
R. Nayak
- Departments
of Radiology and Medical Physics, University
of Wisconsin−Madison, Madison, Wisconsin 53705, United States
| | | | - Todd E. Barnhart
- Departments
of Radiology and Medical Physics, University
of Wisconsin−Madison, Madison, Wisconsin 53705, United States
| | - Weibo Cai
- Departments
of Radiology and Medical Physics, University
of Wisconsin−Madison, Madison, Wisconsin 53705, United States
- Materials
Science Program, University of Wisconsin−Madison, Madison, Wisconsin53706, United States
| | - Shaoqin Gong
- Department
of Biomedical Engineering, University of
Wisconsin−Madison, Madison, Wisconsin 53706, United States
- Wisconsin
Institutes for Discovery, University of
Wisconsin−Madison, Madison, Wisconsin 53715, United States
- Materials
Science Program, University of Wisconsin−Madison, Madison, Wisconsin53706, United States
| |
Collapse
|
25
|
Hu Y, Li CY, Wang XM, Yang YH, Zhu HL. 1,3,4-Thiadiazole: synthesis, reactions, and applications in medicinal, agricultural, and materials chemistry. Chem Rev 2014; 114:5572-610. [PMID: 24716666 DOI: 10.1021/cr400131u] [Citation(s) in RCA: 344] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Yang Hu
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University , Nanjing 210093, People's Republic of China
| | | | | | | | | |
Collapse
|
26
|
Sihver W, Pietzsch J, Krause M, Baumann M, Steinbach J, Pietzsch HJ. Radiolabeled Cetuximab Conjugates for EGFR Targeted Cancer Diagnostics and Therapy. Pharmaceuticals (Basel) 2014; 7:311-38. [PMID: 24603603 PMCID: PMC3978494 DOI: 10.3390/ph7030311] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 02/11/2014] [Accepted: 02/21/2014] [Indexed: 01/09/2023] Open
Abstract
The epidermal growth factor receptor (EGFR) has evolved over years into a main molecular target for the treatment of different cancer entities. In this regard, the anti-EGFR antibody cetuximab has been approved alone or in combination with: (a) chemotherapy for treatment of colorectal and head and neck squamous cell carcinoma and (b) with external radiotherapy for treatment of head and neck squamous cell carcinoma. The conjugation of radionuclides to cetuximab in combination with the specific targeting properties of this antibody might increase its therapeutic efficiency. This review article gives an overview of the preclinical studies that have been performed with radiolabeled cetuximab for imaging and/or treatment of different tumor models. A particularly promising approach seems to be the treatment with therapeutic radionuclide-labeled cetuximab in combination with external radiotherapy. Present data support an important impact of the tumor micromilieu on treatment response that needs to be further validated in patients. Another important challenge is the reduction of nonspecific uptake of the radioactive substance in metabolic organs like liver and radiosensitive organs like bone marrow and kidneys. Overall, the integration of diagnosis, treatment and monitoring as a theranostic approach appears to be a promising strategy for improvement of individualized cancer treatment.
Collapse
Affiliation(s)
- Wiebke Sihver
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, Dresden 01328, Germany.
| | - Jens Pietzsch
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, Dresden 01328, Germany.
| | - Mechthild Krause
- Department of Radiation Oncology and OncoRay, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden 01307, Germany.
| | - Michael Baumann
- Department of Radiation Oncology and OncoRay, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden 01307, Germany.
| | - Jörg Steinbach
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, Dresden 01328, Germany.
| | - Hans-Jürgen Pietzsch
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, Dresden 01328, Germany.
