1
|
Ligeour M, Guillet SG, Maingueneau C, Croyal M, Planchat A, Gestin JF, Galland N, Guérard F. Formation Of Gold-Astatine Bonds in N-Heterocyclic Carbene Complexes. Chemistry 2025:e202500826. [PMID: 40350378 DOI: 10.1002/chem.202500826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 04/26/2025] [Accepted: 05/06/2025] [Indexed: 05/14/2025]
Abstract
With the purpose of exploring alternatives to the poorly stable astatoaryl bond found in most 211At-labeled radiopharmaceuticals, the coordination between astatine and metal-NHC (NHC = N-heterocyclic carbene) complexes was investigated. To orient this work, relativistic DFT calculations were performed, highlighting a superiority of gold(I) over rhodium(I) and iridium(I), both regarding the strength of the metal-astatine bond and the metal's preference for astatine over other halogens. The DFT calculations also revealed a more stable metal-astatine bond with electron-withdrawing NHCs. Accordingly, two Au(I) complexes, [AuCl(IPr)] (IPr = N,N'-bis[2,6-(di-isopropyl)phenyl]imidazol-2-ylidene) and [AuCl(IAd)] (IAd = N,N'-bis[adamantyl]imidazol-2-ylidene) were radiolabeled by halide exchange in excellent radiochemical yields with iodine-125 and astatine-211 in a water/acetonitrile mixture with a reducing agent. While the adamantyl-substituted complex was not investigated further due to its lack of solubility in aqueous or semi aqueous media, the stability of [Au211At(IPr)] in the presence of phosphate-buffered saline was evaluated and proved excellent (96% after two half-lives), encouraging further studies with relevant cancer-targeting vectors.
Collapse
Affiliation(s)
- Mathilde Ligeour
- CRCI2NA, Nantes Université, Inserm, CNRS, Université d'Angers, 8 quai Moncousu, Nantes Cedex, 44007, France
| | - Sébastien G Guillet
- CRCI2NA, Nantes Université, Inserm, CNRS, Université d'Angers, 8 quai Moncousu, Nantes Cedex, 44007, France
| | - Clémence Maingueneau
- CRCI2NA, Nantes Université, Inserm, CNRS, Université d'Angers, 8 quai Moncousu, Nantes Cedex, 44007, France
| | - Mikaël Croyal
- Nantes Université, CHU Nantes, Inserm, CNRS, SFR Santé, Inserm UMS 016, CNRS UMS 3556, Nantes, F-44000, France
| | | | - Jean-François Gestin
- CRCI2NA, Nantes Université, Inserm, CNRS, Université d'Angers, 8 quai Moncousu, Nantes Cedex, 44007, France
| | - Nicolas Galland
- CEISAM, CNRS, Nantes Université, UMR 6230, Nantes, 44000, France
| | - François Guérard
- CRCI2NA, Nantes Université, Inserm, CNRS, Université d'Angers, 8 quai Moncousu, Nantes Cedex, 44007, France
| |
Collapse
|
2
|
Long X, Cheng S, Lan X, Wei W, Jiang D. Trends in nanobody radiotheranostics. Eur J Nucl Med Mol Imaging 2025; 52:2225-2238. [PMID: 39800806 DOI: 10.1007/s00259-025-07077-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 01/04/2025] [Indexed: 04/23/2025]
Abstract
As the smallest antibody fragment with specific binding affinity, nanobody-based nuclear medicine has demonstrated significant potential to revolutionize the field of precision medicine, supported by burgeoning preclinical investigations and accumulating clinical evidence. However, the visualization of nanobodies has also exposed their suboptimal biodistribution patterns, which has spurred collaborative efforts to refine their pharmacokinetic and pharmacodynamic profiles for improved therapeutic efficacy. In this review, we present clinical results that exemplify the benefits of nanobody-based molecular imaging in cancer diagnosis. Moreover, we emphasize the indispensable role of molecular imaging as a tool for evaluating and optimizing nanobodies, thereby expanding their therapeutic potential in cancer treatment in the foreseeable future.
Collapse
Affiliation(s)
- Xingru Long
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave., Wuhan, Hubei, 430022, China
- Hubei Key Laboratory of Molecular Imaging, 1277 Jiefang Ave., Wuhan, Hubei, 430022, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, 1277 Jiefang Ave., Wuhan, Hubei, 430022, China
| | - Sixuan Cheng
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave., Wuhan, Hubei, 430022, China
- Hubei Key Laboratory of Molecular Imaging, 1277 Jiefang Ave., Wuhan, Hubei, 430022, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, 1277 Jiefang Ave., Wuhan, Hubei, 430022, China
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave., Wuhan, Hubei, 430022, China.
- Hubei Key Laboratory of Molecular Imaging, 1277 Jiefang Ave., Wuhan, Hubei, 430022, China.
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, 1277 Jiefang Ave., Wuhan, Hubei, 430022, China.
| | - Weijun Wei
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200233, China.
| | - Dawei Jiang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave., Wuhan, Hubei, 430022, China.
- Hubei Key Laboratory of Molecular Imaging, 1277 Jiefang Ave., Wuhan, Hubei, 430022, China.
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, 1277 Jiefang Ave., Wuhan, Hubei, 430022, China.
| |
Collapse
|
3
|
Tosato M, Favaretto C, Kleynhans J, Burgoyne AR, Gestin JF, van der Meulen NP, Jalilian A, Köster U, Asti M, Radchenko V. Alpha Atlas: Mapping global production of α-emitting radionuclides for targeted alpha therapy. Nucl Med Biol 2025; 142-143:108990. [PMID: 39809026 DOI: 10.1016/j.nucmedbio.2024.108990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 12/06/2024] [Accepted: 12/17/2024] [Indexed: 01/16/2025]
Abstract
Targeted Alpha Therapy has shown great promise in cancer treatment, sparking significant interest over recent decades. However, its broad adoption has been impeded by the scarcity of alpha-emitters and the complexities related to their use. The availability of these radionuclides is often constrained by the intricate production processes and purification, as well as regulatory and logistical challenges. Moreover, the high cost and technical difficulties associated with handling and applying alpha-emitting radionuclides pose additional barriers to their clinical implementation. This Alpha Atlas provides an in-depth overview of the leading alpha-particle emitting radionuclide candidates for clinical use, focusing on their production processes and supply chains. By mapping the current facilities that produce and supply these radionuclides, this atlas aims to assist researchers, clinicians, and industries in initiating or scaling up the applications of alpha-emitters. The Alpha Atlas aspires to act as a strategic guide, facilitating collaboration and driving forward the integration of these potent therapeutic agents into cancer treatment practices.
Collapse
Affiliation(s)
- Marianna Tosato
- Radiopharmaceutical Chemistry Laboratory (RACHEL), Nuclear Medicine Unit, AUSL-IRCCS Reggio Emilia, 42123 Reggio Emilia, Italy.
| | - Chiara Favaretto
- Radiopharmacy and Cyclotron Department, IRCCS Sacro Cuore Don Calabria, Negrar 37024, Verona, Italy
| | - Janke Kleynhans
- Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Andrew R Burgoyne
- Oak Ridge National Laboratory, 1 Bethel Valley Road, Oak Ridge, TN 37830, United States
| | - Jean-François Gestin
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, 44000 Nantes, France
| | - Nicholas P van der Meulen
- PSI Center for Life Sciences, 5232 Villigen-PSI, Switzerland; PSI Center for Nuclear Engineering and Sciences, 5232 Villigen-PSI, Switzerland
| | - Amirreza Jalilian
- Department of Nuclear Safety and Security, International Atomic Energy Agency, 1220 Vienna, Austria
| | - Ulli Köster
- Institut Laue-Langevin, 38042 Grenoble, France
| | - Mattia Asti
- Radiopharmaceutical Chemistry Laboratory (RACHEL), Nuclear Medicine Unit, AUSL-IRCCS Reggio Emilia, 42123 Reggio Emilia, Italy
| | - Valery Radchenko
- Life Sciences Division, TRIUMF, BC V6T 2A3 Vancouver, British Columbia, Canada; Department of Chemistry, University of British Columbia, V6T 1Z1 Vancouver, British Columbia, Canada
| |
Collapse
|
4
|
Biersack HJ, Rojas-Fernandez A, Ting HH, Kramer V, Juweid ME, Mottaghy FM. The promising potential of camelid nanobodies for nuclear medicine. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07136-y. [PMID: 39982493 DOI: 10.1007/s00259-025-07136-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Affiliation(s)
- Hans J Biersack
- Department of Nuclear Medicine, University of Bonn, Bonn, Germany
- Betaklinik Bonn, Bonn, Germany
| | | | - Hong-Hoi Ting
- Nanomab Technology, 720 Centennial Court, Centennial Park, Elstree, WD63SY, Borehamwood, UK
| | - Vasko Kramer
- Acrux Radiopharma Consulting SpA, Santiago, Chile
| | - Malik E Juweid
- Department of Radiology and Nuclear Medicine, University of Jordan, Amman, Jordan
| | - Felix M Mottaghy
- Department of Nuclear Medicine, University Hospital Aachen, Pauwelsstr. 30, D-52072, Aachen, Germany.
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center (MUMC+), Maastricht, The Netherlands.
| |
Collapse
|
5
|
Zuo D, Wang H, Yu B, Li Q, Gan L, Chen W. Astatine-211 and actinium-225: two promising nuclides in targeted alpha therapy. Acta Biochim Biophys Sin (Shanghai) 2024; 57:327-343. [PMID: 39587859 PMCID: PMC11986457 DOI: 10.3724/abbs.2024206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 10/22/2024] [Indexed: 11/27/2024] Open
Abstract
Nuclear medicine therapy offers a promising approach for tumor treatment, as the energy emitted during radionuclide decay causes irreparable damage to tumor cells. Notably, α-decay exhibits an even more significant destructive potential. By conjugating α-nuclides with antibodies or small-molecule inhibitors, targeted alpha therapy (TAT) can enhance tumor destruction while minimizing toxic side effects, making TAT an increasingly attractive antineoplastic strategy. Astatine-211 ( 211At) and actinium-225 ( 225Ac) have emerged as highly effective agents in TAT due to their exceptional physicochemical properties and biological effects. In this review, we highlight the applications of 211At-/ 225Ac-radiopharmaceuticals, particularly in specific tumor targets, such as prostate-specific membrane antigen (PSMA) in prostate cancers, cluster of differentiation (CD) in hematological malignancies, human epidermal growth factor receptor-2 (HER2) in ovarian cancers, and somatostatin receptor (SSTR) in neuroendocrine tumors. We synthesize the progress from preclinical and clinical trials to provide insights into the promising potential of 211At-/ 225Ac-radiopharmaceuticals for future treatments.
Collapse
Affiliation(s)
- Dashan Zuo
- Institute of Modern PhysicsChinese Academy of SciencesLanzhou730000China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in MedicineGansu ProvinceLanzhou730000China
- University of Chinese Academy of SciencesBeijing100049China
| | - Hui Wang
- Institute of Modern PhysicsChinese Academy of SciencesLanzhou730000China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in MedicineGansu ProvinceLanzhou730000China
| | - Boyi Yu
- Institute of Modern PhysicsChinese Academy of SciencesLanzhou730000China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in MedicineGansu ProvinceLanzhou730000China
| | - Qiang Li
- Institute of Modern PhysicsChinese Academy of SciencesLanzhou730000China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in MedicineGansu ProvinceLanzhou730000China
- University of Chinese Academy of SciencesBeijing100049China
- Lanhai Nuclear Medical Research CenterPutian351153China
| | - Lu Gan
- Institute of Modern PhysicsChinese Academy of SciencesLanzhou730000China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in MedicineGansu ProvinceLanzhou730000China
- University of Chinese Academy of SciencesBeijing100049China
| | - Weiqiang Chen
- Institute of Modern PhysicsChinese Academy of SciencesLanzhou730000China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in MedicineGansu ProvinceLanzhou730000China
- University of Chinese Academy of SciencesBeijing100049China
- Lanhai Nuclear Medical Research CenterPutian351153China
| |
Collapse
|
6
|
Liu X, Song Y, Cheng P, Liang B, Xing D. Targeting HER2 in solid tumors: Unveiling the structure and novel epitopes. Cancer Treat Rev 2024; 130:102826. [PMID: 39270365 DOI: 10.1016/j.ctrv.2024.102826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 09/02/2024] [Indexed: 09/15/2024]
Abstract
Human epidermal growth factor receptor-2 (HER2) is overexpressed in various solid tumor types, acting as an established therapeutic target. Over the last three decades, the fast-paced development of diverse HER2-targeted agents, notably marked by the introduction of the antibody-drug conjugate (ADC), yielding substantial improvements in survival rates. However, resistance to anti-HER2 treatments continues to pose formidable challenges. The complex structure and dynamic dimerization properties of HER2 create significant hurdles in the development of novel targeted therapeutics. In this review, we synthesize the latest insights into the structural intricacies of HER2 and present an unprecedented overview of the epitope characteristics of HER2-targeted antibodies and their derivatives. Furthermore, we delve into the correlation between anti-HER2 antibody binding epitopes and their respective functions, with a particular focus on their efficacy against resistant tumors. In addition, we highlight the potential of emerging anti-HER2 agents that target specific sites or non-overlapping epitopes, poised to transform the therapeutic landscape for HER2-positive tumors in the foreseeable future.
Collapse
Affiliation(s)
- Xinlin Liu
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Qingdao Cancer Institute, Qingdao 266071, China
| | - Yunlong Song
- Qingdao Municipal Center for Disease Control and Prevention, Qingdao Institute of Preventive Medicine, Qingdao 266033, China
| | - Panpan Cheng
- Qingdao Municipal Center for Disease Control and Prevention, Qingdao Institute of Preventive Medicine, Qingdao 266033, China
| | - Bing Liang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Qingdao Cancer Institute, Qingdao 266071, China.
| | - Dongming Xing
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Qingdao Cancer Institute, Qingdao 266071, China; School of Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
7
|
Vanermen M, Ligeour M, Oliveira MC, Gestin JF, Elvas F, Navarro L, Guérard F. Astatine-211 radiolabelling chemistry: from basics to advanced biological applications. EJNMMI Radiopharm Chem 2024; 9:69. [PMID: 39365487 PMCID: PMC11452365 DOI: 10.1186/s41181-024-00298-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 09/17/2024] [Indexed: 10/05/2024] Open
Abstract
BACKGROUND 211At-radiopharmaceuticals are currently the subject of growing studies for targeted alpha therapy of cancers, which leads to the widening of the scope of the targeting vectors, from small molecules to peptides and proteins. This has prompted, during the past decade, to a renewed interest in developing novel 211At-labelling approaches and novel prosthetic groups to address the diverse scenarios and to reach improved efficiency and robustness of procedures as well as an appropriate in vivo stability of the label. MAIN BODY Translated from the well-known (radio)iodine chemistry, the long preferred electrophilic astatodemetallation using trialkylaryltin precursors is now complemented by new approaches using electrophilic or nucleophilic At. Alternatives to the astatoaryl moiety have been proposed to improve labelling stability, and the range of prosthetic groups available to label proteins has expanded. CONCLUSION In this report, we cover the evolution of radiolabelling chemistry, from the initial strategies developed in the late 1970's to the most recent findings.
Collapse
Affiliation(s)
- Maarten Vanermen
- Molecular Imaging and Radiology (MIRA), University of Antwerp, Wilrijk, Belgium
| | - Mathilde Ligeour
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, Nantes, France
| | - Maria-Cristina Oliveira
- Departamento de Engenharia e Ciências Nucleares and Centro de Ciências e Tecnologias Nucleares, IST, Universidade de Lisboa, Estrada Nacional 10 (km 139,7), 2695-066, Bobadela LRS, Portugal
| | | | - Filipe Elvas
- Molecular Imaging and Radiology (MIRA), University of Antwerp, Wilrijk, Belgium
| | | | - François Guérard
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, Nantes, France.
| |
Collapse
|
8
|
Tada M, Kaizuka Y, Kannaka K, Suzuki H, Joho T, Takahashi K, Uehara T, Tanaka H. Development of a Neopentyl 211At-Labeled Activated Ester Providing In Vivo Stable 211At-Labeled Antibodies for Targeted Alpha Therapy. ChemMedChem 2024; 19:e202400369. [PMID: 38847493 DOI: 10.1002/cmdc.202400369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/06/2024] [Indexed: 08/10/2024]
Abstract
In this study we developed a neopentyl 211At-labeled activated ester that incorporates a triazole spacer and applied it to the synthesis of an 211At-labeled cetuximab. The activated ester was synthesized via the nucleophilic 211At-astatination of a neopentyl sulfonate carrying two long alkyl chains that serve as a lipid tag, which was followed by the hydrolysis of an acetal. Additionally, we developed a novel Resin-Assisted Purification and Deprotection (RAPD) protocol involving a solid-phase extraction of the protected 211At-labeled compound from the mixture of the labeling reaction, hydrolysis of the acetal on the resin, and finally an elution of the 211At-labeled activator from the resin. This method allows the synthesis of an 211At-labeled activated ester with high purity through a simplified procedure that circumvents the need for HPLC purification. Using this 211At-labeled activated ester, we efficiently synthesized 211At-labeled cetuximab in 27±1 % radiochemical yield with 95 % radiochemical purity. This 211At-activated ester demonstrated high reactivity, and enabled the completion of the reaction with the antibody within 10 min. In comparative biodistribution studies between 211At-labeled cetuximab and the corresponding 125I-labeled cetuximab in normal mice, both the thyroid and stomach showed radioactivity levels that were less than 1.0 % of the injected dose.
Collapse
Affiliation(s)
- Masatoshi Tada
- Department of Chemical Science and Engineering, Tokyo Institute of Technology, 12-12-1-H101 Ookayama, Meguro, Tokyo, 152-8552, Japan
| | - Yuta Kaizuka
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8675, Japan
| | - Kento Kannaka
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8675, Japan
| | - Hiroyuki Suzuki
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8675, Japan
| | - Taiki Joho
- Advanced Clinical Research Center, Fukushima Global Medical Science Center, Fukushima Medical University, 1 Hikariga-oka, Fukushima, 960-1295, Japan
| | - Kazuhiro Takahashi
- Advanced Clinical Research Center, Fukushima Global Medical Science Center, Fukushima Medical University, 1 Hikariga-oka, Fukushima, 960-1295, Japan
| | - Tomoya Uehara
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8675, Japan
| | - Hiroshi Tanaka
- Faculty of Pharmacy, Juntendo University, 6-8-1 Hinode, Urayasu, Chiba, 279-0013, Japan
- Department of Chemical Science and Engineering, Tokyo Institute of Technology, 12-12-1-H101 Ookayama, Meguro, Tokyo, 152-8552, Japan
| |
Collapse
|
9
|
Yssartier T, Liu L, Pardoue S, Le Questel JY, Guérard F, Montavon G, Galland N. In vivo stability of 211At-radiopharmaceuticals: on the impact of halogen bond formation. RSC Med Chem 2024; 15:223-233. [PMID: 38283213 PMCID: PMC10809332 DOI: 10.1039/d3md00579h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 11/22/2023] [Indexed: 01/30/2024] Open
Abstract
211At, when coupled to a targeting agent, is one of the most promising radionuclides for therapeutic applications. The main labelling approach consists in the formation of astatoaryl compounds, which often show a lack of in vivo stability. The hypothesis that halogen bond (XB) interactions with protein functional groups initiate a deastatination mechanism is investigated through radiochemical experiments and DFT modelling. Several descriptors agree on the known mechanism of iodoaryl substrates dehalogenation by iodothyronine deiodinases, supporting the higher in vivo dehalogenation of N-succinimidyl 3-[211At]astatobenzoate (SAB) conjugates in comparison with their iodinated counterparts. The guanidinium group in 3-[211At]astato-4-guanidinomethylbenzoate (SAGMB) prevents the formation of At-mediated XBs with the selenocysteine active site in iodothyronine deiodinases. The initial step of At-aryl bond dissociation is inhibited, elucidating the better in vivo stability of SAGMB conjugates compared with those of SAB. The impact of astatine's ability to form XB interactions on radiopharmaceutical degradation may not be limited to the case of aryl radiolabeling.
Collapse
Affiliation(s)
- Thibault Yssartier
- CNRS, CEISAM UMR 6230, Nantes Université F-44000 Nantes France
- CNRS, SUBATECH UMR 6457, IMT Atlantique F-44307 Nantes France
| | - Lu Liu
- CNRS, IPHC UMR 7178, Université de Strasbourg F-67037 Strasbourg France
| | - Sylvain Pardoue
- CNRS, SUBATECH UMR 6457, IMT Atlantique F-44307 Nantes France
| | | | - François Guérard
- Inserm UMR 1307, CNRS UMR 6075, CRCI2NA, Nantes Université, Université d'Angers F-44000 Nantes France
| | - Gilles Montavon
- CNRS, SUBATECH UMR 6457, IMT Atlantique F-44307 Nantes France
| | - Nicolas Galland
- CNRS, CEISAM UMR 6230, Nantes Université F-44000 Nantes France
| |
Collapse
|
10
|
Coll RP, Bright SJ, Martinus DKJ, Georgiou DK, Sawakuchi GO, Manning HC. Alpha Particle-Emitting Radiopharmaceuticals as Cancer Therapy: Biological Basis, Current Status, and Future Outlook for Therapeutics Discovery. Mol Imaging Biol 2023; 25:991-1019. [PMID: 37845582 PMCID: PMC12054971 DOI: 10.1007/s11307-023-01857-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 09/03/2023] [Accepted: 09/05/2023] [Indexed: 10/18/2023]
Abstract
Critical advances in radionuclide therapy have led to encouraging new options for cancer treatment through the pairing of clinically useful radiation-emitting radionuclides and innovative pharmaceutical discovery. Of the various subatomic particles used in therapeutic radiopharmaceuticals, alpha (α) particles show great promise owing to their relatively large size, delivered energy, finite pathlength, and resulting ionization density. This review discusses the therapeutic benefits of α-emitting radiopharmaceuticals and their pairing with appropriate diagnostics, resulting in innovative "theranostic" platforms. Herein, the current landscape of α particle-emitting radionuclides is described with an emphasis on their use in theranostic development for cancer treatment. Commonly studied radionuclides are introduced and recent efforts towards their production for research and clinical use are described. The growing popularity of these radionuclides is explained through summarizing the biological effects of α radiation on cancer cells, which include DNA damage, activation of discrete cell death programs, and downstream immune responses. Examples of efficient α-theranostic design are described with an emphasis on strategies that lead to cellular internalization and the targeting of proteins involved in therapeutic resistance. Historical barriers to the clinical deployment of α-theranostic radiopharmaceuticals are also discussed. Recent progress towards addressing these challenges is presented along with examples of incorporating α-particle therapy in pharmaceutical platforms that can be easily converted into diagnostic counterparts.
Collapse
Affiliation(s)
- Ryan P Coll
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, 1881 East Rd, Houston, TX, 77054, USA
| | - Scott J Bright
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Blvd, Houston, TX, 77030, USA
| | - David K J Martinus
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Blvd, Houston, TX, 77030, USA
| | - Dimitra K Georgiou
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, 1881 East Rd, Houston, TX, 77054, USA
| | - Gabriel O Sawakuchi
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Blvd, Houston, TX, 77030, USA
| | - H Charles Manning
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, 1881 East Rd, Houston, TX, 77054, USA.
- Cyclotron Radiochemistry Facility, The University of Texas MD Anderson Cancer Center, 1881 East Rd, Houston, TX, 77054, USA.
| |
Collapse
|
11
|
Ducharme M, Hall L, Eckenroad W, Cingoranelli SJ, Houson HA, Jaskowski L, Hunter C, Larimer BM, Lapi SE. Evaluation of [ 89Zr]Zr-DFO-2Rs15d Nanobody for Imaging of HER2-Positive Breast Cancer. Mol Pharm 2023; 20:4629-4639. [PMID: 37552575 PMCID: PMC11606513 DOI: 10.1021/acs.molpharmaceut.3c00360] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2023]
Abstract
One of the most aggressive forms of breast cancer involves the overexpression of human epidermal growth factor receptor 2 (HER2). HER2 is overexpressed in ∼25% of all breast cancers and is associated with increased proliferation, increased rates of metastasis, and poor prognosis. Treatment for HER2-positive breast cancer has vastly improved since the development of the monoclonal antibody trastuzumab (Herceptin) as well as other biological constructs. However, patients still commonly develop resistance, illustrating the need for newer therapies. Nanobodies have become an important focus for potential development as HER2-targeting imaging agents and therapeutics. Nanobodies have many favorable characteristics, including high stability in heat and nonphysiological pH, while maintaining their low-nanomolar affinity for their designed targets. Specifically, the 2Rs15d nanobody has been developed for targeting HER2 and has been evaluated as a diagnostic imaging agent for single-photon emission computed tomography (SPECT) and positron emission tomography (PET). While a construct of 2Rs15d with the positron emitter 68Ga is currently in phase I clinical trials, the only PET images acquired in preclinical or clinical research have been within 3 h postinjection. We evaluated our in-house produced 2Rs15d nanobody, conjugated with the chelator deferoxamine (DFO), and radiolabeled with 89Zr for PET imaging up to 72 h postinjection. [89Zr]Zr-DFO-2Rs15d demonstrated high stability in both phosphate-buffered saline (PBS) and human serum. Cell binding studies showed high binding and specificity for HER2, as well as prominent internalization. Our in vivo PET imaging confirmed high-quality visualization of HER2-positive tumors up to 72 h postinjection, whereas HER2-negative tumors were not visualized. Subsequent biodistribution studies quantitatively supported the significant HER2-positive tumor uptake compared to the negative control. Our studies fill an important gap in understanding the imaging and binding properties of the 2Rs15d nanobody at extended time points. As many therapeutic radioisotopes have single or multiday half-lives, this information will directly benefit the potential of the radiotherapy development of 2Rs15d for HER2-positive breast cancer patients.
Collapse
Affiliation(s)
- Maxwell Ducharme
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama 35233, United States
| | - Lucinda Hall
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama 35233, United States
| | - Whitney Eckenroad
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama 35233, United States
| | - Shelbie J Cingoranelli
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama 35233, United States
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, Alabama 35233, United States
| | - Hailey A Houson
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama 35233, United States
| | - Luke Jaskowski
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama 35233, United States
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, Alabama 35233, United States
| | - Chanelle Hunter
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama 35233, United States
| | - Benjamin M Larimer
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama 35233, United States
| | - Suzanne E Lapi
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama 35233, United States
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, Alabama 35233, United States
| |
Collapse
|
12
|
Hurley K, Cao M, Huang H, Wang Y. Targeted Alpha Therapy (TAT) with Single-Domain Antibodies (Nanobodies). Cancers (Basel) 2023; 15:3493. [PMID: 37444603 DOI: 10.3390/cancers15133493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/23/2023] [Accepted: 06/30/2023] [Indexed: 07/15/2023] Open
Abstract
The persistent threat of cancer necessitates the development of improved and more efficient therapeutic strategies that limit damage to healthy tissues. Targeted alpha therapy (TαT), a novel form of radioimmuno-therapy (RIT), utilizes a targeting vehicle, commonly antibodies, to deliver high-energy, but short-range, alpha-emitting particles specifically to cancer cells, thereby reducing toxicity to surrounding normal tissues. Although full-length antibodies are often employed as targeting vehicles for TαT, their high molecular weight and the presence of an Fc-region lead to a long blood half-life, increased bone marrow toxicity, and accumulation in other tissues such as the kidney, liver, and spleen. The discovery of single-domain antibodies (sdAbs), or nanobodies, naturally occurring in camelids and sharks, has introduced a novel antigen-specific vehicle for molecular imaging and TαT. Given that nanobodies are the smallest naturally occurring antigen-binding fragments, they exhibit shorter relative blood half-lives, enhanced tumor uptake, and equivalent or superior binding affinity and specificity. Nanobody technology could provide a viable solution for the off-target toxicity observed with full-length antibody-based TαT. Notably, the pharmacokinetic properties of nanobodies align better with the decay characteristics of many short-lived α-emitting radionuclides. This review aims to encapsulate recent advancements in the use of nanobodies as a vehicle for TαT.