| |
Collapse
|
27
|
Zhou Y, Baidoo KE, Brechbiel MW. Mapping biological behaviors by application of longer-lived positron emitting radionuclides. Adv Drug Deliv Rev 2013; 65:1098-111. [PMID: 23123291 DOI: 10.1016/j.addr.2012.10.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2012] [Revised: 10/17/2012] [Accepted: 10/23/2012] [Indexed: 02/08/2023]
Abstract
With the technological development of positron emission tomography (PET) and the advent of novel antibody-directed drug delivery systems, longer-lived positron-emitting radionuclides are moving to the forefront to take important roles in tracking the distribution of biotherapeutics such as antibodies, and for monitoring biological processes and responses. Longer half-life radionuclides possess advantages of convenient on-site preparation procedures for both clinical and non-clinical applications. The suitability of the long half-life radionuclides for imaging intact monoclonal antibodies (mAbs) and their respective fragments, which have inherently long biological half-lives, has attracted increased interest in recent years. In this review, we provide a survey of the recent literature as it applies to the development of nine-selected longer-lived positron emitters with half-lives of 9-140h (e.g., (124)I, (64)Cu, (86)Y and (89)Zr), and describe the biological behaviors of radionuclide-labeled mAbs with respect to distribution and targeting characteristics, potential toxicities, biological applications, and clinical translation potentials.
Collapse
Key Words
- (124)I
- (64)Cu
- (86)Y
- (89)Zr
- 1,4,7,10-tetraazacyclododecane-N,N′,N″,N″′-tetraacetic acid
- 1,4,7-triazacyclononane-N,N′,N″-1,4,7-triacetic acid
- 1-N-(4-aminobenzyl)-3,6,10,13,16,19-hexaazabicyclo[6.6.6]eicosane-1,8-diamine
- 1-oxa-4,7,1-tetraazacyclododecane-5-S-(4-isothiocyanatobenzyl)-4,7,10-triacetic acid
- 3,6,9,15-tetraazabicyclo[9.3.1]-pentadeca-1(15),11,13-triene-4-S-(4-isothiocyanatobenzyl)-3,6,9-triacetic acid
- CHX-A″-DTPA
- DOTA
- DOTA-DPhe1-Tyr3-octreotide
- DOTATOC
- DTPA
- HPMA
- Immuno-PET
- Monoclonal antibodies
- N-(2-hydroxypropyl)-methacrylamide
- N-[R-2-amino-3-(p-isothiocyanato-phenyl)propyl]-trans-(S,S)-cyclohexane-1,2-diamine-N,N,N′,N″,N″-pentaacetic acid
- NOTA
- Oncology
- PIB
- PIP
- Radioimmunoimaging
- SATA
- SarAr
- bispecific monoclonal antibody
- bsMAb
- diethylenetriaminepentaacetic acid
- p-SCN-Bn-PCTA
- p-SCN-Bn-oxo-DO3A
- p-iodobenzoate
- para-iodophenyl
- succinimidyl acetylthioacetate
Collapse
|
28
|
Orbay H, Zhang Y, Hong H, Hacker TA, Valdovinos HF, Zagzebski JA, Theuer CP, Barnhart TE, Cai W. Positron emission tomography imaging of angiogenesis in a murine hindlimb ischemia model with 64Cu-labeled TRC105. Mol Pharm 2013; 10:2749-56. [PMID: 23738915 DOI: 10.1021/mp400191w] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The goal of this study was to assess ischemia-induced angiogenesis with (64)Cu-NOTA-TRC105 positron emission tomography (PET) in a murine hindlimb ischemia model of peripheral artery disease (PAD). CD105 binding affinity/specificity of NOTA-conjugated TRC105 (an anti-CD105 antibody) was evaluated by flow cytometry, which exhibited no difference from unconjugated TRC105. BALB/c mice were anesthetized, and the right femoral artery was ligated to induce hindlimb ischemia, with the left hindlimb serving as an internal control. Laser Doppler imaging showed that perfusion in the ischemic hindlimb plummeted to ∼ 20% of the normal level after surgery and gradually recovered to near normal level on day 24. Ischemia-induced angiogenesis was noninvasively monitored and quantified with (64)Cu-NOTA-TRC105 PET on postoperative days 1, 3, 10, 17, and 24. (64)Cu-NOTA-TRC105 uptake in the ischemic hindlimb increased significantly from the control level of 1.6 ± 0.2 %ID/g to 14.1 ± 1.9 %ID/g at day 3 (n = 3) and gradually decreased with time (3.4 ± 1.9 %ID/g at day 24), which correlated well with biodistribution studies performed on days 3 and 24. Blocking studies confirmed the CD105 specificity of tracer uptake in the ischemic hindlimb. Increased CD105 expression on days 3 and 10 following ischemia was confirmed by histology and reverse transcription polymerase chain reaction (RT-PCR). This is the first report of PET imaging of CD105 expression during ischemia-induced angiogenesis. (64)Cu-NOTA-TRC105 PET may play multiple roles in future PAD-related research and improve PAD patient management by identifying the optimal timing of treatment and monitoring the efficacy of therapy.