Collapse
Affiliation(s)
- Kate Hurley
- Radiobiology and Health, Canadian Nuclear Laboratories, Chalk River, ON K0J 1J0, Canada
| | - Meiyun Cao
- Radiobiology and Health, Canadian Nuclear Laboratories, Chalk River, ON K0J 1J0, Canada
| | - Haiming Huang
- Research Center, Forlong Biotechnology Inc., Suzhou 215004, China
| | - Yi Wang
- Radiobiology and Health, Canadian Nuclear Laboratories, Chalk River, ON K0J 1J0, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| |
Collapse
|
13
|
Albertsson P, Bäck T, Bergmark K, Hallqvist A, Johansson M, Aneheim E, Lindegren S, Timperanza C, Smerud K, Palm S. Astatine-211 based radionuclide therapy: Current clinical trial landscape. Front Med (Lausanne) 2023; 9:1076210. [PMID: 36687417 PMCID: PMC9859440 DOI: 10.3389/fmed.2022.1076210] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 12/06/2022] [Indexed: 01/09/2023] Open
Abstract
Astatine-211 (211At) has physical properties that make it one of the top candidates for use as a radiation source for alpha particle-based radionuclide therapy, also referred to as targeted alpha therapy (TAT). Here, we summarize the main results of the completed clinical trials, further describe ongoing trials, and discuss future prospects.
Collapse
Affiliation(s)
- Per Albertsson
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden,Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden,*Correspondence: Per Albertsson ✉
| | - Tom Bäck
- Department of Radiation Physics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Karin Bergmark
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden,Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Andreas Hallqvist
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden,Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Mia Johansson
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden,Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Emma Aneheim
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden,Department of Radiation Physics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Sture Lindegren
- Department of Radiation Physics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Chiara Timperanza
- Department of Radiation Physics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Knut Smerud
- Smerud Medical Research International AS, Oslo, Norway
| | - Stig Palm
- Department of Radiation Physics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
14
|
Feng Y, Meshaw R, Zhao XG, Jannetti S, Vaidyanathan G, Zalutsky MR. Effective Treatment of Human Breast Carcinoma Xenografts with Single-Dose 211At-Labeled Anti-HER2 Single-Domain Antibody Fragment. J Nucl Med 2023; 64:124-130. [PMID: 35618478 PMCID: PMC9841253 DOI: 10.2967/jnumed.122.264071] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 05/25/2022] [Accepted: 05/25/2022] [Indexed: 01/06/2023] Open
Abstract
Single-domain antibody fragments (sdAbs) are attractive for targeted α-particle therapy, particularly with 211At, because of their rapid accumulation in tumor and clearance from normal tissues. Here, we evaluate the therapeutic potential of this strategy with 5F7 and VHH_1028-2 sdAbs that bind with high affinity to domain IV of human epidermal growth factor receptor type 2 (HER2). Methods: The HER2-specific sdAbs and HER2-irrelevant VHH_2001 were labeled using N-succinimidyl-3-211At-astato-5-guanidinomethyl benzoate (iso-211At-SAGMB). The cytotoxicity of iso- 211At-SAGMB-5F7 and iso- 211At-SAGMB-VHH_2001 were compared on HER2-expressing BT474 breast carcinoma cells. Three experiments in mice with subcutaneous BT474 xenografts were performed to evaluate the therapeutic effectiveness of single doses of iso- 211At-SAGMB-5F7 (0.7-3.0 MBq), iso- 211At-SAGMB-VHH_1028 (1.0-3.0 MBq), and iso- 211At-SAGMB-VHH_1028 and iso- 211At-SAGMB-VHH_2001 (∼1.0 MBq). Results: Clonogenic survival of BT474 cells was reduced after exposure to iso- 211At-SAGMB-5F7 (D0 = 1.313 kBq/mL) whereas iso- 211At-SAGMB-VHH_2001 was ineffective. Dose-dependent tumor growth inhibition was observed with 211At-labeled HER2-specific 5F7 and VHH_1028 but not with HER2-irrelevant VHH_2001. At the 3.0-MBq dose, complete tumor regression was seen in 3 of 4 mice treated with iso- 211At-SAGMB-5F7 and 8 of 11 mice treated with iso- 211At-SAGMB-VHH_1028; prolongation in median survival was 495% and 414%, respectively. Conclusion: Combining rapidly internalizing, high-affinity HER2-targeted sdAbs with the iso- 211At-SAGMB residualizing prosthetic agent is a promising strategy for targeted α-particle therapy of HER2-expressing cancers.
Collapse
Affiliation(s)
- Yutian Feng
- Department of Radiology, Duke University Medical Center, Durham, North Carolina
| | - Rebecca Meshaw
- Department of Radiology, Duke University Medical Center, Durham, North Carolina
| | - Xiao-Guang Zhao
- Department of Radiology, Duke University Medical Center, Durham, North Carolina
| | - Stephen Jannetti
- Department of Radiology, Duke University Medical Center, Durham, North Carolina
| | | | - Michael R Zalutsky
- Department of Radiology, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
15
|
Qin X, Guo X, Liu T, Li L, Zhou N, Ma X, Meng X, Liu J, Zhu H, Jia B, Yang Z. High in-vivo stability in preclinical and first-in-human experiments with [ 18F]AlF-RESCA-MIRC213: a 18F-labeled nanobody as PET radiotracer for diagnosis of HER2-positive cancers. Eur J Nucl Med Mol Imaging 2023; 50:302-313. [PMID: 36129493 DOI: 10.1007/s00259-022-05967-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 09/11/2022] [Indexed: 01/10/2023]
Abstract
PURPOSE [18F]AlF-RESCA was introduced as a core particularly useful for 18F-labeling of heat-sensitive biomolecules. However, no translational studies have been reported up to now. Herein, we reported the first-in-human evaluation of an 18F-labeled anti-HER2 nanobody MIRC213 as a PET radiotracer for imaging HER2-positive cancers. METHODS MIRC213 was produced by E. coli and conjugated with ( ±)-H3RESCA-Mal. [18F]AlF-RESCA-MIRC213 was prepared at room temperature. Its radiochemical purity and stability of were determined by radio-HPLC with the size-exclusion chromatographic column. Cell uptake was performed in NCI-N87 (HER2 +) and MCF-7 (HER2-) cells and the cell-binding affinity was verified in SK-OV-3 (HER2 +) cells. Small-animal PET/CT was performed using SK-OV-3, NCI-N87, and MCF-7 tumor-bearing mice at 30 min, 1 h, and 2 h post-injection. For blocking experiment, excess MIRC213 was co-injected with radiotracer. Biodistribution were performed on SKOV-3 and MCF-7 tumor-bearing mice at 2 h post-injection. For clinical study, PET/CT images were acquired at 2 h and 4 h after injection of [18F]AlF-RESCA-MIRC213 (1.85-3.7 MBq/kg) in six breast cancer patients (3 HER2-positive and 3 HER2-negative). All patients underwent [18F]-FDG PET/CT within a week for tissue selection purpose. Distribution and dosimetry were calculated. Standardized uptake values (SUV) were measured in tumors and normal organs. RESULTS MIRC213 was produced with > 95% purity and modified with RESCA to obtain RESCA-MIRC213. [18F]AlF-RESCA-MIRC213 was prepared within 20 min at room temperature with the radiochemical yield of 50.48 ± 7.6% and radiochemical purity of > 98% (n > 10), and remained stable in both PBS (88%) and 5% HSA (92%) after 6 h. The 2 h cellular uptake of [18F]AlF-RESCA-MIRC213 in NCI-N87 cells was 11.22 ± 0.60 AD%/105 cells. Its binding affinity Kd value was determined to be 1.23 ± 0.58 nM. Small-animal PET/CT with [18F]AlF-RESCA-MIRC213 can clearly differentiate SK-OV-3 and NCI-N87 tumors from MCF-7 tumors and background with a high uptake of 4.73 ± 1.18 ID%/g and substantially reduced to 1.70 ± 0.13 ID%/g for the blocking group (p < 0.05) in SK-OV-3 tumors at 2 h post-injection. No significant bone radioactivity was seen in the tumor-bearing animals. In all six breast cancer patients, there was no adverse reaction during study. The uptake of [18F]AlF-RESCA-MIRC213 was mainly in lacrimal gland, parotid gland, submandibular gland, thyroid gland, gallbladder, kidneys, liver, and intestines. There was no significant bone radioactivity accumulation in cancer patients. [18F]AlF-RESCA-MIRC213 had significantly higher tumor uptake in lesions from HER2-positive patients than that lesions from HER2-negative patients (SUVmax of 3.62 ± 1.56 vs. 1.41 ± 0.41, p = 0.0012) at 2 h post-injection. The kidneys received the highest radiation dose of 2.42 × 10-1 mGy/MBq, and the effective dose was 1.56 × 10-2 mSv/MBq. CONCLUSIONS [18F]AlF-RESCA-MIRC213 could be prepared with high radiolabeling yield under mild conditions. [18F]AlF-RESCA-MIRC213 has relatively high stability both in vitro and in vivo. The results from clinical transformation suggest that [18F]AlF-RESCA-MIRC213 PET/CT is a safe procedure with favorable pharmacokinetics and dosimetry profile, and it is a promising new PET radiotracer for noninvasive diagnosis of HER2-positive cancers.
Collapse
Affiliation(s)
- Xue Qin
- Guizhou University School of Medicine, Guizhou University, Guiyang, 550025, China
- Key Laboratory of Carcinogenesis and Translational Research, (Ministry of Education/Beijing), Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Xiaoyi Guo
- Key Laboratory of Carcinogenesis and Translational Research, (Ministry of Education/Beijing), Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Tianyu Liu
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Liqiang Li
- Key Laboratory of Carcinogenesis and Translational Research, (Ministry of Education/Beijing), Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Nina Zhou
- Key Laboratory of Carcinogenesis and Translational Research, (Ministry of Education/Beijing), Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Xiaopan Ma
- Guizhou University School of Medicine, Guizhou University, Guiyang, 550025, China
- Key Laboratory of Carcinogenesis and Translational Research, (Ministry of Education/Beijing), Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Xiangxi Meng
- Key Laboratory of Carcinogenesis and Translational Research, (Ministry of Education/Beijing), Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Jiayue Liu
- Key Laboratory of Carcinogenesis and Translational Research, (Ministry of Education/Beijing), Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Hua Zhu
- Guizhou University School of Medicine, Guizhou University, Guiyang, 550025, China.
- Key Laboratory of Carcinogenesis and Translational Research, (Ministry of Education/Beijing), Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| | - Bing Jia
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing, 100191, China.
| | - Zhi Yang
- Guizhou University School of Medicine, Guizhou University, Guiyang, 550025, China.
- Key Laboratory of Carcinogenesis and Translational Research, (Ministry of Education/Beijing), Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| |
Collapse
|
16
|
Takashima H, Ohnuki K, Manabe S, Koga Y, Tsumura R, Anzai T, Wang Y, Yin X, Sato N, Shigekawa Y, Nambu A, Usuda S, Haba H, Fujii H, Yasunaga M. Tumor Targeting of 211At-Labeled Antibody under Sodium Ascorbate Protection against Radiolysis. Mol Pharm 2022; 20:1156-1167. [PMID: 36573995 PMCID: PMC9906747 DOI: 10.1021/acs.molpharmaceut.2c00869] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Astatine-211 (211At) is an alpha emitter applicable to radioimmunotherapy (RIT), a cancer treatment that utilizes radioactive antibodies to target tumors. In the preparation of 211At-labeled monoclonal antibodies (211At-mAbs), the possibility of radionuclide-induced antibody denaturation (radiolysis) is of concern. Our previous study showed that this 211At-induced radiochemical reaction disrupts the cellular binding activity of an astatinated mAb, resulting in attenuation of in vivo antitumor effects, whereas sodium ascorbate (SA), a free radical scavenger, prevents antibody denaturation, contributing to the maintenance of binding and antitumor activity. However, the influence of antibody denaturation on the pharmacokinetics of 211At-mAbs relating to tumor accumulation, blood circulation time, and distribution to normal organs remains unclear. In this study, we use a radioactive anti-human epidermal growth factor receptor 2 (anti-HER2) mAb to demonstrate that an 211At-induced radiochemical reaction disrupts active targeting via an antigen-antibody interaction, whereas SA helps to maintain targeting. In contrast, there was no difference in blood circulation time as well as distribution to normal organs between the stabilized and denatured immunoconjugates, indicating that antibody denaturation may not affect tumor accumulation via passive targeting based on the enhanced permeability and retention effect. In a high-HER2-expressing xenograft model treated with 1 MBq of 211At-anti-HER2 mAbs, SA-dependent maintenance of active targeting contributed to a significantly better response. In treatment with 0.5 or 0.2 MBq, the stabilized radioactive mAb significantly reduced tumor growth compared to the denatured immunoconjugate. Additionally, through a comparison between a stabilized 211At-anti-HER2 mAb and radioactive nontargeted control mAb, we demonstrate that active targeting significantly enhances tumor accumulation of radioactivity and in vivo antitumor effect. In RIT with 211At, active targeting contributes to efficient tumor accumulation of radioactivity, resulting in a potent antitumor effect. SA-dependent protection that successfully maintains tumor targeting will facilitate the clinical application of alpha-RIT.
Collapse
Affiliation(s)
- Hiroki Takashima
- Division
of Developmental Therapeutics, Exploratory
Oncology Research & Clinical Trial Center, National Cancer Center, 6-5-1 Kashiwanoha, Kashiwa, Chiba 277-8577, Japan
| | - Kazunobu Ohnuki
- Division
of Functional Imaging, Exploratory Oncology
Research & Clinical Trial Center, National Cancer Center, 6-5-1 Kashiwanoha, Kashiwa, Chiba 277-8577, Japan
| | - Shino Manabe
- Laboratory
of Functional Molecule Chemistry, Pharmaceutical Department and Institute
of Medicinal Chemistry, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo 142-8501, Japan,Research
Center for Pharmaceutical Development, Graduate School of Pharmaceutical
Sciences & Faculty of Pharmaceutical Sciences, Tohoku University, 6-3
Aoba, Aramaki, Aoba-ku, Sendai 980-8578, Japan,Glycometabolic
Biochemistry Laboratory, RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Yoshikatsu Koga
- Division
of Developmental Therapeutics, Exploratory
Oncology Research & Clinical Trial Center, National Cancer Center, 6-5-1 Kashiwanoha, Kashiwa, Chiba 277-8577, Japan,Department
of Strategic Programs, Exploratory Oncology
Research & Clinical Trial Center, National Cancer Center, 6-5-1 Kashiwanoha, Kashiwa, Chiba 277-8577, Japan
| | - Ryo Tsumura
- Division
of Developmental Therapeutics, Exploratory
Oncology Research & Clinical Trial Center, National Cancer Center, 6-5-1 Kashiwanoha, Kashiwa, Chiba 277-8577, Japan
| | - Takahiro Anzai
- Division
of Developmental Therapeutics, Exploratory
Oncology Research & Clinical Trial Center, National Cancer Center, 6-5-1 Kashiwanoha, Kashiwa, Chiba 277-8577, Japan
| | - Yang Wang
- Nishina
Center for Accelerator-Based Science, RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Xiaojie Yin
- Nishina
Center for Accelerator-Based Science, RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Nozomi Sato
- Nishina
Center for Accelerator-Based Science, RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Yudai Shigekawa
- Nishina
Center for Accelerator-Based Science, RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Akihiro Nambu
- Nishina
Center for Accelerator-Based Science, RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Sachiko Usuda
- Nishina
Center for Accelerator-Based Science, RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Hiromitsu Haba
- Nishina
Center for Accelerator-Based Science, RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Hirofumi Fujii
- Division
of Functional Imaging, Exploratory Oncology
Research & Clinical Trial Center, National Cancer Center, 6-5-1 Kashiwanoha, Kashiwa, Chiba 277-8577, Japan
| | - Masahiro Yasunaga
- Division
of Developmental Therapeutics, Exploratory
Oncology Research & Clinical Trial Center, National Cancer Center, 6-5-1 Kashiwanoha, Kashiwa, Chiba 277-8577, Japan,Tel.: +81-4-7134-6857. Fax: +81-4-7134-6866.
| |
Collapse
|
17
|
Rodak M, Dekempeneer Y, Wojewódzka M, Caveliers V, Covens P, Miller BW, Sevenois MB, Bruchertseifer F, Morgenstern A, Lahoutte T, D'Huyvetter M, Pruszyński M. Preclinical Evaluation of 225Ac-Labeled Single-Domain Antibody for the Treatment of HER2pos Cancer. Mol Cancer Ther 2022; 21:1835-1845. [PMID: 36129807 PMCID: PMC9716241 DOI: 10.1158/1535-7163.mct-21-1021] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 05/25/2022] [Accepted: 09/16/2022] [Indexed: 01/12/2023]
Abstract
Human epidermal growth factor receptor type 2 (HER2) is overexpressed in various cancers; thus, HER2-targeting single-domain antibodies (sdAb) could offer a useful platform for radioimmunotherapy. In this study, we optimized the labeling of an anti-HER2-sdAb with the α-particle-emitter 225Ac through a DOTA-derivative. The formed radioconjugate was tested for binding affinity, specificity and internalization properties, whereas cytotoxicity was evaluated by clonogenic and DNA double-strand-breaks assays. Biodistribution studies were performed in mice bearing subcutaneous HER2pos tumors to estimate absorbed doses delivered to organs and tissues. Therapeutic efficacy and potential toxicity were assessed in HER2pos intraperitoneal ovarian cancer model and in healthy C57Bl/6 mice. [225Ac]Ac-DOTA-2Rs15d exhibited specific cell uptake and cell-killing capacity in HER2pos cells (EC50 = 3.9 ± 1.1 kBq/mL). Uptake in HER2pos lesions peaked at 3 hours (9.64 ± 1.69% IA/g), with very low accumulation in other organs (<1% IA/g) except for kidneys (11.69 ± 1.10% IA/g). α-camera imaging presented homogeneous uptake of radioactivity in tumors, although heterogeneous in kidneys, with a higher signal density in cortex versus medulla. In mice with HER2pos disseminated tumors, repeated administration of [225Ac]Ac-DOTA-2Rs15d significantly prolonged survival (143 days) compared to control groups (56 and 61 days) and to the group treated with HER2-targeting mAb trastuzumab (100 days). Histopathologic evaluation revealed signs of kidney toxicity after repeated administration of [225Ac]Ac-DOTA-2Rs15d. [225Ac]Ac-DOTA-2Rs15d efficiently targeted HER2pos cells and was effective in treatment of intraperitoneal disseminated tumors, both alone and as an add-on combination with trastuzumab, albeit with substantial signs of inflammation in kidneys. This study warrants further development of [225Ac]Ac-DOTA-2Rs15d.
Collapse
Affiliation(s)
- Magdalena Rodak
- Institute of Nuclear Chemistry and Technology, Warsaw, Poland
| | - Yana Dekempeneer
- Department of Medical Imaging, In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, Brussels, Belgium
| | | | - Vicky Caveliers
- Department of Medical Imaging, In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, Brussels, Belgium
- Nuclear Medicine Department, UZ Brussel, Brussels, Belgium
| | - Peter Covens
- Department of Medical Imaging, In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, Brussels, Belgium
| | - Brian W. Miller
- Department of Medical Imaging, University of Arizona, Tucson, Arizona
| | - Matthijs B. Sevenois
- Department of Medical Imaging, In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, Brussels, Belgium
| | | | | | - Tony Lahoutte
- Department of Medical Imaging, In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, Brussels, Belgium
- Nuclear Medicine Department, UZ Brussel, Brussels, Belgium
| | - Matthias D'Huyvetter
- Department of Medical Imaging, In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, Brussels, Belgium
| | - Marek Pruszyński
- Institute of Nuclear Chemistry and Technology, Warsaw, Poland
- NOMATEN Centre of Excellence, National Centre for Nuclear Research, Otwock, Poland
| |
Collapse
|
18
|
Feng Y, Sarrett SM, Meshaw RL, Vaidyanathan G, Cornejo MA, Zeglis BM, Zalutsky MR. Site-Specific Radiohalogenation of a HER2-Targeted Single-Domain Antibody Fragment Using a Novel Residualizing Prosthetic Agent. J Med Chem 2022; 65:15358-15373. [PMID: 36368007 DOI: 10.1021/acs.jmedchem.2c01331] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Because of their rapid tumor accumulation and normal tissue clearance, single-domain antibody fragments (sdAbs) are an attractive vehicle for developing radiotherapeutics labeled with the α-emitter 211At. Herein, we have evaluated iso-[211At]AGMB-PODS, a prosthetic agent that combines a functionality for residualizing radiohalogens with a phenyloxadiazolyl methylsulfone (PODS) moiety for site-specific sdAb conjugation. Iso-[211At]AGMB-PODS and its radioiodinated analogue were evaluated for thiol-selective conjugation to anti-HER2 5F7 sdAb bearing a C-terminus GGC tail. Both radiohalogenated PODS-5F7GGC conjugates were synthesized in good radiochemical yields and retained high binding affinity on HER2-positive BT474 breast carcinoma cells. Iso-[211At]AGMB-PODS-5F7GGC was considerably more stable in vitro than its maleimide analogue in the presence of cysteine and human serum albumin (HSA) and exhibited excellent tumor uptake and high in vivo stability. Superior tumor-to-kidney activity ratios were seen for both radiohalogenated PODS-5F7GGC conjugates compared with [177Lu]Lu-DOTA-PODS-5F7GGC. These results suggest that iso-[211At]AGMB-PODS-5F7GGC warrants further evaluation for the treatment of HER2-expressing malignancies.
Collapse
Affiliation(s)
- Yutian Feng
- Department of Radiology, Duke University Medical Center, Durham, North Carolina 27710, United States
| | - Samantha M. Sarrett
- Hunter College, City University of New York, New York, New York 10021, United States
- Ph.D. Programs in Biochemistry and Chemistry, The Graduate Center, City University of New York, New York, New York 10021, United States
| | - Rebecca L. Meshaw
- Department of Radiology, Duke University Medical Center, Durham, North Carolina 27710, United States
| | - Ganesan Vaidyanathan
- Department of Radiology, Duke University Medical Center, Durham, North Carolina 27710, United States
| | - Mike A. Cornejo
- Hunter College, City University of New York, New York, New York 10021, United States
- Ph.D. Programs in Biochemistry and Chemistry, The Graduate Center, City University of New York, New York, New York 10021, United States
| | - Brian M. Zeglis
- Hunter College, City University of New York, New York, New York 10021, United States
- Ph.D. Programs in Biochemistry and Chemistry, The Graduate Center, City University of New York, New York, New York 10021, United States
| | - Michael R. Zalutsky
- Department of Radiology, Duke University Medical Center, Durham, North Carolina 27710, United States
| |
Collapse
|
19
|
Uccelli L, Martini P, Urso L, Ghirardi T, Marvelli L, Cittanti C, Carnevale A, Giganti M, Bartolomei M, Boschi A. Rhenium Radioisotopes for Medicine, a Focus on Production and Applications. Molecules 2022; 27:5283. [PMID: 36014521 PMCID: PMC9412410 DOI: 10.3390/molecules27165283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/08/2022] [Accepted: 08/17/2022] [Indexed: 11/28/2022] Open
Abstract
In recent decades, the use of alpha; pure beta; or beta/gamma emitters in oncology, endocrinology, and interventional cardiology rheumatology, has proved to be an important alternative to the most common therapeutic regimens. Among radionuclides used for therapy in nuclear medicine, two rhenium radioisotopes are of particular relevance: rhenium-186 and rhenium-188. The first is routinely produced in nuclear reactors by direct neutron activation of rhenium-186 via 185Re(n,γ)186Re nuclear reaction. Rhenium-188 is produced by the decay of the parent tungsten-188. Separation of rhenium-188 is mainly performed using a chromatographic 188W/188Re generator in which tungsten-188 is adsorbed on the alumina column, similar to the 99Mo/99mTc generator system, and the radionuclide eluted in saline solution. The application of rhenium-186 and rhenium-188 depends on their specific activity. Rhenium-186 is produced in low specific activity and is mainly used for labeling particles or diphosphonates for bone pain palliation. Whereas, rhenium-188 of high specific activity can be used for labeling peptides or bioactive molecules. One of the advantages of rhenium is its chemical similarity with technetium. So, diagnostic technetium analogs labeled with radiorhenium can be developed for therapeutic applications. Clinical trials promoting the use of 186/188Re-radiopharmaceuticals is, in particular, are discussed.
Collapse
Affiliation(s)
- Licia Uccelli
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
- Nuclear Medicine Unit, University Hospital, 44124 Ferrara, Italy
| | - Petra Martini
- Department of Environmental and Prevention Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Luca Urso
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
- Nuclear Medicine Unit, University Hospital, 44124 Ferrara, Italy
| | - Teresa Ghirardi
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Lorenza Marvelli
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Corrado Cittanti
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
- Nuclear Medicine Unit, University Hospital, 44124 Ferrara, Italy
| | - Aldo Carnevale
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
- Radiology Unit, University Hospital, 44124 Ferrara, Italy
| | - Melchiore Giganti
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
- Radiology Unit, University Hospital, 44124 Ferrara, Italy
| | - Mirco Bartolomei
- Nuclear Medicine Unit, University Hospital, 44124 Ferrara, Italy
| | - Alessandra Boschi
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
20
|
Caers J, Duray E, Vrancken L, Marcion G, Bocuzzi V, De Veirman K, Krasniqi A, Lejeune M, Withofs N, Devoogdt N, Dumoulin M, Karlström AE, D’Huyvetter M. Radiotheranostic Agents in Hematological Malignancies. Front Immunol 2022; 13:911080. [PMID: 35865548 PMCID: PMC9294596 DOI: 10.3389/fimmu.2022.911080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 06/06/2022] [Indexed: 12/23/2022] Open
Abstract
Radioimmunotherapy (RIT) is a cancer treatment that combines radiation therapy with tumor-directed monoclonal antibodies (Abs). Although RIT had been introduced for the treatment of CD20 positive non-Hodgkin lymphoma decades ago, it never found a broad clinical application. In recent years, researchers have developed theranostic agents based on Ab fragments or small Ab mimetics such as peptides, affibodies or single-chain Abs with improved tumor-targeting capacities. Theranostics combine diagnostic and therapeutic capabilities into a single pharmaceutical agent; this dual application can be easily achieved after conjugation to radionuclides. The past decade has seen a trend to increased specificity, fastened pharmacokinetics, and personalized medicine. In this review, we discuss the different strategies introduced for the noninvasive detection and treatment of hematological malignancies by radiopharmaceuticals. We also discuss the future applications of these radiotheranostic agents.
Collapse
Affiliation(s)
- Jo Caers
- Laboratory of Hematology, GIGA I³, University of Liège, Liège, Belgium
- Department of Hematology, CHU de Liège, Liège, Belgium
- *Correspondence: Jo Caers,
| | - Elodie Duray
- Laboratory of Hematology, GIGA I³, University of Liège, Liège, Belgium
- Centre for Protein Engineering, Inbios, University of Liège, Liège, Belgium
| | - Louise Vrancken
- Laboratory of Hematology, GIGA I³, University of Liège, Liège, Belgium
- Department of Hematology, CHU de Liège, Liège, Belgium
| | - Guillaume Marcion
- Laboratory of Hematology, GIGA I³, University of Liège, Liège, Belgium
| | - Valentina Bocuzzi
- Laboratory of Hematology, GIGA I³, University of Liège, Liège, Belgium
| | - Kim De Veirman
- Department of Hematology and Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Ahmet Krasniqi
- Laboratory of In Vivo Cellular and Molecular Imaging Laboratory (ICMI), Vrije Universiteit Brussel, Brussels, Belgium
| | - Margaux Lejeune
- Laboratory of Hematology, GIGA I³, University of Liège, Liège, Belgium
| | - Nadia Withofs
- Department of Nuclear Medicine, CHU de Liège, Liège, Belgium
| | - Nick Devoogdt
- Laboratory of In Vivo Cellular and Molecular Imaging Laboratory (ICMI), Vrije Universiteit Brussel, Brussels, Belgium
| | - Mireille Dumoulin
- Centre for Protein Engineering, Inbios, University of Liège, Liège, Belgium
| | - Amelie Eriksson Karlström
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Matthias D’Huyvetter
- Laboratory of In Vivo Cellular and Molecular Imaging Laboratory (ICMI), Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
21
|
Awad RM, Meeus F, Ceuppens H, Ertveldt T, Hanssens H, Lecocq Q, Mateusiak L, Zeven K, Valenta H, De Groof TWM, De Vlaeminck Y, Krasniqi A, De Veirman K, Goyvaerts C, D'Huyvetter M, Hernot S, Devoogdt N, Breckpot K. Emerging applications of nanobodies in cancer therapy. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 369:143-199. [PMID: 35777863 DOI: 10.1016/bs.ircmb.2022.03.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Cancer is a heterogeneous disease, requiring treatment tailored to the unique phenotype of the patient's tumor. Monoclonal antibodies (mAbs) and variants thereof have enabled targeted therapies to selectively target cancer cells. Cancer cell-specific mAbs have been used for image-guided surgery and targeted delivery of radionuclides or toxic agents, improving classical treatment strategies. Cancer cell-specific mAbs can further inhibit tumor cell growth or can stimulate immune-mediated destruction of cancer cells, a feature that has also been achieved through mAb-mediated manipulation of immune cells and pathways. Drawbacks of mAbs and their variants, together with the discovery of camelid heavy chain-only antibodies and the many advantageous features of their variable domains, referred to as VHHs, single domain antibodies or nanobodies (Nbs), resulted in the exploration of Nbs as an alternative targeting moiety. We therefore review the state-of-the-art as well as novel exploitation strategies of Nbs for targeted cancer therapy.