Collapse
Affiliation(s)
- Hakan Orbay
- Department of Radiology, ‡Department of Medical Physics, and §Department of Medicine, Division of Cardiovascular Medicine, University of Wisconsin-Madison , Madison, Wisconsin 53706, United States
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
A novel 99mTc-labeled molecular probe for tumor angiogenesis imaging in hepatoma xenografts model: a pilot study. PLoS One 2013; 8:e61043. [PMID: 23573294 PMCID: PMC3616001 DOI: 10.1371/journal.pone.0061043] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Accepted: 03/05/2013] [Indexed: 02/07/2023] Open
Abstract
Introduction Visualization of tumor angiogenesis using radionuclide targeting provides important diagnostic information. In previous study, we proved that an arginine-arginine-leucine (RRL) peptide should be a tumor endothelial cell specific binding sequence. The overall aim of this study was to evaluate whether 99mTc-radiolabeled RRL could be noninvasively used for imaging of malignant tumors in vivo, and act as a new molecular probe targeting tumor angiogenesis. Methods The RRL peptide was designed and radiosynthesized with 99mTc by a one-step method. The radiolabeling efficiency and radiochemical purity were then characterized in vitro. 99mTc-RRL was injected intravenously in HepG2 xenograft-bearing BALB/c nude mice. Biodistribution and in vivo imaging were performed periodically. The relationship between tumor size and %ID uptake of 99mTc-RRL was also explored. Results The labeling efficiencies of 99mTc-RRL reached 76.9%±4.5% (n = 6) within 30–60 min at room temperature, and the radiochemical purity exceeded 96% after purification. In vitro stability experiment revealed the radiolabeled peptide was stable. Biodistribution data showed that 99mTc-RRL rapidly cleared from the blood and predominantly accumulated in the kidneys and tumor. The specific uptake of 99mTc-RRL in tumor was significantly higher than that of unlabeled RRL blocking and free pertechnetate control test after injection (p<0.05). The ratio of the tumor-to-muscle exceeded 6.5, tumor-to-liver reached 1.98 and tumor-to-blood reached 1.95. In planar gamma imaging study, the tumors were imaged clearly at 2–6 h after injection of 99mTc-RRL, whereas the tumor was not imaged clearly in blocking group. The tumor-to-muscle ratio of images with 99mTc-RRL was comparable with that of 18F-FDG PET images. Immunohistochemical analysis verified the excessive vasculature of tumor. There was a linear relationship between the tumor size and uptake of 99mTc-RRL with R2 = 0.821. Conclusion 99mTc-RRL can be used as a potential candidate for visualization of tumor angiogenesis in malignant carcinomas.