Collapse
Affiliation(s)
- Robin Maximilian Awad
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Fien Meeus
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Hannelore Ceuppens
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Thomas Ertveldt
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Heleen Hanssens
- In Vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| | - Quentin Lecocq
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Lukasz Mateusiak
- In Vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| | - Katty Zeven
- In Vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| | - Hana Valenta
- Lab for Nanobiology, Department of Chemistry, KU Leuven, Leuven, Belgium
| | - Timo W M De Groof
- In Vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| | - Yannick De Vlaeminck
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Ahmet Krasniqi
- In Vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| | - Kim De Veirman
- Laboratory for Hematology and Immunology, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Cleo Goyvaerts
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Matthias D'Huyvetter
- In Vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| | - Sophie Hernot
- In Vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| | - Nick Devoogdt
- In Vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| | - Karine Breckpot
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
22
|
Shoari A, Tahmasebi M, Khodabakhsh F, Cohan RA, Oghalaie A, Behdani M. Angiogenic biomolecules specific nanobodies application in cancer imaging and therapy; review and updates. Int Immunopharmacol 2022; 105:108585. [DOI: 10.1016/j.intimp.2022.108585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/18/2022] [Accepted: 01/25/2022] [Indexed: 11/05/2022]
|
23
|
Recent progress of astatine-211 in endoradiotherapy: Great advances from fundamental properties to targeted radiopharmaceuticals. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.03.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
24
|
Feng Y, Meshaw R, McDougald D, Zhou Z, Zhao XG, Jannetti SA, Reiman RE, Pippen E, Marjoram R, Schaal JL, Vaidyanathan G, Zalutsky MR. Evaluation of an 131I-labeled HER2-specific single domain antibody fragment for the radiopharmaceutical therapy of HER2-expressing cancers. Sci Rep 2022; 12:3020. [PMID: 35194100 PMCID: PMC8864007 DOI: 10.1038/s41598-022-07006-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 02/09/2022] [Indexed: 11/17/2022] Open
Abstract
Radiopharmaceutical therapy (RPT) is an attractive strategy for treatment of disseminated cancers including those overexpressing the HER2 receptor including breast, ovarian and gastroesophageal carcinomas. Single-domain antibody fragments (sdAbs) exemplified by the HER2-targeted VHH_1028 evaluated herein are attractive for RPT because they rapidly accumulate in tumor and clear faster from normal tissues than intact antibodies. In this study, VHH_1028 was labeled using the residualizing prosthetic agent N-succinimidyl 3-guanidinomethyl 5-[131I]iodobenzoate (iso-[131I]SGMIB) and its tissue distribution evaluated in the HER2-expressing SKOV-3 ovarian and BT474 breast carcinoma xenograft models. In head-to-head comparisons to [131I]SGMIB-2Rs15d, a HER2-targeted radiopharmaceutical currently under clinical investigation, iso-[131I]SGMIB-VHH_1028 exhibited significantly higher tumor uptake and significantly lower kidney accumulation. The results demonstrated 2.9 and 6.3 times more favorable tumor-to-kidney radiation dose ratios in the SKOV-3 and BT474 xenograft models, respectively. Iso-[131I]SGMIB-VHH_1028 was prepared using a solid-phase extraction method for purification of the prosthetic agent intermediate Boc2-iso-[131I]SGMIB that reproducibly scaled to therapeutic-level doses and obviated the need for its HPLC purification. Single-dose (SKOV-3) and multiple-dose (BT474) treatment regimens demonstrated that iso-[131I]SGMIB-VHH_1028 was well tolerated and provided significant tumor growth delay and survival prolongation. This study suggests that iso-[131I]SGMIB-VHH_1028 is a promising candidate for RPT of HER2-expressing cancers and further development is warranted.
Collapse
Affiliation(s)
- Yutian Feng
- Department of Radiology, Duke University Medical Center, Durham, NC, USA
| | - Rebecca Meshaw
- Department of Radiology, Duke University Medical Center, Durham, NC, USA
| | - Darryl McDougald
- Department of Radiology, Duke University Medical Center, Durham, NC, USA.,Cereius Inc, Durham, NC, USA
| | - Zhengyuan Zhou
- Department of Radiology, Duke University Medical Center, Durham, NC, USA
| | - Xiao-Guang Zhao
- Department of Radiology, Duke University Medical Center, Durham, NC, USA
| | - Stephen A Jannetti
- Department of Radiology, Duke University Medical Center, Durham, NC, USA
| | - Robert E Reiman
- Department of Radiology, Duke University Medical Center, Durham, NC, USA
| | | | | | | | | | - Michael R Zalutsky
- Department of Radiology, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
25
|
Radiolabeling chemistry with heavy halogens iodine and astatine. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00013-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
26
|
Radiobiology of Targeted Alpha Therapy. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00093-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
27
|
Liu Y, Zhou Z, Feng Y, Zhao XG, Vaidyanathan G, Zalutsky MR, Vo-Dinh T. Gold Nanostars: A Novel Platform for Developing 211At-Labeled Agents for Targeted Alpha-Particle Therapy. Int J Nanomedicine 2021; 16:7297-7305. [PMID: 34737567 PMCID: PMC8560129 DOI: 10.2147/ijn.s327577] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 08/28/2021] [Indexed: 12/20/2022] Open
Abstract
Aim To develop an innovative 211At nanoplatform with high radiolabeling efficiency and low in vivo deastatination for future targeted alpha-particle therapy (TAT) to treat cancer. Methods Star-shaped gold nanoparticles, gold nanostars (GNS), were used as the platform for 211At radiolabeling. Radiolabeling efficiency under different reaction conditions was tested. Uptake in the thyroid and stomach after systemic administration was used to evaluate the in vivo stability of 211At-labeled GNS. A subcutaneous U87MG human glioma xenograft murine model was used to preliminarily evaluate the therapeutic efficacy of 211At-labeled GNS after intratumoral administration. Results The efficiency of labeling GNS with 211At was almost 100% using a simple and rapid synthesis process that was completed in only 1 min. In vitro stability test in serum showed that more than 99% of the 211At activity remained on the GNS after 24 h incubation at 37°C. In vivo biodistribution results showed low uptake in the thyroid (0.44–0.64%ID) and stomach (0.21–0.49%ID) between 0.5 and 21 h after intravenous injection, thus indicating excellent in vivo stability of 211At-labeled GNS. The preliminary therapeutic efficacy study demonstrated that 211At labeled GNS substantially reduced tumor growth (P < 0.001; two-way ANOVA) after intratumoral administration. Conclusion The new 211At radiolabeling strategy based on GNS has the advantages of a simple process, high labeling efficiency, and minimal in vivo dissociation, making it an attractive potential platform for developing TAT agents that warrants further evaluation in future preclinical studies directed to evaluating prospects for clinical translation.
Collapse
Affiliation(s)
- Yang Liu
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Zhengyuan Zhou
- Department of Radiology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Yutian Feng
- Department of Radiology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Xiao-Guang Zhao
- Department of Radiology, Duke University Medical Center, Durham, NC, 27710, USA
| | | | - Michael R Zalutsky
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA.,Department of Radiology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Tuan Vo-Dinh
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA.,Department of Chemistry, Duke University, Durham, NC, 27708, USA.,Fitzpatrick Institute for Photonics, Duke University, Durham, NC, 27708, USA
| |
Collapse
|
28
|
Feng Y, Zalutsky MR. Production, purification and availability of 211At: Near term steps towards global access. Nucl Med Biol 2021; 100-101:12-23. [PMID: 34144505 PMCID: PMC8448941 DOI: 10.1016/j.nucmedbio.2021.05.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 05/28/2021] [Accepted: 05/31/2021] [Indexed: 10/21/2022]
Abstract
The promising characteristics of the 7.2-h radiohalogen 211At have long been recognized; including having chemical properties suitable for labeling targeting vectors ranging from small organic molecules to proteins, and the emission of only one α-particle per decay, providing greater control over off-target effects. Unfortunately, the impact of 211At within the targeted α-particle therapy domain has been constrained by its limited availability. Paradoxically, the most commonly used production method - via the 209Bi(α,2n)211At reaction - utilizes a widely available natural material (bismuth) as the target and straightforward cyclotron irradiation methodology. On the other hand, the most significant impediment to widespread 211At availability is the need for an accelerator capable of generating ≥28 MeV α-particles with sufficient beam intensities to make clinically relevant levels of 211At. In this review, current methodologies for the production and purification of 211At - both by the direct production route noted above and via a 211Rn generator system - will be discussed. The capabilities of cyclotrons that currently produce 211At will be summarized and the characteristics of other accelerators that could be utilized for this purpose will be described. Finally, the logistics of networks, both academic and commercial, for facilitating 211At distribution to locations remote from production sites will be addressed.
Collapse
Affiliation(s)
- Yutian Feng
- Department of Radiology, Duke University Medical Center, Durham, NC, USA
| | - Michael R Zalutsky
- Department of Radiology, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
29
|
Zhao L, Liu C, Xing Y, He J, O'Doherty J, Huang W, Zhao J. Development of a 99mTc-Labeled Single-Domain Antibody for SPECT/CT Assessment of HER2 Expression in Breast Cancer. Mol Pharm 2021; 18:3616-3622. [PMID: 34328338 DOI: 10.1021/acs.molpharmaceut.1c00569] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Accurate determination of human epidermal growth factor receptor 2 (HER2) expression is essential for HER2-targeted therapy in patients with cancer. HER2 expression in a complex environment, such as in a heterogeneous tumor, makes the precise assessment of the HER2 status difficult using current methods. In this study, we developed a novel 99mTc-labeled anti-HER2 single-domain antibody (99mTc-NM-02) as a molecular imaging tracer for the noninvasive detection of HER2 expression and investigated its safety, radiation dosimetry, biodistribution, and tumor-targeting potential in 10 patients with breast cancer. Our data showed that no drug-related adverse reactions occurred. The tracer mainly accumulated in the kidneys and liver with mild uptake in the spleen, intestines, and thyroid; however, only background tracer levels were observed in other organs where primary tumors and metastases typically occurred. The mean effective dose was 6.56 × 10-3 mSv/MBq, and tracer uptake was visually observed in the primary tumors and metastases. A maximal standard uptake value of 1.5 was determined as a reasonable cutoff for identifying HER2 positivity using SPECT/CT imaging. Our 99mTc-NM-02 tracer is safe for use in breast cancer imaging, with reasonable radiation doses, favorable biodistribution, and imaging characteristics. 99mTc-NM-02 SPECT imaging may be an accurate and noninvasive method to detect the HER2 status in patients with breast cancer.
Collapse
Affiliation(s)
- Lingzhou Zhao
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100 Haining Road, Shanghai 200080, China
| | - Changcun Liu
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100 Haining Road, Shanghai 200080, China
| | - Yan Xing
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100 Haining Road, Shanghai 200080, China
| | - Jin He
- Department of Pathology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Jim O'Doherty
- Siemens Healthineers, Malvern, Pennsylvania 19355, United States
| | - Wenhua Huang
- Nanomab Technology Limited, Shanghai 200080, China
| | - Jinhua Zhao
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100 Haining Road, Shanghai 200080, China
| |
Collapse
|
30
|
Farzipour S, Shaghaghi Z, Abbasi S, Albooyeh H, Alvandi M. Recent Achievements about Targeted Alpha Therapy-Based Targeting Vectors and Chelating Agents. Anticancer Agents Med Chem 2021; 22:1496-1510. [PMID: 34315393 DOI: 10.2174/1871520621666210727120308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/22/2021] [Accepted: 06/28/2021] [Indexed: 11/22/2022]
Abstract
One of the most rapidly growing options in the management of cancer therapy is Targeted Alpha Therapy (TAT) through which lethal α-emitting radionuclides conjugated to tumor-targeting vectors selectively deliver high amount of radiation to cancer cells.225Ac, 212Bi, 211At, 213Bi, and 223Ra have been investigated by plenty of clinical trials and preclinical researches for the treatment of smaller tumor burdens, micro-metastatic disease, and post-surgery residual disease. In order to send maximum radiation to tumor cells while minimizing toxicity in normal cells, a high affinity of targeting vectors to cancer tissue is essential. Besides that, the stable and specific complex between chelating agent and α-emitters was found as a crucial parameter. The present review was planned to highlight recent achievements about TAT-based targeting vectors and chelating agents and provide further insight for future researches.
Collapse
Affiliation(s)
- Soghra Farzipour
- Cardiovascular Diseases Research Center, Department of Cardiology, Heshmat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Zahra Shaghaghi
- Department of Nuclear Medicine and Molecular Imaging, Clinical Development Research Unit of Farshchian Heart Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Sahar Abbasi
- Department of Radiology, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hajar Albooyeh
- Department of Nuclear Medicine, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Alvandi
- Department of Nuclear Medicine and Molecular Imaging, Clinical Development Research Unit of Farshchian Heart Center, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
31
|
Eychenne R, Chérel M, Haddad F, Guérard F, Gestin JF. Overview of the Most Promising Radionuclides for Targeted Alpha Therapy: The "Hopeful Eight". Pharmaceutics 2021; 13:pharmaceutics13060906. [PMID: 34207408 PMCID: PMC8234975 DOI: 10.3390/pharmaceutics13060906] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/02/2021] [Accepted: 06/08/2021] [Indexed: 12/11/2022] Open
Abstract
Among all existing radionuclides, only a few are of interest for therapeutic applications and more specifically for targeted alpha therapy (TAT). From this selection, actinium-225, astatine-211, bismuth-212, bismuth-213, lead-212, radium-223, terbium-149 and thorium-227 are considered as the most suitable. Despite common general features, they all have their own physical characteristics that make them singular and so promising for TAT. These radionuclides were largely studied over the last two decades, leading to a better knowledge of their production process and chemical behavior, allowing for an increasing number of biological evaluations. The aim of this review is to summarize the main properties of these eight chosen radionuclides. An overview from their availability to the resulting clinical studies, by way of chemical design and preclinical studies is discussed.
Collapse
Affiliation(s)
- Romain Eychenne
- Groupement d’Intérêt Public ARRONAX, 1 Rue Aronnax, F-44817 Saint-Herblain, France;
- Université de Nantes, Inserm, CNRS, Centre de Recherche en Cancérologie et Immunologie Nantes—Angers (CRCINA)—UMR 1232, ERL 6001, F-44000 Nantes, France; (M.C.); (F.G.)
- Correspondence: (R.E.); (J.-F.G.)
| | - Michel Chérel
- Université de Nantes, Inserm, CNRS, Centre de Recherche en Cancérologie et Immunologie Nantes—Angers (CRCINA)—UMR 1232, ERL 6001, F-44000 Nantes, France; (M.C.); (F.G.)
| | - Férid Haddad
- Groupement d’Intérêt Public ARRONAX, 1 Rue Aronnax, F-44817 Saint-Herblain, France;
- Laboratoire Subatech, UMR 6457, Université de Nantes, IMT Atlantique, CNRS, Subatech, F-44000 Nantes, France
| | - François Guérard
- Université de Nantes, Inserm, CNRS, Centre de Recherche en Cancérologie et Immunologie Nantes—Angers (CRCINA)—UMR 1232, ERL 6001, F-44000 Nantes, France; (M.C.); (F.G.)
| | - Jean-François Gestin
- Université de Nantes, Inserm, CNRS, Centre de Recherche en Cancérologie et Immunologie Nantes—Angers (CRCINA)—UMR 1232, ERL 6001, F-44000 Nantes, France; (M.C.); (F.G.)
- Correspondence: (R.E.); (J.-F.G.)
| |
Collapse
|
32
|
Manabe S, Takashima H, Ohnuki K, Koga Y, Tsumura R, Iwata N, Wang Y, Yokokita T, Komori Y, Usuda S, Mori D, Haba H, Fujii H, Yasunaga M, Matsumura Y. Stabilization of an 211At-Labeled Antibody with Sodium Ascorbate. ACS OMEGA 2021; 6:14887-14895. [PMID: 34151070 PMCID: PMC8209801 DOI: 10.1021/acsomega.1c00684] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 05/20/2021] [Indexed: 06/13/2023]
Abstract
211At, an α-particle emitter, has recently attracted attention for radioimmunotherapy of intractable cancers. However, our sodium dodecyl sulfate polyacrylamide gel electrophoresis and flow cytometry analyses revealed that 211At-labeled immunoconjugates are easily disrupted. Luminol assay revealed that reactive oxygen species generated from radiolysis of water caused the disruption of 211At-labeled immunoconjugates. To retain their functions, we explored methods to protect 211At-immunoconjugates from oxidation and enhance their stability. Among several other reducing agents, sodium ascorbate most safely and successfully protected 211At-labeled trastuzumab from oxidative stress and retained the stability of the 211At-labeled antibody and its cytotoxicity against antigen-expressing cells for several days.
Collapse
Affiliation(s)
- Shino Manabe
- Pharmaceutical
Department, Hoshi University 2-4-41, Ebara, Shinagawa, Tokyo 142-8501, Japan
- Research
Center for Pharmaceutical Development Graduate School of Pharmaceutical
Sciences & Faculty of Pharmaceutical Sciences, Tohoku University, 6-3
Aoba, Aramaki, Aoba-ku, Sendai 980-8578, Japan
- Glycometabolic
Biochemistry Laboratory, RIKEN, Hirosawa, Wako, Saitama 351-0198, Japan
| | - Hiroki Takashima
- Division
of Developmental Therapeutics, Exploratory Oncology Research and Clinical
Trial Center, National Cancer Center, 6-5-1 Kahiwanoha, Kashiwa City 277-8577, Japan
| | - Kazunobu Ohnuki
- Division
of Functional Imaging, Exploratory Oncology Research and Clinical
Trial Center, National Cancer Center, 6-5-1 Kahiwanoha, Kashiwa City 277-8577, Japan
| | - Yoshikatsu Koga
- Division
of Developmental Therapeutics, Exploratory Oncology Research and Clinical
Trial Center, National Cancer Center, 6-5-1 Kahiwanoha, Kashiwa City 277-8577, Japan
- Department
of Strategic Programs, Exploratory Oncology Research and Clinical
Trial Center, National Cancer Center, 6-5-1 Kahiwanoha, Kashiwa City 277-8577, Japan
| | - Ryo Tsumura
- Division
of Developmental Therapeutics, Exploratory Oncology Research and Clinical
Trial Center, National Cancer Center, 6-5-1 Kahiwanoha, Kashiwa City 277-8577, Japan
| | - Nozomi Iwata
- Division
of Developmental Therapeutics, Exploratory Oncology Research and Clinical
Trial Center, National Cancer Center, 6-5-1 Kahiwanoha, Kashiwa City 277-8577, Japan
| | - Yang Wang
- Nishina
Center for Accelerator-Based Science, RIKEN, Hirosawa, Wako-shi, Saitama 351-0198 Japan
| | - Takuya Yokokita
- Nishina
Center for Accelerator-Based Science, RIKEN, Hirosawa, Wako-shi, Saitama 351-0198 Japan
| | - Yukiko Komori
- Nishina
Center for Accelerator-Based Science, RIKEN, Hirosawa, Wako-shi, Saitama 351-0198 Japan
| | - Sachiko Usuda
- Nishina
Center for Accelerator-Based Science, RIKEN, Hirosawa, Wako-shi, Saitama 351-0198 Japan
| | - Daiki Mori
- Nishina
Center for Accelerator-Based Science, RIKEN, Hirosawa, Wako-shi, Saitama 351-0198 Japan
| | - Hiromitsu Haba
- Nishina
Center for Accelerator-Based Science, RIKEN, Hirosawa, Wako-shi, Saitama 351-0198 Japan
| | - Hirofumi Fujii
- Division
of Functional Imaging, Exploratory Oncology Research and Clinical
Trial Center, National Cancer Center, 6-5-1 Kahiwanoha, Kashiwa City 277-8577, Japan
| | - Masahiro Yasunaga
- Division
of Developmental Therapeutics, Exploratory Oncology Research and Clinical
Trial Center, National Cancer Center, 6-5-1 Kahiwanoha, Kashiwa City 277-8577, Japan
| | - Yasuhiro Matsumura
- Department
of Immune Medicine, National Cancer Center
Research Institute, 5-1-1 Tsukiji, Chuo-ku, 104-0045 Tokyo, Japan
| |
Collapse
|
33
|
Altunay B, Morgenroth A, Beheshti M, Vogg A, Wong NCL, Ting HH, Biersack HJ, Stickeler E, Mottaghy FM. HER2-directed antibodies, affibodies and nanobodies as drug-delivery vehicles in breast cancer with a specific focus on radioimmunotherapy and radioimmunoimaging. Eur J Nucl Med Mol Imaging 2021; 48:1371-1389. [PMID: 33179151 PMCID: PMC8113197 DOI: 10.1007/s00259-020-05094-1] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 10/26/2020] [Indexed: 02/07/2023]
Abstract
PURPOSE The aim of the present paper is to review the role of HER2 antibodies, affibodies and nanobodies as vehicles for imaging and therapy approaches in breast cancer, including a detailed look at recent clinical data from antibody drug conjugates and nanobodies as well as affibodies that are currently under development. RESULTS Clinical and preclinical studies have shown that the use of monoclonal antibodies in molecular imaging is impaired by slow blood clearance, associated with slow and low tumor uptake and with limited tumor penetration potential. Antibody fragments, such as nanobodies, on the other hand, can be radiolabelled with short-lived radioisotopes and provide high-contrast images within a few hours after injection, allowing early diagnosis and reduced radiation exposure of patients. Even in therapy, the small radioactively labeled nanobodies prove to be superior to radioactively labeled monoclonal antibodies due to their higher specificity and their ability to penetrate the tumor. CONCLUSION While monoclonal antibodies are well established drug delivery vehicles, the current literature on molecular imaging supports the notion that antibody fragments, such as affibodies or nanobodies, might be superior in this approach.
Collapse
Affiliation(s)
- Betül Altunay
- Department of Nuclear Medicine, University Hospital Aachen, RWTH Aachen University, 52074, Aachen, Germany
| | - Agnieszka Morgenroth
- Department of Nuclear Medicine, University Hospital Aachen, RWTH Aachen University, 52074, Aachen, Germany
| | - Mohsen Beheshti
- Department of Nuclear Medicine, University Hospital Aachen, RWTH Aachen University, 52074, Aachen, Germany
- Center of Integrated Oncology (CIO), Universities of Aachen, Bonn, Cologne and Düsseldorf, Kerpener Str. 62, 50937, Cologne, Germany
- Division of Molecular PET-Imaging and Theranostics , Paracelsus Medical University , Salzburg, 5020, Austria
| | - Andreas Vogg
- Department of Nuclear Medicine, University Hospital Aachen, RWTH Aachen University, 52074, Aachen, Germany
| | | | - Hong Hoi Ting
- Nanomab Technology Limited, Shanghai, People's Republic of China
| | | | - Elmar Stickeler
- Center of Integrated Oncology (CIO), Universities of Aachen, Bonn, Cologne and Düsseldorf, Kerpener Str. 62, 50937, Cologne, Germany
- Department of Gynecology and Obstetrics, RWTH Aachen, Aachen, Germany
| | - Felix M Mottaghy
- Department of Nuclear Medicine, University Hospital Aachen, RWTH Aachen University, 52074, Aachen, Germany.
- Center of Integrated Oncology (CIO), Universities of Aachen, Bonn, Cologne and Düsseldorf, Kerpener Str. 62, 50937, Cologne, Germany.
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center (MUMC+), 6202, Maastricht, The Netherlands.
| |
Collapse
|
34
|
Baudhuin H, Puttemans J, Hanssens H, Vanwolleghem P, Hernot S, Raes G, Xavier C, Lahoutte T, Debie P. Decorating sdAbs with Chelators: Effect of Conjugation on Biodistribution and Functionality. Pharmaceuticals (Basel) 2021; 14:ph14050407. [PMID: 33923088 PMCID: PMC8146233 DOI: 10.3390/ph14050407] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 04/21/2021] [Accepted: 04/22/2021] [Indexed: 11/16/2022] Open
Abstract
Single domain antibodies (sdAbs) have proven to be valuable probes for molecular imaging. In order to produce such probes, one strategy is the functionalization of the reactive amine side chain of lysines with a chelator, resulting in a mixture of compounds with a different degree of conjugation. In this study, we implemented anion exchange chromatography (AEX) to separate the different compounds or fractions that were further characterized and evaluated to study the impact of the conjugation degree on pharmacokinetic properties and functionality. Anti-HER2 and anti-MMR sdAbs were functionalized with NOTA or DTPA chelator. Anion exchange chromatography was performed using 0.02 mol/L Tris pH 7.5 as the first solvent and 0.25 M or 0.4 M NaCl (in case of NOTA chelator or DTPA chelator, respectively) as the second solvent applied as a gradient. The fractions were characterized via mass spectrometry (MS), surface plasmon resonance (SPR), and isoelectric focusing gel electrophoresis (IEF), while in vivo studies were performed after radiolabeling with either 68Ga (NOTA) or 111In (DTPA) to assess the impact of the conjugation degree on pharmacokinetics. AEX could successfully be applied to separate fractions of (chelator)n-anti-HER2 and (chelator)n-anti-MMR sdAb constructs. MS confirmed the identity of different peaks obtained in the separation process. SPR measurement suggests a small loss of affinity for (chelator)3-anti-sdAb, while IEF revealed a correlated decrease in isoelectric point (pI) with the number of conjugated chelators. Interestingly, both the reduction in affinity and in pI was stronger with the DTPA chelator than with NOTA for both sdAbs. In vivo data showed no significant differences in organ uptake for any construct, except for (DTPA)n-anti-MMR, which showed a significantly higher liver uptake for (DTPA)1-anti-MMR compared to (DTPA)2-anti-MMR and (DTPA)3-anti-MMR. For all constructs in general, high kidney uptake was observed, due to the typical renal clearance of sdAb-based tracers. The kidney uptake showed significant differences between fractions of a same construct and indicates that a higher conjugation degree improves kidney clearance. AEX allows the separation of sdAbs with a different degree of conjugation and provides the opportunity to further characterize individual fractions. The conjugation of a chelator to sdAbs can alter some properties of the tracers, such as pI; however, the impact on the general biodistribution profile and tumor targeting was minimal.
Collapse
Affiliation(s)
- Henri Baudhuin
- Department of Medical Imaging (MIMA), Vrije Universiteit Brussel, Laarbeeklaan 103, B-1090 Brussels, Belgium; (J.P.); (H.H.); (P.V.); (S.H.); (C.X.); (T.L.); (P.D.)
- Correspondence: ; Tel.: +32-2-477-49-91
| | - Janik Puttemans
- Department of Medical Imaging (MIMA), Vrije Universiteit Brussel, Laarbeeklaan 103, B-1090 Brussels, Belgium; (J.P.); (H.H.); (P.V.); (S.H.); (C.X.); (T.L.); (P.D.)
| | - Heleen Hanssens
- Department of Medical Imaging (MIMA), Vrije Universiteit Brussel, Laarbeeklaan 103, B-1090 Brussels, Belgium; (J.P.); (H.H.); (P.V.); (S.H.); (C.X.); (T.L.); (P.D.)
| | - Philippe Vanwolleghem
- Department of Medical Imaging (MIMA), Vrije Universiteit Brussel, Laarbeeklaan 103, B-1090 Brussels, Belgium; (J.P.); (H.H.); (P.V.); (S.H.); (C.X.); (T.L.); (P.D.)
| | - Sophie Hernot
- Department of Medical Imaging (MIMA), Vrije Universiteit Brussel, Laarbeeklaan 103, B-1090 Brussels, Belgium; (J.P.); (H.H.); (P.V.); (S.H.); (C.X.); (T.L.); (P.D.)
| | - Geert Raes
- Unit of Cellular and Molecular Immunology (CMIM), Vrije Universiteit Brussel, Pleinlaan 2, B-1050 Brussels, Belgium;
- Myeloid Cell Immunology Laboratory, VIB Center for Inflammation Research, Pleinlaan 2, B-1050 Brussels, Belgium
| | - Catarina Xavier
- Department of Medical Imaging (MIMA), Vrije Universiteit Brussel, Laarbeeklaan 103, B-1090 Brussels, Belgium; (J.P.); (H.H.); (P.V.); (S.H.); (C.X.); (T.L.); (P.D.)
| | - Tony Lahoutte
- Department of Medical Imaging (MIMA), Vrije Universiteit Brussel, Laarbeeklaan 103, B-1090 Brussels, Belgium; (J.P.); (H.H.); (P.V.); (S.H.); (C.X.); (T.L.); (P.D.)