Collapse
|
30
|
Zhao Q, Yan P, Yin L, Li L, Chen XQ, Ma C, Wang RF. Validation study of ¹³¹I-RRL: assessment of biodistribution, SPECT imaging and radiation dosimetry in mice. Mol Med Rep 2013; 7:1355-60. [PMID: 23440460 DOI: 10.3892/mmr.2013.1338] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Accepted: 02/14/2013] [Indexed: 11/05/2022] Open
Abstract
Tumor angiogenesis is important in the growth and metastasis of malignant tumors. In our previous study, we demonstrated that an arginine-arginine-leucine (RRL) peptide is a tumor endothelial cell-specific binding sequence that may be used as a molecular probe for the imaging of malignant tumors in vivo. The aim of the present study was to further explore the characteristics of 131I‑RRL by biodistribution tests, and to estimate the radiation dosimetry of 131I‑RRL for humans using mice data. The RRL peptide was radiolabeled with 131I by a chloramine-T (CH-T) method. The radiolabeling efficiency and radiochemical purity were then characterized in vitro. 131I‑RRL was injected intravenously into B16 xenograft-bearing Kunming mice. Biodistribution analysis and in vivo imaging were performed periodically. The radiation dosimetry in humans was calculated according to the organ distribution and the standard medical internal radiation dose (MIRD) method in mice. All data were analyzed by statistical and MIRDOSE 3.1 software. The labeling efficiency of 131I‑RRL reached 70.0±2.91% (n=5), and the radiochemical purity exceeded 95% following purification. In mice bearing B16 xenografts, 131I‑RRL rapidly cleared from the blood and predominantly accumulated in the kidneys, the stomach and the tumor tissue. The specific uptake of 131I‑RRL in the tumor increased over time and was significantly higher than that of the other organs, 24-72 h following injection (P<0.05). The ratio of tumor-to-skeletal muscle (T/SM) tissue exceeded 4.75, and the ratio of the tumor-to-blood (T/B) tissue peaked at 3.36. In the single-photon emission computed tomography (SPECT) imaging of Kunming mice bearing B16 xenografts, the tumors were clearly identifiable at 6 h, and significant uptake was evident 24-72 h following administration of 131I‑RRL. The effective dose for the adult male dosimetric model was estimated to be 0.0293 mSv/MBq. Higher absorbed doses were estimated for the stomach (0.102 mGy/MBq), the small intestines (0.0699 mGy/MBq), the kidneys (0.0611 mGy/MBq) and the liver (0.055 mGy/MBq). These results highlight the potential of 131I‑RRL as a ligand for the SPECT imaging of tumors. Administration of 131I‑RRL led to a reasonable radiation dose burden and was safe for human use.
Collapse
Affiliation(s)
- Qian Zhao
- Department of Nuclear Medicine, Peking University First Hospital, West District, Beijing 100034, P.R. China
| | | | | | | | | | | | | |
Collapse
|
31
|
Orbay H, Hong H, Zhang Y, Cai W. PET/SPECT imaging of hindlimb ischemia: focusing on angiogenesis and blood flow. Angiogenesis 2012; 16:279-87. [PMID: 23117521 DOI: 10.1007/s10456-012-9319-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Accepted: 10/22/2012] [Indexed: 12/12/2022]
Abstract
Peripheral artery disease (PAD) is a result of the atherosclerotic narrowing of blood vessels to the extremities, and the subsequent tissue ischemia can lead to the up-regulation of angiogenic growth factors and formation of new vessels as a recovery mechanism. Such formation of new vessels can be evaluated with various non-invasive molecular imaging techniques, where serial images from the same subjects can be obtained to allow the documentation of disease progression and therapeutic response. The most commonly used animal model for preclinical studies of PAD is the murine hindlimb ischemia model, and a number of radiotracers have been investigated for positron emission tomography (PET) and single photon emission computed tomography (SPECT) imaging of PAD. In this review article, we summarize the PET/SPECT tracers that have been tested in the murine hindlimb ischemia model as well as those used clinically to assess the extremity blood flow.
Collapse
Affiliation(s)
- Hakan Orbay
- Department of Radiology, School of Medicine and Public Health, University of Wisconsin, Madison, 1111 Highland Ave, Room 7137, Madison, WI 53705-2275, USA
| | | | | | | |
Collapse
|