- Nuclear Medicine Department (NUCG), Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, B-1090 Brussels, Belgium
| | - Pieterjan Debie
- Department of Medical Imaging (MIMA), Vrije Universiteit Brussel, Laarbeeklaan 103, B-1090 Brussels, Belgium; (J.P.); (H.H.); (P.V.); (S.H.); (C.X.); (T.L.); (P.D.)
| |
Collapse
|
35
|
Piramoon M, Khodadust F, Hosseinimehr SJ. Radiolabeled nanobodies for tumor targeting: From bioengineering to imaging and therapy. Biochim Biophys Acta Rev Cancer 2021; 1875:188529. [PMID: 33647388 DOI: 10.1016/j.bbcan.2021.188529] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 02/13/2021] [Accepted: 02/18/2021] [Indexed: 02/08/2023]
Abstract
So far, numerous molecules and biomolecules have been evaluated for tumor targeting purposes for radionuclide-based imaging and therapy modalities. Due to the high affinity and specificity against tumor antigens, monoclonal antibodies are appropriate candidates for tumor targeting. However, their large size prevents their comprehensive application in radionuclide-based tumor imaging or therapy, since it leads to their low tumor penetration, low blood clearance, and thus inappropriate tumor-to-background ratio. Nowadays, the variable domain of heavy-chain antibodies from the Camelidae family, known as nanobodies (Nbs), turn into exciting candidates for medical research. Considering several innate advantages of these new tumor-targeting agents, including excellent affinity and specificity toward antigen, high solubility, high stability, fast washout from blood, convenient production, ease of selection, and low immunogenicity, it assumes that they may overcome generic problems of monoclonal antibodies, their fragments, and other vectors used for tumor imaging/therapy. After three decades of Nbs discovery, the increasing number of their preclinical and clinical investigations, which have led to outstanding results, confirm their application for tumor targeting purposes. This review describes Nbs characteristics, the diagnostic and therapeutic application of their radioconjugates, and their recent advances.
Collapse
Affiliation(s)
- Majid Piramoon
- Department of Medicinal Chemistry and Radiopharmacy, School of Pharmacy, Lorestan University of Medical Sciences, Khorramabad, Iran; Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Fatemeh Khodadust
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran; Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyed Jalal Hosseinimehr
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
36
|
Aldridge MD, Peet C, Wan S, Shankar A, Gains JE, Bomanji JB, Gaze MN. Paediatric Molecular Radiotherapy: Challenges and Opportunities. Clin Oncol (R Coll Radiol) 2021; 33:80-91. [PMID: 33246658 DOI: 10.1016/j.clon.2020.11.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/17/2020] [Accepted: 11/12/2020] [Indexed: 12/18/2022]
Abstract
The common contemporary indications for paediatric molecular radiotherapy (pMRT) are differentiated thyroid cancer and neuroblastoma. It may also have value in neuroendocrine cancers, and it is being investigated in clinical trials for other diseases. pMRT is the prototypical biomarker-driven, precision therapy, with a unique mode of delivery and mechanism of action. It is safe and well tolerated, compared with other treatments. However, its full potential has not yet been achieved, and its wider use faces a number of challenges and obstacles. Paradoxically, the success of radioactive iodine as a curative treatment for metastatic thyroid cancer has led to a 'one size fits all' approach and limited academic enquiry into optimisation of the conventional treatment regimen, until very recently. Second, the specialised requirements for the delivery of pMRT are available in only a very limited number of centres. This limited capacity and geographical coverage results in reduced accessibility. With few enthusiastic advocates for this treatment modality, investment in research to improve treatments and broaden indications from both industry and national and charitable research funders has historically been suboptimal. Nonetheless, there is now an increasing interest in the opportunities offered by pMRT. Increased research funding has been allocated, and technical developments that will permit innovative approaches in pMRT are available for exploration. A new portfolio of clinical trials is being assembled. These studies should help to move at least some paediatric treatments from simply palliative use into potentially curative protocols. Therapeutic strategies require modification and optimisation to achieve this. The delivery should be personalised and tailored appropriately, with a comprehensive evaluation of tumour and organ-at-risk dosimetry, in alignment with the external beam model of radiotherapy. This article gives an overview of the current status of pMRT, indicating the barriers to progress and identifying ways in which these may be overcome.
Collapse
Affiliation(s)
- M D Aldridge
- Department of Oncology, University College London Hospitals NHS Foundation Trust, London, UK; Department of Nuclear Medicine, University College London Hospitals NHS Foundation Trust, London, UK
| | - C Peet
- Department of Oncology, University College London Hospitals NHS Foundation Trust, London, UK
| | - S Wan
- Department of Nuclear Medicine, University College London Hospitals NHS Foundation Trust, London, UK
| | - A Shankar
- Department of Paediatric and Adolescent Oncology, University College London Hospitals NHS Foundation Trust, London, UK
| | - J E Gains
- Department of Oncology, University College London Hospitals NHS Foundation Trust, London, UK
| | - J B Bomanji
- Department of Nuclear Medicine, University College London Hospitals NHS Foundation Trust, London, UK
| | - M N Gaze
- Department of Nuclear Medicine, University College London Hospitals NHS Foundation Trust, London, UK.
| |
Collapse
|
37
|
Feng Y, Zhou Z, McDougald D, Meshaw RL, Vaidyanathan G, Zalutsky MR. Site-specific radioiodination of an anti-HER2 single domain antibody fragment with a residualizing prosthetic agent. Nucl Med Biol 2021; 92:171-183. [PMID: 32448731 PMCID: PMC7657985 DOI: 10.1016/j.nucmedbio.2020.05.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/06/2020] [Accepted: 05/07/2020] [Indexed: 11/28/2022]
Abstract
INTRODUCTION As a consequence of their small size, high stability and high affinity, single domain antibody fragments (sdAbs) are appealing targeting vectors for radiopharmaceutical development. With sdAbs binding to internalizing receptors like HER2, residualizing prosthetic agents can enhance tumor retention of radioiodine, which until now has been done with random labeling approaches. Herein we evaluate a site-specific strategy utilizing a radioiodinated, residualizing maleimido moiety and the anti-HER2 sdAb 5F7 bearing a GGC tail for conjugation. METHODS Maleimidoethyl 3-(guanidinomethyl)-5-iodobenzoate ([131I]MEGMB) and its N-succinimidyl ester analogue, iso-[125I]SGMIB, were labeled by halodestannylation and conjugated with 5F7GGC and 5F7, respectively. Radiochemical purity, immunoreactivity and binding affinity were determined. Paired-label experiments directly compared iso-[125I]SGMIB-5F7 and [131I]MEGMIB-5F7GGC with regard to internalization/residualization and affinity on HER2-expressing SKOV-3 ovarian carcinoma cells as well as biodistribution and metabolite distribution in athymic mice with subcutaneous SKOV-3 xenografts. RESULTS [131I]MEGMIB-5F7GGC had an immunoreactivity of 81.3% and Kd = 0.94 ± 0.27 nM. Internalization assays demonstrated high intracellular trapping for both conjugates, For example, at 1 h, intracellular retention was 50.30 ± 3.36% for [131I]MEGMIB-5F7GGC and 55.95 ± 3.27% for iso-[125I]SGMIB-5F7, while higher retention was seen for iso-[125I]SGMIB-5F7 at later time points. Peak tumor uptake was similar for both conjugates (8.35 ± 2.66%ID/g and 8.43 ± 2.84%ID/g for iso-[125I]SGMIB-5F7 and [131I]MEGMIB-5F7GGC at 1 h, respectively); however, more rapid normal tissue clearance was seen for [131I]MEGMIB-5F7GGC, with a 2-fold higher tumor-to-kidney ratio and a 3-fold higher tumor-to-liver ratio compared with co-injected iso-[125I]SGMIB-5F7. Consisted with this, generation of labeled catabolites in the kidneys was higher for [131I]MEGMIB-5F7GGC. CONCLUSION [131I]MEGMIB-5F7GGC offers similar tumor targeting as iso-[125I]SGMIB-5F7 but with generally lower normal tissue uptake. ADVANCES IN KNOWLEDGE AND IMPLICATION FOR PATIENT CARE The site specific nature of the [131I]MEGMIB reagent may facilitate clinical translation, particularly for sdAb with compromised affinity after random labeling.
Collapse
Affiliation(s)
- Yutian Feng
- Department of Radiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Zhengyuan Zhou
- Department of Radiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Darryl McDougald
- Department of Radiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Rebecca L Meshaw
- Department of Radiology, Duke University Medical Center, Durham, NC 27710, USA
| | | | - Michael R Zalutsky
- Department of Radiology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
38
|
Targeted Alpha Therapy: Progress in Radionuclide Production, Radiochemistry, and Applications. Pharmaceutics 2020; 13:pharmaceutics13010049. [PMID: 33396374 PMCID: PMC7824049 DOI: 10.3390/pharmaceutics13010049] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 12/15/2020] [Accepted: 12/23/2020] [Indexed: 12/17/2022] Open
Abstract
This review outlines the accomplishments and potential developments of targeted alpha (α) particle therapy (TAT). It discusses the therapeutic advantages of the short and highly ionizing path of α-particle emissions; the ability of TAT to complement and provide superior efficacy over existing forms of radiotherapy; the physical decay properties and radiochemistry of common α-emitters, including 225Ac, 213Bi, 224Ra, 212Pb, 227Th, 223Ra, 211At, and 149Tb; the production techniques and proper handling of α-emitters in a radiopharmacy; recent preclinical developments; ongoing and completed clinical trials; and an outlook on the future of TAT.
Collapse
|
39
|
Bathula NV, Bommadevara H, Hayes JM. Nanobodies: The Future of Antibody-Based Immune Therapeutics. Cancer Biother Radiopharm 2020; 36:109-122. [PMID: 32936001 DOI: 10.1089/cbr.2020.3941] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Targeted therapy is a fast evolving treatment strategy to reduce unwanted damage to healthy tissues, while increasing efficacy and specificity. Driven by state-of-the-art technology, this therapeutic approach is especially true of cancer. Antibodies with their remarkable specificity have revolutionized therapeutic strategies for autoimmune conditions and cancer, particularly blood-borne cancers, but have severe limitations in treating solid tumors. This is mainly due to their large molecular size, low stability, tumor-tissue penetration difficulties, and pharmacokinetic properties. The tumor microenvironment, rich in immune-suppressing molecules is also a major barrier in targeting solid tumors by antibody-based drugs. Nanobodies have recently emerged as an alternative therapeutic agent to overcome some of the drawbacks of traditional antibody treatment. Nanobodies are the VHH domains found on the heavy-chain only antibodies of camelids and are the smallest naturally available antibody fragments with excellent antigen-binding specificity and affinity, equivalent to conventional antibodies but with molecular weights as low as 15 kDa. The compact size, high stability, enhanced hydrophilicity, particularly in framework regions, excellent epitope interactions with protruding CDR3 regions, and improved tissue penetration make nanobodies the next-generation therapeutics (Nano-BioDrugs). In this review, the authors discuss the interesting properties of nanobodies and their advantages over their conventional counterparts and provide insight into how nanobodies are being utilized as agonists and antagonists, bispecific constructs, and drug and enzyme-conjugates to combat the tumor microenvironment and treat disease.
Collapse
Affiliation(s)
- Nuthan V Bathula
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Hemashree Bommadevara
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Jerrard M Hayes
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
40
|
Lindegren S, Albertsson P, Bäck T, Jensen H, Palm S, Aneheim E. Realizing Clinical Trials with Astatine-211: The Chemistry Infrastructure. Cancer Biother Radiopharm 2020; 35:425-436. [PMID: 32077749 PMCID: PMC7465635 DOI: 10.1089/cbr.2019.3055] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Despite the consensus around the clinical potential of the α-emitting radionuclide astatine-211 (211At), there are only a limited number of research facilities that work with this nuclide. There are three main reasons for this: (1) Scarce availability of the nuclide. Despite a relatively large number of globally existing cyclotrons capable of producing 211At, few cyclotron facilities produce the nuclide on a regular basis. (2) Lack of a chemical infrastructure, that is, isolation of 211At from irradiated targets and the subsequent synthesis of an astatinated product. At present, the research groups that work with 211At depend on custom systems for recovering 211At from the irradiated targets. Setting up and implementing such custom units require long lead times to provide a proper working system. (3) The chemistry of 211At. Compared with radiometals there are no well-established and generally accepted synthesis methods for forming sufficiently stable bonds between 211At and the tumor-specific vector to allow for systemic applications. Herein we present an overview of the infrastructure of producing 211At radiopharmaceuticals, from target to radiolabeled product including chemical strategies to overcome hurdles for advancement into clinical trials with 211At.
Collapse
Affiliation(s)
- Sture Lindegren
- Department of Radiation Physics and Targeted Alpha Therapy Group, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Per Albertsson
- Department of Oncology, Targeted Alpha Therapy Group, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Oncology, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Tom Bäck
- Department of Radiation Physics and Targeted Alpha Therapy Group, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Holger Jensen
- Cyclotron and PET unit KF-3982, Copenhagen University Hospital, Copenhagen, Denmark
| | - Stig Palm
- Department of Radiation Physics and Targeted Alpha Therapy Group, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Emma Aneheim
- Department of Radiation Physics and Targeted Alpha Therapy Group, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Oncology, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
41
|
Dekempeneer Y, Caveliers V, Ooms M, Maertens D, Gysemans M, Lahoutte T, Xavier C, Lecocq Q, Maes K, Covens P, Miller BW, Bruchertseifer F, Morgenstern A, Cardinaels T, D’Huyvetter M. Therapeutic Efficacy of 213Bi-labeled sdAbs in a Preclinical Model of Ovarian Cancer. Mol Pharm 2020; 17:3553-3566. [DOI: 10.1021/acs.molpharmaceut.0c00580] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Yana Dekempeneer
- Institute for Nuclear Materials Science, Belgian Nuclear Research Center (SCK CEN), 2400 Mol, Belgium
- In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium
| | - Vicky Caveliers
- In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium
- Department of Nuclear Medicine, UZ Brussel, 1090 Brussels, Belgium
| | - Maarten Ooms
- Institute for Nuclear Materials Science, Belgian Nuclear Research Center (SCK CEN), 2400 Mol, Belgium
| | - Dominic Maertens
- Institute for Nuclear Materials Science, Belgian Nuclear Research Center (SCK CEN), 2400 Mol, Belgium
| | - Mireille Gysemans
- Institute for Nuclear Materials Science, Belgian Nuclear Research Center (SCK CEN), 2400 Mol, Belgium
| | - Tony Lahoutte
- In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium
- Department of Nuclear Medicine, UZ Brussel, 1090 Brussels, Belgium
| | - Catarina Xavier
- In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium
| | - Quentin Lecocq
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel (VUB), 1050 Brussels, Belgium
| | - Ken Maes
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel (VUB), 1050 Brussels, Belgium
| | - Peter Covens
- In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium
| | - Brian W. Miller
- Department of Radiation Oncology, University of Colorado School of Medicine, Aurora, Colorado 80045, United States
| | - Frank Bruchertseifer
- Directorate for Nuclear Safety and Security, European Commission−Joint Research Centre, Karlsruhe 76344, Germany
| | - Alfred Morgenstern
- Directorate for Nuclear Safety and Security, European Commission−Joint Research Centre, Karlsruhe 76344, Germany
| | - Thomas Cardinaels
- Institute for Nuclear Materials Science, Belgian Nuclear Research Center (SCK CEN), 2400 Mol, Belgium
- Department of Chemistry, KU Leuven, Heverlee, 3000 Leuven, Belgium
| | - Matthias D’Huyvetter
- In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium
| |
Collapse
|
42
|
Guerra Liberal FDC, O'Sullivan JM, McMahon SJ, Prise KM. Targeted Alpha Therapy: Current Clinical Applications. Cancer Biother Radiopharm 2020; 35:404-417. [PMID: 32552031 DOI: 10.1089/cbr.2020.3576] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
α-Emitting radionuclides have been approved for cancer treatment since 2013, with increasing degrees of success. Despite this clinical utility, little is known regarding the mechanisms of action of α particles in this setting, and accurate assessments of the dosimetry underpinning their effectiveness are lacking. However, targeted alpha therapy (TAT) is gaining more attention as new targets, synthetic chemistry approaches, and α particle emitters are identified, constructed, developed, and realized. From a radiobiological perspective, α particles are more effective at killing cells compared to low linear energy transfer radiation. Also, from these direct effects, it is now evident from preclinical and clinical data that α emitters are capable of both producing effects in nonirradiated bystander cells and stimulating the immune system, extending the biological effects of TAT beyond the range of α particles. The short range of α particles makes them a potent tool to irradiate single-cell lesions or treat solid tumors by minimizing unwanted irradiation of normal tissue surrounding the cancer cells, assuming a high specificity of the radiopharmaceutical and good stability of its chemical bonds. Clinical approval of 223RaCl2 in 2013 was a major milestone in the widespread application of TAT as a safe and effective strategy for cancer treatment. In addition, 225Ac-prostate specific membrane antigen treatment benefit in metastatic castrate-resistant prostate cancer patients, refractory to standard therapies, is another game-changing piece in the short history of TAT clinical application. Clinical applications of TAT are growing with different radionuclides and combination therapies, and in different clinical settings. Despite the remarkable advances in TAT dosimetry and imaging, it has not yet been used to its full potential. Labeled 227Th and 225Ac appear to be promising candidates and could represent the next generation of agents able to extend patient survival in several clinical scenarios.
Collapse
Affiliation(s)
- Francisco D C Guerra Liberal
- The Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom.,Faculdade de Ciências e Tenclonogia, Universidade Nova de Lisboa, Caparica, Portugal
| | - Joe M O'Sullivan
- The Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom.,Clinical Oncology, Northern Ireland Cancer Centre, Belfast Health and Social Care Trust, Belfast, United Kingdom
| | - Stephen J McMahon
- The Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom
| | - Kevin M Prise
- The Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom
| |
Collapse
|
43
|
The five “W”s and “How” of Targeted Alpha Therapy: Why? Who? What? Where? When? and How? RENDICONTI LINCEI-SCIENZE FISICHE E NATURALI 2020. [DOI: 10.1007/s12210-020-00900-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
44
|
Deken MM, Kijanka MM, Beltrán Hernández I, Slooter MD, de Bruijn HS, van Diest PJ, van Bergen En Henegouwen PMP, Lowik CWGM, Robinson DJ, Vahrmeijer AL, Oliveira S. Nanobody-targeted photodynamic therapy induces significant tumor regression of trastuzumab-resistant HER2-positive breast cancer, after a single treatment session. J Control Release 2020; 323:269-281. [PMID: 32330574 PMCID: PMC7116241 DOI: 10.1016/j.jconrel.2020.04.030] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 04/18/2020] [Accepted: 04/20/2020] [Indexed: 12/18/2022]
Abstract
Rationale A substantial number of breast cancer patients with an overexpression of the human epidermal growth factor receptor 2 (HER2) have residual disease after neoadjuvant therapy or become resistant to trastuzumab. Photodynamic therapy (PDT) using nanobodies targeted to HER2 is a promising treatment option for these patients. Here we investigate the in vitro and in vivo antitumor efficacy of HER2-targeted nanobody-photosensitizer (PS) conjugate PDT. Methods Nanobodies targeting HER2 were obtained from phage display selections. Monovalent nanobodies were engineered into a biparatopic construct. The specificity of selected nanobodies was tested in immunofluorescence assays and their affinity was evaluated in binding studies, both performed in a panel of breast cancer cells varying in HER2 expression levels. The selected HER2-targeted nanobodies 1D5 and 1D5-18A12 were conjugated to the photosensitizer IRDye700DX and tested in in vitro PDT assays. Mice bearing orthotopic HCC1954 trastuzumab-resistant tumors with high HER2 expression or MCF-7 tumors with low HER2 expression were intravenously injected with nanobody-PS conjugates. Quantitative fluorescence spectroscopy was performed for the determination of the local pharmacokinetics of the fluorescence conjugates. After nanobody-PS administration, tumors were illuminated to a fluence of 100 J∙cm-2, with a fluence rate of 50 mW∙cm-2, and thereafter tumor growth was measured with a follow-up until 30 days. Results The selected nanobodies remained functional after conjugation to the PS, binding specifically and with high affinity to HER2-positive cells. Both nanobody-PS conjugates potently and selectively induced cell death of HER2 overexpressing cells, either sensitive or resistant to trastuzumab, with low nanomolar LD50 values. In vivo, quantitative fluorescence spectroscopy showed specific accumulation of nanobody-PS conjugates in HCC1954 tumors and indicated 2 h post injection as the most suitable time point to apply light. Nanobody-targeted PDT with 1D5-PS and 1D5-18A12-PS induced significant tumor regression of trastuzumab-resistant high HER2 expressing tumors, whereas in low HER2 expressing tumors only a slight growth delay was observed. Conclusion Nanobody-PS conjugates accumulated selectively in vivo and their fluorescence could be detected through optical imaging. Upon illumination, they selectively induced significant tumor regression of HER2 overexpressing tumors with a single treatment session. Nanobody-targeted PDT is therefore suggested as a new additional treatment for HER2-positive breast cancer, particularly of interest for trastuzumab-resistant HER2-positive breast cancer. Further studies are now needed to assess the value of this approach in clinical practice.
Collapse
Affiliation(s)
- Marion M Deken
- Dept. of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Marta M Kijanka
- Division of Cell Biology, Neurobiology and Biophysics, Dept. of Biology, Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | - Irati Beltrán Hernández
- Pharmaceutics, Dept. of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | - Maxime D Slooter
- Dept. of Radiology, Division of Molecular Imaging, Leiden University Medical Center, Leiden, the Netherlands
| | - Henriette S de Bruijn
- Dept. of Otorhinolaryngology & Head and Neck Surgery, Center for Optical Diagnostics and Therapy, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Paul J van Diest
- Dept. of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Paul M P van Bergen En Henegouwen
- Division of Cell Biology, Neurobiology and Biophysics, Dept. of Biology, Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | - Clemens W G M Lowik
- Dept. of Radiology, Optical Molecular Imaging, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Dominic J Robinson
- Dept. of Surgery, Leiden University Medical Center, Leiden, the Netherlands; Dept. of Otorhinolaryngology & Head and Neck Surgery, Center for Optical Diagnostics and Therapy, Erasmus Medical Center, Rotterdam, the Netherlands
| | | | - Sabrina Oliveira
- Division of Cell Biology, Neurobiology and Biophysics, Dept. of Biology, Faculty of Science, Utrecht University, Utrecht, the Netherlands; Pharmaceutics, Dept. of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
45
|
Preclinical Targeted α- and β --Radionuclide Therapy in HER2-Positive Brain Metastasis Using Camelid Single-Domain Antibodies. Cancers (Basel) 2020; 12:cancers12041017. [PMID: 32326199 PMCID: PMC7226418 DOI: 10.3390/cancers12041017] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 04/13/2020] [Accepted: 04/16/2020] [Indexed: 12/13/2022] Open
Abstract
HER2-targeted therapies have drastically improved the outcome for breast cancer patients. However, when metastasis to the brain is involved, current strategies fail to hold up to the same promise. Camelid single-domain antibody-fragments (sdAbs) have been demonstrated to possess favorable properties for detecting and treating cancerous lesions in vivo using different radiolabeling methods. Here we evaluate the anti-HER2 sdAb 2Rs15d, coupled to diagnostic γ- and therapeutic α- and β−-emitting radionuclides for the detection and treatment of HER2pos brain lesions in a preclinical setting. 2Rs15d was radiolabeled with 111In, 225Ac and 131I using DTPA- and DOTA-based bifunctional chelators and Sn-precursor of SGMIB respectively and evaluated in orthotopic tumor-bearing athymic nude mice. Therapeutic efficacy as well as systemic toxicity were determined for 131I- and 225Ac-labeled sdAbs and compared to anti-HER2 monoclonal antibody (mAb) trastuzumab in two different HER2pos tumor models. Radiolabeled 2Rs15d showed high and specific tumor uptake in both HER2pos SK-OV-3-Luc-IP1 and HER2pos MDA-MB-231Br brain lesions, whereas radiolabeled trastuzumab was unable to accumulate in intracranial SK-OV-3-Luc-IP1 tumors. Administration of [131I]-2Rs15d and [225Ac]-2Rs15d alone and in combination with trastuzumab showed a significant increase in median survival in 2 tumor models that remained largely unresponsive to trastuzumab treatment alone. Histopathological analysis revealed no significant early toxicity. Radiolabeled sdAbs prove to be promising vehicles for molecular imaging and targeted radionuclide therapy of metastatic lesions in the brain. These data demonstrate the potential of radiolabeled sdAbs as a valuable add-on treatment option for patients with difficult-to-treat HER2pos metastatic cancer.
Collapse
|
46
|
Woen DH, Eiroa-Lledo C, Akin AC, Anderson NH, Bennett KT, Birnbaum ER, Blake AV, Brugh M, Dalodière E, Dorman EF, Ferrier MG, Hamlin DK, Kozimor SA, Li Y, Lilley LM, Mocko V, Thiemann SL, Wilbur DS, White FD. A Solid-State Support for Separating Astatine-211 from Bismuth. Inorg Chem 2020; 59:6137-6146. [PMID: 32302134 DOI: 10.1021/acs.inorgchem.0c00221] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Increasing access to the short-lived α-emitting radionuclide astatine-211 (211At) has the potential to advance targeted α-therapeutic treatment of disease and to solve challenges facing the medical community. For example, there are numerous technical needs associated with advancing the use of 211At in targeted α-therapy, e.g., improving 211At chelates, developing more effective 211At targeting, and characterizing in vivo 211At behavior. There is an insufficient understanding of astatine chemistry to support these efforts. The chemistry of astatine is one of the least developed of all elements on the periodic table, owing to its limited supply and short half-life. Increasing access to 211At could help address these issues and advance understanding of 211At chemistry in general. We contribute here an extraction chromatographic processing method that simplifies 211At production in terms of purification. It utilizes the commercially available Pre-Filter resin to rapidly (<1.5 h) isolate 211At from irradiated bismuth targets (Bi decontamination factors ≥876 000), in reasonable yield (68-55%) and in a form that is compatible for subsequent in vivo study. We are excited about the potential of this procedure to address 211At supply and processing/purification problems.
Collapse
Affiliation(s)
- David H Woen
- Los Alamos National Laboratory, Los Alamos, New Mexico 87545, United States
| | | | - Andrew C Akin
- Los Alamos National Laboratory, Los Alamos, New Mexico 87545, United States
| | | | - Kevin T Bennett
- Los Alamos National Laboratory, Los Alamos, New Mexico 87545, United States
| | - Eva R Birnbaum
- Los Alamos National Laboratory, Los Alamos, New Mexico 87545, United States
| | - Anastasia V Blake
- Los Alamos National Laboratory, Los Alamos, New Mexico 87545, United States
| | - Mark Brugh
- Los Alamos National Laboratory, Los Alamos, New Mexico 87545, United States
| | - Elodie Dalodière
- Los Alamos National Laboratory, Los Alamos, New Mexico 87545, United States
| | - Eric F Dorman
- Department of Radiation Oncology, University of Washington, Seattle, Washington 98195, United States
| | - Maryline G Ferrier
- Department of Radiation Oncology, University of Washington, Seattle, Washington 98195, United States
| | - Donald K Hamlin
- Department of Radiation Oncology, University of Washington, Seattle, Washington 98195, United States
| | - Stosh A Kozimor
- Los Alamos National Laboratory, Los Alamos, New Mexico 87545, United States
| | - Yawen Li
- Department of Radiation Oncology, University of Washington, Seattle, Washington 98195, United States
| | - Laura M Lilley
- Los Alamos National Laboratory, Los Alamos, New Mexico 87545, United States
| | - Veronika Mocko
- Los Alamos National Laboratory, Los Alamos, New Mexico 87545, United States
| | - Sara L Thiemann
- Los Alamos National Laboratory, Los Alamos, New Mexico 87545, United States
| | - D Scott Wilbur
- Department of Radiation Oncology, University of Washington, Seattle, Washington 98195, United States
| | - Frankie D White
- Los Alamos National Laboratory, Los Alamos, New Mexico 87545, United States
| |
Collapse
|